1
|
Hsu CL, Chang YS, Li HP. Molecular diagnosis of nasopharyngeal carcinoma: Past and future. Biomed J 2025; 48:100748. [PMID: 38796105 PMCID: PMC11772973 DOI: 10.1016/j.bj.2024.100748] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2024] [Revised: 05/01/2024] [Accepted: 05/12/2024] [Indexed: 05/28/2024] Open
Abstract
Nasopharyngeal carcinoma (NPC) is a malignant tumor originated from the nasopharynx epithelial cells and has been linked with Epstein-Barr virus (EBV) infection, dietary habits, environmental and genetic factors. It is a common malignancy in Southeast Asia, especially with gender preference among men. Due to its non-specific symptoms, NPC is often diagnosed at a late stage. Thus, the molecular diagnosis of NPC plays a crucial role in early detection, treatment selection, disease monitoring, and prognosis prediction. This review aims to provide a summary of the current state and the latest emerging molecular diagnostic techniques for NPC, including EBV-related biomarkers, gene mutations, liquid biopsy, and DNA methylation. Challenges and potential future directions of NPC molecular diagnosis will be discussed.
Collapse
Affiliation(s)
- Cheng-Lung Hsu
- Division of Hematology-Oncology, Department of Internal Medicine, Chang Gung Memorial Hospital, Chang Gung University, Taoyuan, Taiwan
- School of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Yu-Sun Chang
- Graduate Institute of Biomedical Sciences, Chang Gung University, Taoyuan, Taiwan
- Molecular Medicine Research Center, Chang Gung University, Taoyuan, Taiwan
- Department of Otolaryngology-Head and Neck Surgery, Chang Gung Memorial Hospital, Chang Gung University, Taoyuan, Taiwan
| | - Hsin-Pai Li
- Division of Hematology-Oncology, Department of Internal Medicine, Chang Gung Memorial Hospital, Chang Gung University, Taoyuan, Taiwan
- Graduate Institute of Biomedical Sciences, Chang Gung University, Taoyuan, Taiwan
- Molecular Medicine Research Center, Chang Gung University, Taoyuan, Taiwan
- Department of Microbiology and Immunology, Chang Gung University, Taoyuan, Taiwan
| |
Collapse
|
2
|
Torne AS, Robertson ES. Epigenetic Mechanisms in Latent Epstein-Barr Virus Infection and Associated Cancers. Cancers (Basel) 2024; 16:991. [PMID: 38473352 PMCID: PMC10931536 DOI: 10.3390/cancers16050991] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Revised: 02/25/2024] [Accepted: 02/25/2024] [Indexed: 03/14/2024] Open
Abstract
The Epstein-Barr Virus (EBV) is a double-stranded DNA-based human tumor virus that was first isolated in 1964 from lymphoma biopsies. Since its initial discovery, EBV has been identified as a major contributor to numerous cancers and chronic autoimmune disorders. The virus is particularly efficient at infecting B-cells but can also infect epithelial cells, utilizing an array of epigenetic strategies to establish long-term latent infection. The association with histone modifications, alteration of DNA methylation patterns in host and viral genomes, and microRNA targeting of host cell factors are core epigenetic strategies that drive interactions between host and virus, which are necessary for viral persistence and progression of EBV-associated diseases. Therefore, understanding epigenetic regulation and its role in post-entry viral dynamics is an elusive area of EBV research. Here, we present current outlooks of EBV epigenetic regulation as it pertains to viral interactions with its host during latent infection and its propensity to induce tumorigenesis. We review the important epigenetic regulators of EBV latency and explore how the strategies involved during latent infection drive differential epigenetic profiles and host-virus interactions in EBV-associated cancers.
Collapse
Affiliation(s)
| | - Erle S. Robertson
- Tumor Virology Program, Department of Otorhinolaryngology-Head and Neck Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA;
| |
Collapse
|
3
|
Li HP, Huang CY, Lui KW, Chao YK, Yeh CN, Lee LY, Huang Y, Lin TL, Kuo YC, Huang MY, Fan HC, Lin AC, Hsieh CH, Chang KP, Lin CY, Wang HM, Chao M, Liu JS, Chang YS, Hsu CL. Nasopharyngeal carcinoma patient-derived xenograft mouse models reveal potential drugs targeting cell cycle, mTOR, and autophagy pathways. Transl Oncol 2023; 38:101785. [PMID: 37713975 PMCID: PMC10509698 DOI: 10.1016/j.tranon.2023.101785] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Revised: 09/05/2023] [Accepted: 09/08/2023] [Indexed: 09/17/2023] Open
Abstract
BACKGROUND Nasopharyngeal carcinoma (NPC) is associated with Epstein-Barr virus (EBV) infection. To test preclinical NPC drugs, we established two patient-derived xenograft (PDX) mouse models, EBV-positive PDX-B13 and EBV-negative PDX-Li41, for drug screening. METHODS Based on next generation sequencing (NGS) studies, PDX-B13 had CCND1 copy number (CN) gain but CDKN2A CN loss, whereas PDX-Li41 had CDKN2A and RB1 CN loss, TSC1 (negative regulator of mTOR) frameshift deletion mutation, and increased activation of mTOR, a serine/threonine kinase that governs metabolism, autophagy, and apoptosis. Increased mTOR was also associated with poor NPC prognosis. RESULTS Everolimus, an mTOR inhibitor, suppressed tumor growth in the two PDX NPC models and had an additive antitumor effect with palbociclib, a CDK4/6 inhibitor. PDX tumors treated with various drugs or untreated were subjected to RNA sequencing, transcriptome profile analysis, and selective Western blotting to understand the interactions between these drugs and gene expression profiles. Palbociclib also suppressed EB viral nuclear antigen (EBNA1) expression in PDX-B13. Everolimus together with autophagy inhibitor, hydroxychloroquine, had additive anti-tumor effect on PDX-B13 tumor. Immunohistochemistry revealed that high mTOR levels were correlated with poor overall survival in patients with metastatic NPC (N = 90). CONCLUSIONS High mTOR levels are a poor prognostic factor in NPC, and cell cycle, mTOR and autophagy pathways may serve as therapeutic targets in NPC. In addition, PDX models can be used for efficiently testing potential NPC drugs.
Collapse
Affiliation(s)
- Hsin-Pai Li
- Department of Microbiology and Immunology, Chang Gung University, Taoyuan 33305, Taiwan; Graduate Institute of Biomedical Sciences, Chang Gung University, Taoyuan 33305, Taiwan; Molecular Medicine Research Center, Chang Gung University, Taoyuan 33305, Taiwan; Division of Hematology-Oncology, Department of Internal Medicine, Chang Gung Memorial Hospital, Chang Gung University, Taoyuan 33305, Taiwan
| | - Chen-Yang Huang
- Division of Hematology-Oncology, Department of Internal Medicine, Chang Gung Memorial Hospital, Chang Gung University, Taoyuan 33305, Taiwan
| | - Kar-Wai Lui
- Department of Medical Imaging and Intervention, Chang Gung Memorial Hospital, Chang Gung University, Taoyuan 33305, Taiwan
| | - Yin-Kai Chao
- Division of Thoracic and Cardiovascular Surgery, Department of Surgery, Chang Gung Memorial Hospital, Chang Gung University, Taoyuan 33305, Taiwan
| | - Chun-Nan Yeh
- Department of General Surgery, Liver Research Center, Chang Gung Memorial Hospital, Chang Gung University, Taoyuan 33305, Taiwan
| | - Li-Yu Lee
- Department of Pathology, Chang Gung Memorial Hospital, Chang Gung University, Taoyuan 33305, Taiwan
| | - Yenlin Huang
- Department of Pathology, Chang Gung Memorial Hospital, Chang Gung University, Taoyuan 33305, Taiwan
| | - Tung-Liang Lin
- Division of Hematology-Oncology, Department of Internal Medicine, Chang Gung Memorial Hospital, Chang Gung University, Taoyuan 33305, Taiwan
| | - Yung-Chia Kuo
- Division of Hematology-Oncology, Department of Internal Medicine, Chang Gung Memorial Hospital, Chang Gung University, Taoyuan 33305, Taiwan
| | - Mei-Yuan Huang
- Department of Microbiology and Immunology, Chang Gung University, Taoyuan 33305, Taiwan
| | - Hsien-Chi Fan
- Division of Hematology-Oncology, Department of Internal Medicine, Chang Gung Memorial Hospital, Chang Gung University, Taoyuan 33305, Taiwan
| | - An-Chi Lin
- Division of Hematology-Oncology, Department of Internal Medicine, Chang Gung Memorial Hospital, Chang Gung University, Taoyuan 33305, Taiwan
| | - Chia-Hsun Hsieh
- Division of Hematology-Oncology, Department of Internal Medicine, Chang Gung Memorial Hospital, Chang Gung University, Taoyuan 33305, Taiwan
| | - Kai-Ping Chang
- Department of Otolaryngology-Head and Neck Surgery, Chang Gung Memorial Hospital, Chang Gung University, Taoyuan 33305, Taiwan
| | - Chien-Yu Lin
- Department of Radiation, Chang Gung Memorial Hospital, Chang Gung University, Taoyuan 33305, Taiwan
| | - Hung-Ming Wang
- Division of Hematology-Oncology, Department of Internal Medicine, Chang Gung Memorial Hospital, Chang Gung University, Taoyuan 33305, Taiwan
| | - Mei Chao
- Department of Microbiology and Immunology, Chang Gung University, Taoyuan 33305, Taiwan; Graduate Institute of Biomedical Sciences, Chang Gung University, Taoyuan 33305, Taiwan; Department of Hepato-Gastroenterology, Chang Gung Memorial Hospital, Chang Gung University, Taoyuan 33305, Taiwan
| | - Jai-Shin Liu
- Department of Biotechnology and Pharmaceutical Technology, Yuanpei University of Medical Technology, Hsinchu City 30015, Taiwan
| | - Yu-Sun Chang
- Department of Microbiology and Immunology, Chang Gung University, Taoyuan 33305, Taiwan; Molecular Medicine Research Center, Chang Gung University, Taoyuan 33305, Taiwan; Department of Otolaryngology-Head and Neck Surgery, Chang Gung Memorial Hospital, Chang Gung University, Taoyuan 33305, Taiwan
| | - Cheng-Lung Hsu
- Division of Hematology-Oncology, Department of Internal Medicine, Chang Gung Memorial Hospital, Chang Gung University, Taoyuan 33305, Taiwan; School of Medicine, Chang Gung University, Taoyuan 33305, Taiwan.
| |
Collapse
|
4
|
Sarshari B, Ravanshad M, Rabbani A, Zareh-Khoshchehreh R, Mokhtari F, Khanabadi B, Mohebbi SR, Asadzadeh Aghdaei H. Quantitative analysis of Epstein-Barr virus DNA in plasma and stomach biopsies of patients with gastric cancer. Virus Genes 2023; 59:351-358. [PMID: 36757510 DOI: 10.1007/s11262-023-01977-1] [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: 09/18/2022] [Accepted: 01/29/2023] [Indexed: 02/10/2023]
Abstract
Epstein-Barr virus (EBV) associated gastric carcinoma (EBVaGC) is a subtype of gastric cancer with distinct histological and molecular features. The study aimed to assess the EBV DNA copy number and the prevalence of EBVaGC in gastric cancer samples taken from Iranian patients. The next aim was to assess whether the DNA and microRNAs EBV are present in plasma. EBV load was analyzed in 68 gastric cancer biopsies and compared with the results of EBV-encoded small RNA in situ hybridization (EBER-ISH) test in these patients. After the detection of 6 EBV miRNAs in gastric tissue by stem-loop RT-PCR, plasma samples were evaluated for the viral load and EBV miRNAs. Four gastric cancer cases were EBER -ISH positive (5.8%), with a significantly higher viral load than the remaining cases, 47,781 vs. 1909 copies/μg of tissue DNA. Here, was also found a significant difference in plasma EBV load between EBER-positive and EBER-negative cases. Although EBV miRNAs were detectable in all the EBER-positive tumors, the test did not detect any of these miRNAs among the plasma samples tested. Our data indicate that the prevalence of EBVaGC among Iranian patients with gastric cancer is lower than the global prevalence and although none of the EBV miRNAs were detected in plasma, evaluation of EBV microRNAs in tumor tissue, especially miR-BART7-3p, may constitute useful biomarkers for diagnosis of EBVaGC.
Collapse
Affiliation(s)
- Behrang Sarshari
- Department of Medical Virology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
- Iranian Research Center for HIV/AIDS, Iranian Institute for Reduction of High Risk Behaviors, Tehran University of Medical Sciences, Tehran, Iran
| | - Mehrdad Ravanshad
- Department of Medical Virology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran.
| | - Amirhassan Rabbani
- Department of Transplant & Hepatobiliary Surgery, Taleghani Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Raziyeh Zareh-Khoshchehreh
- Department of Medical Virology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
- Food and Drug Administration, Iran Ministry of Health and Medical Education, Tehran, Iran
| | - Fedra Mokhtari
- Basic and Molecular Epidemiology of Gastrointestinal Disorders Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Binazir Khanabadi
- Basic and Molecular Epidemiology of Gastrointestinal Disorders Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Seyed Reza Mohebbi
- Basic and Molecular Epidemiology of Gastrointestinal Disorders Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Hamid Asadzadeh Aghdaei
- Basic and Molecular Epidemiology of Gastrointestinal Disorders Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
5
|
Thakur A, Kumar M. Integration of Human and Viral miRNAs in Epstein-Barr Virus-Associated Tumors and Implications for Drug Repurposing. OMICS : A JOURNAL OF INTEGRATIVE BIOLOGY 2023; 27:93-108. [PMID: 36927073 DOI: 10.1089/omi.2023.0005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/18/2023]
Abstract
Epstein-Barr virus (EBV) is associated with several tumors, and has substantial relevance for public health. Therapeutics innovation for EBV-related disorders is much needed. In this context, miRNAs are noncoding RNA molecules that play vital roles in EBV infection. miRNA-Seq and RNA-Seq data for EBV-associated clinical samples and cell lines have been generated, but their detailed integrative analyses, and exploitation for drug repurposing against EBV are lacking. Hence, we identified and analyzed the differentially expressed miRNAs (DEmiRs) in EBV-infected cell lines (28) and infected (28) and uninfected human tissue (20) samples using an in-house pipeline. We found significantly enriched host miRNAs like hsa-mir-3651, hsa-mir-1248, and hsa-mir-29c-3p in EBV-infected samples from EBV-associated nasopharyngeal carcinoma and Hodgkin's lymphoma, among others. Furthermore, we also identified significantly enriched novel miRNAs such as hsa-mir-29c-3p, hsa-mir-3651, and hsa-mir-98-3p, which were not previously reported in EBV-related tumors. Differentially expressed mRNAs (DEMs) were identified in EBV-infected cell lines (21) and uninfected human tissue (14) samples. We predicted and selected 1572 DEMs (upregulated) that are targeted by 547 DEmiRs (downregulated). These were further classified into essential (870) and nonessential (702) genes. Moreover, a miRNA-mRNA network was developed for the hub miRNAs. Importantly, we used the DEMs during EBV latent infection types I, II, and III to identify the candidate drugs for repurposing: Glyburide, Levodopa, Nateglinide, and Stiripentol, among others. To the best of our knowledge, this is the first integrative analyses that identified DEmiRs and DEMs as potential therapeutic targets and predicted drugs as potential candidates for repurposing against EBV-related tumors.
Collapse
Affiliation(s)
- Anamika Thakur
- Virology Unit and Bioinformatics Centre, Institute of Microbial Technology, Council of Scientific and Industrial Research (CSIR), Sector 39-A, Chandigarh, India.,Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| | - Manoj Kumar
- Virology Unit and Bioinformatics Centre, Institute of Microbial Technology, Council of Scientific and Industrial Research (CSIR), Sector 39-A, Chandigarh, India.,Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| |
Collapse
|
6
|
Wen KW, Wang L, Menke JR, Damania B. Cancers associated with human gammaherpesviruses. FEBS J 2022; 289:7631-7669. [PMID: 34536980 PMCID: PMC9019786 DOI: 10.1111/febs.16206] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2021] [Revised: 08/10/2021] [Accepted: 09/16/2021] [Indexed: 01/14/2023]
Abstract
Epstein-Barr virus (EBV; human herpesvirus 4; HHV-4) and Kaposi sarcoma-associated herpesvirus (KSHV; human herpesvirus 8; HHV-8) are human gammaherpesviruses that have oncogenic properties. EBV is a lymphocryptovirus, whereas HHV-8/KSHV is a rhadinovirus. As lymphotropic viruses, EBV and KSHV are associated with several lymphoproliferative diseases or plasmacytic/plasmablastic neoplasms. Interestingly, these viruses can also infect epithelial cells causing carcinomas and, in the case of KSHV, endothelial cells, causing sarcoma. EBV is associated with Burkitt lymphoma, classic Hodgkin lymphoma, nasopharyngeal carcinoma, plasmablastic lymphoma, lymphomatoid granulomatosis, leiomyosarcoma, and subsets of diffuse large B-cell lymphoma, post-transplant lymphoproliferative disorder, and gastric carcinoma. KSHV is implicated in Kaposi sarcoma, primary effusion lymphoma, multicentric Castleman disease, and KSHV-positive diffuse large B-cell lymphoma. Pathogenesis by these two herpesviruses is intrinsically linked to viral proteins expressed during the lytic and latent lifecycles. This comprehensive review intends to provide an overview of the EBV and KSHV viral cycles, viral proteins that contribute to oncogenesis, and the current understanding of the pathogenesis and clinicopathology of their related neoplastic entities.
Collapse
Affiliation(s)
- Kwun Wah Wen
- Department of Pathology and Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, CA 94158
| | - Linlin Wang
- Department of Laboratory Medicine, University of California, San Francisco, CA 94158
| | - Joshua R. Menke
- Department of Pathology, Stanford University, Palo Alto, CA 94304
| | - Blossom Damania
- Department of Microbiology & Immunology & Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC 27599
| |
Collapse
|
7
|
Zhang T, Chen Z, Deng J, Xu K, Che D, Lin J, Jiang P, Gu X, Xu B. Epstein-Barr virus-encoded microRNA BART22 serves as novel biomarkers and drives malignant transformation of nasopharyngeal carcinoma. Cell Death Dis 2022; 13:664. [PMID: 35907914 PMCID: PMC9338958 DOI: 10.1038/s41419-022-05107-x] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Revised: 07/14/2022] [Accepted: 07/15/2022] [Indexed: 01/21/2023]
Abstract
Nasopharyngeal carcinoma (NPC) is an epithelial malignancy ubiquitously associated with Epstein-Barr virus (EBV). EBV generates various viral microRNAs (miRNAs) by processing the BHRF1 and BamHI A rightward (BART) transcripts. These BART miRNAs are abundantly expressed in NPC, but their functions and molecular mechanisms remain largely unknown. Our study found that the EBV-encoded microRNA BART-22 was significantly upregulated in NPC tissues and positively correlated with tumor progression. Furthermore, we found that EBV-miR-BART-22 was a significant predictor of poor prognosis in NPC. A reliable nomogram model to predict the preoperative overall survival (OS) of NPC patients was established. The area under the receiver operating characteristic (ROC) curve value for 5-year survival was 0.91. Elevated levels of EBV-miR-BART-22 significantly promoted the epithelial-mesenchymal transition (EMT) and metastasis of NPC cells in vivo and in vitro. We found that EBV-miR-BART-22 directly targets the 3'-UTR of MOSPD2 mRNA to promote the EMT and metastasis of NPC cells by activating the Wnt/β-catenin signaling pathway. Our findings provide a potential prognostic biomarker and new insight into the molecular mechanisms of NPC metastasis.
Collapse
Affiliation(s)
- Ting Zhang
- Department of Laboratory Medicine, the Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou, 510180, China
| | - Zui Chen
- Department of Laboratory Medicine, the Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou, 510180, China
| | - Jing Deng
- Department of Laboratory Medicine, the Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou, 510180, China
| | - Kaixiong Xu
- Department of Laboratory Medicine, the Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou, 510180, China
| | - Di Che
- Department of Clinical Biological Resource Bank, Guangzhou Institute of Pediatrics, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, China
| | - Jiamin Lin
- Department of Laboratory Medicine, the Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou, 510180, China
| | - Ping Jiang
- Department of Laboratory Medicine, the Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou, 510180, China
| | - Xiaoqiong Gu
- Department of Clinical Biological Resource Bank, Guangzhou Institute of Pediatrics, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, China.
| | - Banglao Xu
- Department of Laboratory Medicine, the Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou, 510180, China.
| |
Collapse
|
8
|
Lin C, Lin K, Zhang B, Su Y, Guo Q, Lu T, Xu Y, Lin S, Zong J, Pan J. OUP accepted manuscript. Oncologist 2022; 27:e340-e349. [PMID: 35380720 PMCID: PMC8982379 DOI: 10.1093/oncolo/oyac024] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2020] [Accepted: 11/13/2021] [Indexed: 11/23/2022] Open
Abstract
Background Nasopharyngeal carcinoma is an Epstein-Barr virus (EBV)-associated tumor that is highly common in southern China. Our previous sequencing data demonstrated that the EBV-encoded microRNA BART8-3p was most upregulated in nasopharyngeal carcinoma (NPC) and was closely associated with the metastasis of NPC. However, the values of plasma BART8-3p in NPC patients have not yet been well characterized. Material and Methods We quantified plasma BART8-3p expression by quantitative real-time PCR in 205 newly diagnosed NPC patients. Kaplan-Meier analysis was used to compare overall survival (OS), distant metastasis-free survival (DMFS), and locoregional relapse-free survival (LRRFS) between the groups. Results Plasma pretreatment BART8-3p was highly expressed in NPC patients compared with healthy controls. Pretreatment BART8-3p yielded a 92% predictive value for detecting NPC. Importantly, BART8-3p decreased dramatically after therapy relative to pretreatment levels. High levels of pretreatment or post-treatment BART8-3p were associated with worse OS, DMFS, and LRRFS. Multivariate analysis showed that high pretreatment or post-treatment BART8-3p was an independent unfavorable prognostic marker for OS (HR 3.82, 95% CI 1.77-8.24, P = .001 or HR 2.74, 95% CI 1.27-5.91, P = .010), DMFS (HR 2.82, 95% CI 1.36-5.85, P = .005 or HR 3.27, 95% CI 1.57-6.81, P = .002), and LRRFS (HR 1.94, 95% CI 1.12-3.35, P = .018 or HR 2.03, 95% CI 1.14-3.62, P = .016) in NPC. Subgroup analysis revealed that for patients with locally advanced NPC with high levels of pretreatment BART8-3p (n = 58), more cycles of chemotherapy (≥6 cycles) tended to prolong OS (P = .070). Over 50% (6/11) patients with high levels of post-treatment BART8-3p presented distant metastasis. Conclusion Plasma BART8-3p is a promising biomarker for the detection and prognosis of NPC.
Collapse
Affiliation(s)
- Cheng Lin
- Department of Radiation Oncology, Fujian Medical University Cancer Hospital & Fujian Cancer Hospital, Fuzhou, People’s Republic of China
- The School of Clinical Medicine and Fujian Medical University, Fuzhou, People’s Republic of China
| | - Keyu Lin
- Department of Radiation Biology, Fujian Medical University Cancer Hospital & Fujian Cancer Hospital, Fuzhou, People’s Republic of China
| | - Bin Zhang
- Department of Genetics and Genomic Sciences, Icahn Institute of Genomics and Multiscale Biology, Mount Sinai Center for Transformative Disease Modeling, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Ying Su
- Department of Radiation Biology, Fujian Medical University Cancer Hospital & Fujian Cancer Hospital, Fuzhou, People’s Republic of China
| | - Qiaojuan Guo
- Department of Radiation Oncology, Fujian Medical University Cancer Hospital & Fujian Cancer Hospital, Fuzhou, People’s Republic of China
| | - Tianzhu Lu
- Department of Radiation Oncology, Fujian Medical University Cancer Hospital & Fujian Cancer Hospital, Fuzhou, People’s Republic of China
| | - Yuanji Xu
- Department of Radiation Oncology, Fujian Medical University Cancer Hospital & Fujian Cancer Hospital, Fuzhou, People’s Republic of China
| | - Shaojun Lin
- Department of Radiation Oncology, Fujian Medical University Cancer Hospital & Fujian Cancer Hospital, Fuzhou, People’s Republic of China
| | - Jingfeng Zong
- Department of Radiation Oncology, Fujian Medical University Cancer Hospital & Fujian Cancer Hospital, Fuzhou, People’s Republic of China
- Corresponding author: Jianji Pan and Jingfeng Zong, Department of Radiation Oncology, Fujian Cancer Hospital & Fujian Medical University Cancer Hospital No. 420 Fuma Road, Fuzhou 350014, People’s Republic of China. ;
| | - Jianji Pan
- Department of Radiation Oncology, Fujian Medical University Cancer Hospital & Fujian Cancer Hospital, Fuzhou, People’s Republic of China
- Corresponding author: Jianji Pan and Jingfeng Zong, Department of Radiation Oncology, Fujian Cancer Hospital & Fujian Medical University Cancer Hospital No. 420 Fuma Road, Fuzhou 350014, People’s Republic of China. ;
| |
Collapse
|
9
|
Zhu QY, Zhao GX, Li Y, Talakatta G, Mai HQ, Le QT, Young LS, Zeng MS. Advances in pathogenesis and precision medicine for nasopharyngeal carcinoma. MedComm (Beijing) 2021; 2:175-206. [PMID: 34766141 PMCID: PMC8491203 DOI: 10.1002/mco2.32] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2020] [Revised: 07/28/2020] [Accepted: 07/29/2020] [Indexed: 12/13/2022] Open
Abstract
Nasopharyngeal carcinoma (NPC) is a squamous carcinoma with apparent geographical and racial distribution, mostly prevalent in East and Southeast Asia, particularly concentrated in southern China. The epidemiological trend over the past decades has suggested a substantial reduction in the incidence rate and mortality rate due to NPC. These results may reflect changes in lifestyle and environment, and more importantly, a deeper comprehension of the pathogenic mechanism of NPC, leading to much progress in the preventing, screening, and treating for this cancer. Herein, we present the recent advances on the key signal pathways involved in pathogenesis of NPC, the mechanism of Epstein‐Barr virus (EBV) entry into the cell, and the progress of EBV vaccine and screening biomarkers. We will also discuss in depth the development of various therapeutic approaches including radiotherapy, chemotherapy, surgery, targeted therapy, and immunotherapy. These research advancements have led to a new era of precision medicine in NPC.
Collapse
Affiliation(s)
- Qian-Ying Zhu
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy Sun Yat-sen University Cancer Center (SYSUCC) Guangzhou China
| | - Ge-Xin Zhao
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy Sun Yat-sen University Cancer Center (SYSUCC) Guangzhou China
| | - Yan Li
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy Sun Yat-sen University Cancer Center (SYSUCC) Guangzhou China
| | - Girish Talakatta
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy Sun Yat-sen University Cancer Center (SYSUCC) Guangzhou China
| | - Hai-Qiang Mai
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy Sun Yat-sen University Cancer Center (SYSUCC) Guangzhou China
| | - Quynh-Thu Le
- Department of Radiation Oncology Stanford California
| | - Lawrence S Young
- Warwick Medical School University of Warwick Coventry United Kingdom
| | - Mu-Sheng Zeng
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy Sun Yat-sen University Cancer Center (SYSUCC) Guangzhou China
| |
Collapse
|
10
|
Zhang YJ, Mu ZL, Deng P, Liang YD, Wu LC, Yang LL, Zhou Z, Yu ZP. 8-Formylophiopogonanone B induces ROS-mediated apoptosis in nasopharyngeal carcinoma CNE-1 cells. Toxicol Res (Camb) 2021; 10:1052-1063. [PMID: 34733490 DOI: 10.1093/toxres/tfab087] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2021] [Revised: 07/28/2021] [Accepted: 08/05/2021] [Indexed: 12/24/2022] Open
Abstract
Cancer is one of the leading causes of death in the world. It is very important to find drugs with high efficiency, low toxicity, and low side effects for the treatment of cancer. Flavonoids and their derivatives with broad biological functions have been recognized as anti-tumor chemicals. 8-Formylophiopogonanone B (8-FOB), a naturally existed homoisoflavonoids with rarely known biological functions, needs pharmacological evaluation. In order to explore the possible anti-tumor action of 8-FOB, we used six types of tumor cells to evaluate in vitro effects of this agent on cell viability and tested the effects on clone formation ability, scratching wound-healing, and apoptosis. In an attempt to elucidate the mechanism of pharmacological action, we examined 8-FOB-induced intracellular oxidative stress and -disrupted mitochondrial function. Results suggested that 8-FOB could suppress tumor cell viability, inhibit cell migration and invasion, induce apoptosis, and elicit intracellular ROS production. Among these six types of tumor cells, the nasopharyngeal carcinoma CNE-1 cells were the most sensitive cancer cells to 8-FOB treatment. Intracellular ROS production played a pivotal role in the anti-tumor action of 8-FOB. Our present study is the first to document that 8-FOB has anti-tumor activity in vitro and increases intracellular ROS production, which might be responsible for its anti-tumor action. The anti-tumor pharmacological effect of 8-FOB is worthy of further investigation.
Collapse
Affiliation(s)
- Ya-Jing Zhang
- Medical College, Guangxi University, 100 University East Road, Xixiangtang District, Nanning, Guangxi, 530004, P. R. China
| | - Zhen-Lin Mu
- Medical College, Guangxi University, 100 University East Road, Xixiangtang District, Nanning, Guangxi, 530004, P. R. China
| | - Ping Deng
- Department of Occupational Health, Third Military Medical University, 30 Gaotanyan Zhengjie, Shapingba District, Chongqing, 400038, P. R. China
| | - Yi-Dan Liang
- Medical College, Guangxi University, 100 University East Road, Xixiangtang District, Nanning, Guangxi, 530004, P. R. China
| | - Li-Chuan Wu
- Medical College, Guangxi University, 100 University East Road, Xixiangtang District, Nanning, Guangxi, 530004, P. R. China
| | - Ling-Ling Yang
- Department of Occupational Health, Third Military Medical University, 30 Gaotanyan Zhengjie, Shapingba District, Chongqing, 400038, P. R. China
| | - Zhou Zhou
- Department of Environmental Medicine, and Department of Emergency Medicine of the First Affiliated Hospital, Zhejiang University, 866 Yuhangtang Road, Xihu District, Hangzhou, Zhejiang, 310000, P. R. China
| | - Zheng-Ping Yu
- Medical College, Guangxi University, 100 University East Road, Xixiangtang District, Nanning, Guangxi, 530004, P. R. China
| |
Collapse
|
11
|
Cui X, Snapper CM. Epstein Barr Virus: Development of Vaccines and Immune Cell Therapy for EBV-Associated Diseases. Front Immunol 2021; 12:734471. [PMID: 34691042 PMCID: PMC8532523 DOI: 10.3389/fimmu.2021.734471] [Citation(s) in RCA: 100] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Accepted: 09/15/2021] [Indexed: 11/13/2022] Open
Abstract
Epstein-Barr virus (EBV) is the first human tumor virus discovered and is strongly implicated in the etiology of multiple lymphoid and epithelial cancers. Each year EBV associated cancers account for over 200,000 new cases of cancer and cause 150,000 deaths world-wide. EBV is also the primary cause of infectious mononucleosis, and up to 70% of adolescents and young adults in developed countries suffer from infectious mononucleosis. In addition, EBV has been shown to play a critical role in the pathogenesis of multiple sclerosis. An EBV prophylactic vaccine that induces neutralizing antibodies holds great promise for prevention of EBV associated diseases. EBV envelope proteins including gH/gL, gB and gp350 play key roles in EBV entry and infection of target cells, and neutralizing antibodies elicited by each of these proteins have shown to prevent EBV infection of target cells and markedly decrease EBV titers in the peripheral blood of humanized mice challenged with lethal dose EBV. Recent studies demonstrated that immunization with the combination of gH/gL, gB and/or gp350 induced markedly increased synergistic EBV neutralizing activity compared to immunization with individual proteins. As previous clinical trials focused on gp350 alone were partially successful, the inclusion of gH/gL and gB in a vaccine formulation with gp350 represents a promising approach of EBV prophylactic vaccine development. Therapeutic EBV vaccines have also been tested clinically with encouraging results. Immunization with various vaccine platforms expressing the EBV latent proteins EBNA1, LMP1, and/or LMP2 promoted specific CD4+ and CD8+ cytotoxic responses with anti-tumor activity. The addition of EBV envelope proteins gH/gL, gB and gp350 has the potential to increase the efficacy of a therapeutic EBV vaccine. The immune system plays a critical role in the control of tumors, and immune cell therapy has emerged as a promising treatment of cancers. Adoptive T-cell therapy has been successfully used in the prevention and treatment of post-transplant lymphoproliferative disorder. Chimeric antigen receptor T cell therapy and T cell receptor engineered T cell therapy targeting EBV latent proteins LMP1, LMP2 and/or EBNA1 have been in development, with the goal to increase the specificity and efficacy of treatment of EBV associated cancers.
Collapse
Affiliation(s)
- Xinle Cui
- Genitourinary Malignancies Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States.,The Institute for Vaccine Research and Department of Pathology, Uniformed Services University of the Health Sciences, Bethesda, MD, United States
| | - Clifford M Snapper
- The Institute for Vaccine Research and Department of Pathology, Uniformed Services University of the Health Sciences, Bethesda, MD, United States.,Citranvi Biosciences LLC, Chapel Hill, NC, United States
| |
Collapse
|
12
|
PPRV-induced novel miR-3 contributes to inhibit type I IFN production by targeting IRAK1. J Virol 2021; 95:JVI.02045-20. [PMID: 33504605 PMCID: PMC8103702 DOI: 10.1128/jvi.02045-20] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023] Open
Abstract
Peste des petits ruminants virus (PPRV) is an important pathogen that seriously influences the productivity of small ruminants worldwide. PPRV has evolved several mechanisms to evade IFN-I responses. We report that a novel microRNA in goat PBMCs, novel miR-3, was upregulated by PPRV to facilitate virus infection. Furthermore, PPRV V protein alone was sufficient to induce novel miR-3 expression, and NF-κB and p38 pathway may involved in the induction of novel miR-3 during PPRV infection. Importantly, we demonstrated that novel miR-3 was a potent negative regulator of IFN-α production by targeting IRAK1, which resulted in the enhancement of PPRV infection. In addition, we found that PPRV infection can activated ISGs through IFN independent and IRF3 dependent pathway. Moreover, our data revealed that novel miR-3 mediated regulation of IFN-α production may involve in the differential susceptibility between goat and sheep to PPRV. Taken together, our findings identified a new strategy taken by PPRV to escape IFN-I-mediated antiviral immune responses by engaging cellular microRNA and, thus, improve our understanding of its pathogenesis.IMPORTANCE: Peste des petits ruminants virus (PPRV) induce in the hosts a transient but severe immunosuppression, which threatens both small livestock and endangered susceptible wildlife populations in many countries. Despite extensive research has been explored, the mechanism underlying PPRV immune system evasion remains elusive. Our data provided the first direct evidence that novel microRNA-3 (novel miR-3) feedback inhibits type I IFN signaling when goat PBMCs are infected with PPRV vaccine strain N75/1, thus promoting the infection. In this study, the target of novel miR-3, IRAK1, which are important for PPRV-induced type I IFN production, have also been found. Moreover, we identified NF-κB and p38 pathways may involve in novel miR-3 induction in response to PPRV infection. Taken together, our research has provided new insight into understanding the effects of miRNA on host-virus interactions, and revealed a potential therapeutic target for antiviral intervention.
Collapse
|
13
|
Liao Y, Zhuang G, Sun A, Khan OA, Lupiani B, Reddy SM. Marek's Disease Virus Cluster 3 miRNAs Restrict Virus' Early Cytolytic Replication and Pathogenesis. Viruses 2020; 12:v12111317. [PMID: 33212952 PMCID: PMC7698348 DOI: 10.3390/v12111317] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Revised: 11/12/2020] [Accepted: 11/13/2020] [Indexed: 12/21/2022] Open
Abstract
Herpesvirus-encoded microRNAs (miRNAs) have been discovered in infected cells; however, lack of a suitable animal model has hampered functional analyses of viral miRNAs in vivo. Marek’s disease virus (MDV) (Gallid alphaherpesvirus 2, GaHV-2) genome contains 14 miRNA precursors, which encode 26 mature miRNAs, grouped into three clusters. In this study, the role of MDV-encoded cluster 3 miRNAs, also known as mdv1-miR-M8-M10, in pathogenesis was evaluated in chickens, the natural host of MDV. Our results show that deletion of cluster 3 miRNAs did not affect virus replication and plaque size in cell culture, but increased early cytolytic replication of MDV in chickens. We also observed that deletion of cluster 3 miRNAs resulted in significantly higher virus reactivation from peripheral blood lymphocytes. In addition, pathogenesis studies showed that deletion of cluster 3 miRNAs resulted in more severe atrophy of lymphoid organs and reduced mean death time, but did not affect the incidence of MDV-associated visceral tumors. We confirmed these results by generating a cluster 3 miRNA revertant virus in which the parental MDV phenotype was restored. To the best of our knowledge, our study provides the first evidence that MDV cluster 3 miRNAs play an important role in modulating MDV pathogenesis.
Collapse
|
14
|
Kang Y, He W, Ren C, Qiao J, Guo Q, Hu J, Xu H, Jiang X, Wang L. Advances in targeted therapy mainly based on signal pathways for nasopharyngeal carcinoma. Signal Transduct Target Ther 2020; 5:245. [PMID: 33093441 PMCID: PMC7582884 DOI: 10.1038/s41392-020-00340-2] [Citation(s) in RCA: 53] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2020] [Revised: 09/12/2020] [Accepted: 09/16/2020] [Indexed: 02/07/2023] Open
Abstract
Nasopharyngeal carcinoma (NPC) is a malignant epithelial carcinoma of the head and neck region which mainly distributes in southern China and Southeast Asia and has a crucial association with the Epstein-Barr virus. Based on epidemiological data, both incidence and mortality of NPC have significantly declined in recent decades grounded on the improvement of living standard and medical level in an endemic region, in particular, with the clinical use of individualized chemotherapy and intensity-modulated radiotherapy (IMRT) which profoundly contributes to the cure rate of NPC patients. To tackle the challenges including local recurrence and distant metastasis in the current NPC treatment, we discussed the implication of using targeted therapy against critical molecules in various signal pathways, and how they synergize with chemoradiotherapy in the NPC treatment. Combination treatment including targeted therapy and IMRT or concurrent chemoradiotherapy is presumably to be future options, which may reduce radiation or chemotherapy toxicities and open new avenues for the improvement of the expected functional outcome for patients with advanced NPC.
Collapse
Affiliation(s)
- Yuanbo Kang
- Department of Neurosurgery, Cancer Research Institute, Xiangya Hospital, Central South University, 410008, Changsha, Hunan, China
- Cancer Research Institute, Collaborative Innovation Center for Cancer Medicine, School of Basic Medical Science, Central South University, 410008, Changsha, Hunan, China
- The NHC Key Laboratory of Carcinogenesis and The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Xiangya Hospital, Central South University, 410008, Changsha, Hunan, China
| | - Weihan He
- Department of Neurosurgery, Cancer Research Institute, Xiangya Hospital, Central South University, 410008, Changsha, Hunan, China
- Cancer Research Institute, Collaborative Innovation Center for Cancer Medicine, School of Basic Medical Science, Central South University, 410008, Changsha, Hunan, China
- The NHC Key Laboratory of Carcinogenesis and The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Xiangya Hospital, Central South University, 410008, Changsha, Hunan, China
| | - Caiping Ren
- Department of Neurosurgery, Cancer Research Institute, Xiangya Hospital, Central South University, 410008, Changsha, Hunan, China.
- Cancer Research Institute, Collaborative Innovation Center for Cancer Medicine, School of Basic Medical Science, Central South University, 410008, Changsha, Hunan, China.
- The NHC Key Laboratory of Carcinogenesis and The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Xiangya Hospital, Central South University, 410008, Changsha, Hunan, China.
| | - Jincheng Qiao
- Department of Neurosurgery, Cancer Research Institute, Xiangya Hospital, Central South University, 410008, Changsha, Hunan, China
- Cancer Research Institute, Collaborative Innovation Center for Cancer Medicine, School of Basic Medical Science, Central South University, 410008, Changsha, Hunan, China
- The NHC Key Laboratory of Carcinogenesis and The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Xiangya Hospital, Central South University, 410008, Changsha, Hunan, China
| | - Qiuyong Guo
- Department of Neurosurgery, Cancer Research Institute, Xiangya Hospital, Central South University, 410008, Changsha, Hunan, China
- Cancer Research Institute, Collaborative Innovation Center for Cancer Medicine, School of Basic Medical Science, Central South University, 410008, Changsha, Hunan, China
- The NHC Key Laboratory of Carcinogenesis and The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Xiangya Hospital, Central South University, 410008, Changsha, Hunan, China
| | - Jingyu Hu
- Department of Neurosurgery, Cancer Research Institute, Xiangya Hospital, Central South University, 410008, Changsha, Hunan, China
- Cancer Research Institute, Collaborative Innovation Center for Cancer Medicine, School of Basic Medical Science, Central South University, 410008, Changsha, Hunan, China
- The NHC Key Laboratory of Carcinogenesis and The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Xiangya Hospital, Central South University, 410008, Changsha, Hunan, China
| | - Hongjuan Xu
- Department of Neurosurgery, Cancer Research Institute, Xiangya Hospital, Central South University, 410008, Changsha, Hunan, China
- Cancer Research Institute, Collaborative Innovation Center for Cancer Medicine, School of Basic Medical Science, Central South University, 410008, Changsha, Hunan, China
- The NHC Key Laboratory of Carcinogenesis and The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Xiangya Hospital, Central South University, 410008, Changsha, Hunan, China
| | - Xingjun Jiang
- Department of Neurosurgery, Cancer Research Institute, Xiangya Hospital, Central South University, 410008, Changsha, Hunan, China
| | - Lei Wang
- Department of Neurosurgery, Cancer Research Institute, Xiangya Hospital, Central South University, 410008, Changsha, Hunan, China.
- Cancer Research Institute, Collaborative Innovation Center for Cancer Medicine, School of Basic Medical Science, Central South University, 410008, Changsha, Hunan, China.
- The NHC Key Laboratory of Carcinogenesis and The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Xiangya Hospital, Central South University, 410008, Changsha, Hunan, China.
| |
Collapse
|
15
|
Wardana T, Gunawan L, Herawati C, Oktriani R, Anwar SL, Astuti I, Aryandhono T, Mubarika S. Circulation EBV Mir-Bart-7 Relating to Clinical Manifestation in Nasopharyngeal Carcinoma. Asian Pac J Cancer Prev 2020; 21:2777-2782. [PMID: 32986380 PMCID: PMC7779452 DOI: 10.31557/apjcp.2020.21.9.2777] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2020] [Accepted: 09/25/2020] [Indexed: 12/02/2022] Open
Abstract
OBJECTIVE Nasopharyngeal Carcinoma (NPC) is an endemic head and neck malignancy in Asia Pacific regions that is associated with chronic infection by Epstein-Barr virus (EBV). EBV miR-BART-7 is a microRNA (miRNA) encoded by EBV that regulates malignant behavior of NPC. However, the role and function of miR-BART7 are not clear, particularly the relation of circulating levels and patient's clinical presentation. METHODS Circulating miR-BART-7 levels were measured by using qRT-PCR and were correlated with clinical and pathological data. RESULT Of 52 NPC patients included in this study, 85% were diagnosed in the late stages (Stage III-IV). 73% of tumors were non-keratinizing undifferentiated NPC, 92% of tumors were WHO class III histology and all cases were EBV-IgA positive. Over-expression of miR-BART7-3p was correlated with positive regional lymph nodes in newly diagnosed (4.61 fold changes, p <0.05). CONCLUSION Over-expression of circulating EBV miR-BART7 correlated with positive regional lymph nodes reflecting the diagnostic and prognostic values of circulating miR-BART7 for patients with NPC.
Collapse
Affiliation(s)
- Tirta Wardana
- Department of Biomedicine, School of Dentistry, Faculty of Medicine, Jenderal Soedirman University, Purwokerto, Indonesia.
| | - Lisa Gunawan
- Postgraduate Student, Center for Biotechnology, Universitas Gadjah Mada, Yogyakarta, Indonesia.
| | - Cita Herawati
- Department of Ear, Nose and Throat, Dharmais National Cancer Hospital, Jakarta, Indonesia.
| | - Risky Oktriani
- Department of Biochemistry, Faculty of Medicine, Public Health and Nursing, Universitas Gadjah Mada, Yogyakarta, Indonesia.
| | - Sumadi Lukman Anwar
- Division of Surgical Oncology, Department of Surgery, Faculty of Medicine, Public Health and Nursing, Universitas Gadjah Mada, Yogyakarta, Indonesia.
| | - Indwiani Astuti
- Department of Pharmacology and Therapy, Faculty of Medicine, Public Health and Nursing, Universitas Gadjah Mada, Yogyakarta, Indonesia.
| | - Teguh Aryandhono
- Division of Surgical Oncology, Department of Surgery, Faculty of Medicine, Public Health and Nursing, Universitas Gadjah Mada, Yogyakarta, Indonesia.
| | - Sofia Mubarika
- Department of Histology and Cell Biology, Faculty of Medicine, Public Health and Nursing, Universitas Gadjah Mada, Yogyakarta, Indonesia.
| |
Collapse
|
16
|
Jiang C, Cheng Z, Jiang T, Xu Y, Wang B. MicroRNA-34a inhibits cell invasion and epithelial-mesenchymal transition via targeting AXL/PI3K/AKT/Snail signaling in nasopharyngeal carcinoma. Genes Genomics 2020; 42:971-978. [PMID: 32648233 DOI: 10.1007/s13258-020-00963-3] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2019] [Accepted: 06/29/2020] [Indexed: 01/19/2023]
Abstract
BACKGROUND MicroRNA-34a (miR-34a) has been reported to inhibit TGF-β (transforming growth factor-β)-induced epithelial-mesenchymal transition (EMT) in nasopharyngeal carcinoma (NPC). However, the underlying mechanism remain unclear. Using the bioinformatics, we found that the AXL receptor tyrosine kinase (AXL) is a predicted target of miR-34a. OBJECTIVE we aimed to reveal the relationship between miR-34a and AXL, and investigate the effect and mechanism of miR-34a in NPC progression. METHODS The expression patterns of miR-34a and AXL in 30 paired NPC tissues and the adjacent tissues were examined by quantitative real time PCR (qRT-PCR). The target relationship between miR-34a and AXL was evaluated by the luciferase gene reporter assay. Cell migration and invasion were assessed by wound healing and transwell chamber assays, respectively. RESULTS miR-34a level was dramatically decreased in the NPC tissues compared to the adjacent tissues, while AXL expression was increased. Overexpression of miR-34a significantly reduced the luciferase activity of the luciferase vector of AXL (pGL3-AXL-WT), whereas this effect was abrogated when binding sites between miR-34a and AXL were mutated. In addition, ectopic expression of miR-34a dramatically inhibited Sune-1 cell migration and invasion abilities, decreased the levels of N-cadherin and Vimentin and increased E-cadherin and γ-catenin expressions, as well as induced significant reductions in the expressions of p-AKT and Snail. However, these effects were attenuated when the cells were treated with recombinant human AXL protein. CONCLUSIONS Our results demonstrate that miR-34a/AXL can inhibit NPC cell migration, invasion and EMT through inhibition of AKT/Snail signaling.
Collapse
Affiliation(s)
- Chengyi Jiang
- Department of Otolaryngology Head and Neck Surgery, The First Affiliated Hospital of Bengbu Medical College, No. 287 Changhuai Road, Bengbu City, 233004, Anhui Province, China.
| | - Zhongqiang Cheng
- Department of Otolaryngology Head and Neck Surgery, The First Affiliated Hospital of Bengbu Medical College, No. 287 Changhuai Road, Bengbu City, 233004, Anhui Province, China
| | - Tao Jiang
- Department of Otolaryngology Head and Neck Surgery, The First Affiliated Hospital of Bengbu Medical College, No. 287 Changhuai Road, Bengbu City, 233004, Anhui Province, China
| | - Yajia Xu
- Department of Otolaryngology Head and Neck Surgery, The First Affiliated Hospital of Bengbu Medical College, No. 287 Changhuai Road, Bengbu City, 233004, Anhui Province, China
| | - Bin Wang
- Department of Otolaryngology Head and Neck Surgery, The First Affiliated Hospital of Bengbu Medical College, No. 287 Changhuai Road, Bengbu City, 233004, Anhui Province, China
| |
Collapse
|
17
|
Tagawa T, Serquiña A, Kook I, Ziegelbauer J. Viral non-coding RNAs: Stealth strategies in the tug-of-war between humans and herpesviruses. Semin Cell Dev Biol 2020; 111:135-147. [PMID: 32631785 DOI: 10.1016/j.semcdb.2020.06.015] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2020] [Revised: 06/20/2020] [Accepted: 06/24/2020] [Indexed: 12/23/2022]
Abstract
Oncogenic DNA viruses establish lifelong infections in humans, and they cause cancers, often in immunocompromised patients, despite anti-viral immune surveillance targeted against viral antigens. High-throughput sequencing techniques allowed the field to identify novel viral non-coding RNAs (ncRNAs). ncRNAs are ideal factors for DNA viruses to exploit; they are non-immunogenic to T cells, thus viral ncRNAs can manipulate host cells without evoking adaptive immune responses. Viral ncRNAs may still trigger the host innate immune response, but many viruses encode decoys/inhibitors to counter-act and evade recognition. In addition, ncRNAs can be secreted to the extracellular space and influence adjacent cells to create a pro-viral microenvironment. In this review, we present recent progress in understanding interactions between oncoviruses and ncRNAs including small and long ncRNAs, microRNAs, and recently identified viral circular RNAs. In addition, potential clinical applications for ncRNA will be discussed. Extracellular ncRNAs are suggested to be diagnostic and prognostic biomarkers and, with the realization of the importance of viral ncRNAs in tumorigenesis, approaches to target critical viral ncRNAs are emerging. Further understanding of viral utilization of ncRNAs will advance anti-viral therapeutics beyond conventional medication and vaccination.
Collapse
Affiliation(s)
- Takanobu Tagawa
- HIV and AIDS Malignancy Branch, National Cancer Institute, Bethesda, Maryland 20892, United States
| | - Anna Serquiña
- HIV and AIDS Malignancy Branch, National Cancer Institute, Bethesda, Maryland 20892, United States
| | - Insun Kook
- HIV and AIDS Malignancy Branch, National Cancer Institute, Bethesda, Maryland 20892, United States
| | - Joseph Ziegelbauer
- HIV and AIDS Malignancy Branch, National Cancer Institute, Bethesda, Maryland 20892, United States.
| |
Collapse
|
18
|
Predictive Value of CD8 Expression and FoxP3 Methylation in Nasopharyngeal Carcinoma Patients Treated with Chemoradiotherapy in a Non-endemic Area. Pathol Oncol Res 2020; 26:2459-2467. [PMID: 32564263 DOI: 10.1007/s12253-020-00859-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/01/2019] [Accepted: 06/16/2020] [Indexed: 10/24/2022]
Abstract
Undifferentiated Nasopharyngeal Carcinoma (UNPC) is associated with Epstein-Barr Virus (EBV) and characterized by an abundant immune infiltrate potentially influencing the prognosis. Thus, we retrospectively assessed the significance of immunosuppression in the UNPC microenvironment as prognostic biomarker of treatment failure in a non-endemic area, and monitored the variation of systemic EBV-specific immunity before and after chemoradiotherapy (CRT). DNA and RNA were extracted from diagnostic biopsies obtained by tumor and adjacent mucosa from 63 consecutive EBV+ UNPC patients who underwent radical CRT. Among these patients 11 relapsed within 2 years. The expression of the EBV-derived UNPC-specific BARF1 gene and several immune-related genes was monitored through quantitative RT-PCR and methylation-specific PCR analyses. Peripheral T cell responses against EBV and BARF1 were measured in 14 patients (7 relapses) through IFN-γ ELISPOT assay. We found significantly higher expression levels of BARF1, CD8, IFN-γ, IDO, PD-L1, and PD-1 in UNPC samples compared to healthy tissues. CD8 expression was significantly reduced in both tumor and healthy tissues in UNPC patients who relapsed within two years. We observed a hypomethylated FOXP3 intron 1 exclusively in relapsed UNPC patients. Finally, we noticed a significant decrease in EBV- and BARF1-specific T-cells after CRT only in relapsing patients. Our data suggest that a high level of immunosuppression (low CD8, hypomethylated FoxP3) in UNPC microenvironment may predict treatment failure and may allow an early identification of patients who could benefit from the addition of immune modulating strategies to improve first line CRT.
Collapse
|
19
|
EBV microRNA-BHRF1-2-5p targets the 3'UTR of immune checkpoint ligands PD-L1 and PD-L2. Blood 2020; 134:2261-2270. [PMID: 31856276 DOI: 10.1182/blood.2019000889] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2019] [Accepted: 10/28/2019] [Indexed: 01/08/2023] Open
Abstract
Epstein-Barr virus-positive (EBV+) diffuse large B-cell lymphomas (DLBCLs) express high levels of programmed death ligand 1 (PD-L1) and PD-L2. MicroRNA (miR) regulation is an important mechanism for the fine-tuning of gene expression via 3'-untranslated region (3'UTR) targeting, and we have previously demonstrated strong EBV miR expression in EBV+ DLBCL. Whereas the EBV latent membrane protein-1 (LMP1) is known to induce PD-L1/L2, a potential counterregulatory role of EBV miR in the fine-tuning of PD-L1/L2 expression remains to be established. To examine this, a novel in vitro model of EBV+ DLBCL was developed, using the viral strain EBV WIL, which unlike common laboratory strains retains intact noncoding regions where several EBV miRs reside. This enabled interrogation of the relationship among EBV latency genes, cell of origin (COO), PD-L1, PD-L2, and EBV miRs. The model successfully recapitulated the full spectrum of B-cell differentiation, with 4 discrete COO phases: early and late germinal center B cells (GCBs) and early and late activated B cells (ABCs). Interestingly, PD-L1/L2 levels increased markedly during transition from late GCB to early ABC phase, after LMP1 upregulation. EBV miR-BamHI fragment H rightward open reading frame 1 (BHRF1)-2-5p clustered apart from other EBV miRs, rising during late GCB phase. Bioinformatic prediction, together with functional validation, confirmed EBV miR-BHRF1-2-5p bound to PD-L1 and PD-L2 3'UTRs to reduce PD-L1/L2 surface protein expression. Results indicate a novel mechanism by which EBV miR-BHRF1-2-5p plays a context-dependent counterregulatory role to fine-tune the expression of the LMP1-driven amplification of these inhibitory checkpoint ligands. Further identification of immune checkpoint-targeting miRs may enable potential novel RNA-based therapies to emerge.
Collapse
|
20
|
Jiang C, Li L, Xiang YQ, Lung ML, Zeng T, Lu J, Tsao SW, Zeng MS, Yun JP, Kwong DLW, Guan XY. Epstein-Barr Virus miRNA BART2-5p Promotes Metastasis of Nasopharyngeal Carcinoma by Suppressing RND3. Cancer Res 2020; 80:1957-1969. [PMID: 32060148 DOI: 10.1158/0008-5472.can-19-0334] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2019] [Revised: 07/08/2019] [Accepted: 02/11/2020] [Indexed: 11/16/2022]
Abstract
Nasopharyngeal carcinoma is an Epstein-Barr virus (EBV)-related malignancy. Recently, we found that the EBV-encoded miRNA BART2-5p was increased in the serum of patients with preclinical nasopharyngeal carcinoma and that the copy number positively correlated with disease progression. In this study, we established its role in nasopharyngeal carcinoma progression and explored underlying mechanisms and clinical significance. BART2-5p was an independent unfavorable prognostic factor for progression-free survival and its circulating abundance positively associated with distant metastasis. Ectopic expression of BART2-5p promoted migration and invasion of EBV-negative nasopharyngeal carcinoma cells, whereas genetic downregulation of BART2-5p in EBV-positive nasopharyngeal carcinoma cells decreased aggressiveness. Mechanistically, BART2-5p targeted RND3, a negative regulator of Rho signaling. Downregulation of RND3 phenocopied the effect of BART2-5p and reconstitution of RND3 rescued the phenotype. By suppressing RND3, BART2-5p activated Rho signaling to enhance cell motility. These findings suggest a novel role for EBV miRNA BART2-5p in promoting nasopharyngeal carcinoma metastasis and its potential value as a prognostic indicator or therapeutic target. SIGNIFICANCE: This study shows that EBV-encoded BART2-5p miRNA suppresses expression of the RND3 Rho family GTPase, consequently promoting ROCK signaling, cell motility, and metastatic behavior of NPC cells.
Collapse
Affiliation(s)
- Chen Jiang
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Sun Yat-sen University Cancer Center, Guangzhou, China.,Department of Pathology, Sun Yat-sen University Cancer Center, Guangzhou, China.,Department of Clinical Oncology, The University of Hong Kong, Hong Kong SAR, China
| | - Lei Li
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Sun Yat-sen University Cancer Center, Guangzhou, China.,Department of Clinical Oncology, The University of Hong Kong, Hong Kong SAR, China
| | - Yan-Qun Xiang
- Department of Nasopharyngeal Carcinoma, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Maria Li Lung
- Department of Clinical Oncology, The University of Hong Kong, Hong Kong SAR, China
| | - Tingting Zeng
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Jiabin Lu
- Department of Pathology, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Sai Wah Tsao
- School of Biomedical Sciences, The University of Hong Kong, Hong Kong SAR, China
| | - Mu-Sheng Zeng
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Jing-Ping Yun
- Department of Pathology, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Dora L W Kwong
- Department of Clinical Oncology, The University of Hong Kong, Hong Kong SAR, China
| | - Xin-Yuan Guan
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Sun Yat-sen University Cancer Center, Guangzhou, China. .,Department of Clinical Oncology, The University of Hong Kong, Hong Kong SAR, China
| |
Collapse
|
21
|
Zhang H, Zou X, Wu L, Zhang S, Wang T, Liu P, Zhu W, Zhu J. Identification of a 7-microRNA signature in plasma as promising biomarker for nasopharyngeal carcinoma detection. Cancer Med 2020; 9:1230-1241. [PMID: 31856390 PMCID: PMC6997065 DOI: 10.1002/cam4.2676] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2019] [Revised: 10/20/2019] [Accepted: 10/21/2019] [Indexed: 01/06/2023] Open
Abstract
BACKGROUND Circulating microRNAs (miRNAs) have become reliable sources of non-invasive biomarkers for cancer diagnosis. Identification of promising miRNA biomarkers in plasma might benefit a lot to the detection of nasopharyngeal carcinoma (NPC). METHODS The Exiqon miRNA qPCR panel was used in the screening stage to identify candidate miRNAs, which were further verified by quantitative reverse transcription polymerase chain reaction (qRT-PCR) in the following three stages among plasma samples from 200 NPC patients and 189 healthy donors (as normal controls [NCs]). The identified miRNAs were further explored in tissue specimens (48 NPC vs 32 NCs) and plasma exosomes (32 NPC vs 32 NCs). Survival analyses were ultimately conducted by Cox regression models and Kaplan-Meier curves using log-rank tests. RESULTS We identified a 7-miRNA signature including let-7b-5p, miR-140-3p, miR-144-3p, miR-17-5p, miR-20a-5p, miR-20b-5p, and miR-205-5p in plasma for NPC diagnosis after four-stage validation. The areas under the receiver operating characteristic curve (AUCs) for the signature were 0.879, 0.884, 0.921, and 0.807 for the training, testing, external validation stage, and the combined three stages, respectively. In NPC tissues, miR-144-3p, miR-17-5p, miR-20a-5p, and miR-205-5p were consistently up-regulated while let-7b-5p and miR-140-3p were significantly down-regulated compared to NCs. However, none of the seven identified miRNAs were dysregulated in plasma-derived exosomes in NPC patients. As to survival analysis, none of the seven miRNAs seemed to be associated with NPC prognosis. CONCLUSION We identified a 7-miRNA signature in plasma as promising non-invasive biomarkers for NPC detection.
Collapse
Affiliation(s)
- Huo Zhang
- Department of OncologyFirst Affiliated Hospital of Nanjing Medical UniversityNanjingJiangsuPR China
- Department of Medical OncologyNorthern Jiangsu People's Hospital Affiliated to Yangzhou UniversityYangzhouJiangsuPR China
| | - Xuan Zou
- Fudan University Shanghai Cancer CenterXuhuiShanghaiPR China
| | - Lirong Wu
- Department of Radiation OncologyNanjing Medical University Affiliated Cancer HospitalJiangsu Cancer HospitalJiangsu Institute of Cancer ResearchNanjingPR China
| | - Shiyu Zhang
- Department of OncologyFirst Affiliated Hospital of Nanjing Medical UniversityNanjingJiangsuPR China
| | - Tongshan Wang
- Department of OncologyFirst Affiliated Hospital of Nanjing Medical UniversityNanjingJiangsuPR China
| | - Ping Liu
- Department of OncologyFirst Affiliated Hospital of Nanjing Medical UniversityNanjingJiangsuPR China
| | - Wei Zhu
- Department of OncologyFirst Affiliated Hospital of Nanjing Medical UniversityNanjingJiangsuPR China
| | - Jun Zhu
- Department of Radiation OncologyNanjing Medical University Affiliated Cancer HospitalJiangsu Cancer HospitalJiangsu Institute of Cancer ResearchNanjingPR China
| |
Collapse
|
22
|
Mishra R, Kumar A, Ingle H, Kumar H. The Interplay Between Viral-Derived miRNAs and Host Immunity During Infection. Front Immunol 2020; 10:3079. [PMID: 32038626 PMCID: PMC6989438 DOI: 10.3389/fimmu.2019.03079] [Citation(s) in RCA: 112] [Impact Index Per Article: 22.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2019] [Accepted: 12/17/2019] [Indexed: 01/01/2023] Open
Abstract
MicroRNAs are short non-coding RNAs that play a crucial role in the regulation of gene expression during cellular processes. The host-encoded miRNAs are known to modulate the antiviral defense during viral infection. In the last decade, multiple DNA and RNA viruses have been shown to produce miRNAs known as viral miRNAs (v-miRNAs) so as to evade the host immune response. In this review, we highlight the origin and biogenesis of viral miRNAs during the viral lifecycle. We also explore the role of viral miRNAs in immune evasion and hence in maintaining chronic infection and disease. Finally, we offer insights into the underexplored role of viral miRNAs as potential targets for developing therapeutics for treating complex viral diseases.
Collapse
Affiliation(s)
- Richa Mishra
- Laboratory of Immunology and Infectious Disease Biology, Department of Biological Sciences, Indian Institute of Science Education and Research, Bhopal, India
| | - Ashish Kumar
- Department of Dermatology, School of Medicine, University of California, Davis, Sacramento, CA, United States
| | - Harshad Ingle
- Department of Medicine, Washington University School of Medicine, Saint Louis, MO, United States
| | - Himanshu Kumar
- Laboratory of Immunology and Infectious Disease Biology, Department of Biological Sciences, Indian Institute of Science Education and Research, Bhopal, India
- Laboratory of Host Defense, WPI Immunology, Frontier Research Centre, Osaka University, Osaka, Japan
| |
Collapse
|
23
|
Sun L, Wang Y, Shi J, Zhu W, Wang X. Association of Plasma Epstein-Barr Virus LMP1 and EBER1 with Circulating Tumor Cells and the Metastasis of Nasopharyngeal Carcinoma. Pathol Oncol Res 2019; 26:1893-1901. [PMID: 31832991 DOI: 10.1007/s12253-019-00777-z] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/02/2019] [Accepted: 11/04/2019] [Indexed: 01/16/2023]
Abstract
Epstein-Barr virus (EBV) has been widely recognized to contribute to the development of nasopharyngeal carcinoma (NPC). The present study was to explore the association of plasma Epstein-Barr Virus LMP1 and EBER1 with circulating tumor cells (CTCs) and the metastasis of nasopharyngeal carcinoma. In the present study, we quantified the plasma levels of EBV DNA/RNAs, such as LMP1, LMP2, BART and EBER1 with real-time quantitative PCR, and CTCs with a CellSpotter Analyzer in NPC patients, with or without metastasis. Then the correlation of each biomarker with other biomarkers and tumor metastasis was analyzed. Our data indicated that the plasma levels of EBV LMP1, BART, EBER1, along with CTCs were significantly higher in metastatic NPC patients than in non-metastatic patients. Plasma LMP1 DNA and EBER1 discriminate metastatic NPC patients from non-metastatic patients, correlate with tumor stage and node stage for metastatic NPC patients. In summary, there were significantly higher plasma levels of Epstein-Barr Virus DNAs / RNAs in nasopharyngeal carcinoma patients. LMP1 DNA and EBER1 RNA correlated with the metastasis of nasopharyngeal carcinoma.
Collapse
Affiliation(s)
- Le Sun
- Department of Otolaryngology, Head & Neck Surgery, First Hospital of Jilin University, 71#, Xinmin Street, Changchun, 130021, People's Republic of China
| | - Yusheng Wang
- Department of Otolaryngology, Head & Neck Surgery, First Hospital of Jilin University, 71#, Xinmin Street, Changchun, 130021, People's Republic of China
| | - Jinfeng Shi
- Department of Otolaryngology, Head & Neck Surgery, First Hospital of Jilin University, 71#, Xinmin Street, Changchun, 130021, People's Republic of China
| | - Wei Zhu
- Department of Otolaryngology, Head & Neck Surgery, First Hospital of Jilin University, 71#, Xinmin Street, Changchun, 130021, People's Republic of China.
| | - Xin Wang
- Department of Otolaryngology, Head & Neck Surgery, First Hospital of Jilin University, 71#, Xinmin Street, Changchun, 130021, People's Republic of China.
| |
Collapse
|
24
|
Limam S, Missaoui N, Mestiri S, Yacoubi M, Krifa H, Selmi B, Mokni M. Epstein-Barr virus infection in gliomas. Curr Res Transl Med 2019; 67:129-133. [DOI: 10.1016/j.retram.2019.06.002] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2019] [Revised: 06/07/2019] [Accepted: 06/30/2019] [Indexed: 12/23/2022]
|
25
|
Liu Y, Jiang Q, Liu X, Lin X, Tang Z, Liu C, Zhou J, Zhao M, Li X, Cheng Z, Li L, Xie Y, Liu Z, Fang W. Cinobufotalin powerfully reversed EBV-miR-BART22-induced cisplatin resistance via stimulating MAP2K4 to antagonize non-muscle myosin heavy chain IIA/glycogen synthase 3β/β-catenin signaling pathway. EBioMedicine 2019; 48:386-404. [PMID: 31594754 PMCID: PMC6838365 DOI: 10.1016/j.ebiom.2019.08.040] [Citation(s) in RCA: 64] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2019] [Revised: 08/16/2019] [Accepted: 08/19/2019] [Indexed: 01/17/2023] Open
Abstract
BACKGROUND Nasopharyngeal carcinoma (NPC) is an Epstein-Barr virus (EBV)-related tumor. The role of EBV-encoding miR-BART22 is still unclear in NPC. This study aimed to identify the detailed mechanisms by which EBV-miR-BART22 functions as a tumor-promoting factor and evaluate the action of cinobufotalin in treating EBV-miR-BART22-overexpressing NPC cells. METHODS Using real-time PCR, western blotting, immunohistochemistry, and In situ hybridization, we detected the expression of miR-BART22 and MAP2K4 in tissues and cells, as well as evaluated their clinical relevance in NPC patients. The effects of miR-BART22 on cell metastasis, stemness and DDP chemoresistance were examined by sphere formation assay, side population analysis, transwell, boyden, in vivo xenograft tumor mouse model et al. Western blotting, immunofluorescence staining, luciferase reporter assay, ChIP, EMSA and Co-IP assay et al. were performed to explore the detailed molecular mechanism of EBV-miR-BART22 in NPC. Finally, we estimated the effects and molecular basis of Cinobufotalin on EBV-miR-BART22-overexpressing NPC cells in vitro and in vivo assays. FINDINGS We observed that EBV-miR-BART22 not only promoted tumor stemness and metastasis, but also enhanced the resistance to Cisplatin (DDP) in vitro and in vivo. Mechanistic analysis indicated that EBV-miR-BART22 directly targeted the MAP2K4 and upregulated non-muscle myosin heavy chain IIA (MYH9) expression by PI3K/AKT/c-Jun-induced transcription. Further, MYH9 interacted with glycogen synthase 3β(GSK3β) protein and induced its ubiquitin degradation by activating PI3K/AKT/c-Jun-induced ubiquitin transcription and the latter combined with increased TRAF6 E3 ligase, which further bound to GSK3β protein. Reductions in the GSK3β protein thus promoted β-catenin expression and nuclear translocation, which induced tumor stemness and the epithelial-to-mesenchymal transition (EMT) signals. Furthermore, we observed that cinobufotalin, a new chemically synthesized compound, significantly suppressed EBV-miR-BART22-induced DDP chemoresistance by upregulating MAP2K4 to suppress MYH9/GSK3β/β-catenin and its downstream tumor stemness and EMT signals in NPC. Finally, clinical data revealed that increased miR-BART22 and reduced MAP2K4 expression caused the poor prognoses of NPC patients. INTERPRETATION Our study provides a novel mechanism that cinobufotalin reversed the DDP chemoresistance and EMT induced by EBV-miR-BART22 in NPC.
Collapse
Affiliation(s)
- Yiyi Liu
- Cancer Center, Integrated Hospital of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China
| | - Qingping Jiang
- Cancer Institute, School of Basic Medical Science, Southern Medical University, Guangzhou, China; Department of Pathology, Third Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Xiong Liu
- Cancer Institute, School of Basic Medical Science, Southern Medical University, Guangzhou, China; Department of Otolaryngology, Head and Neck Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, People's Republic of China
| | - Xian Lin
- Cancer Center, Integrated Hospital of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China
| | - ZiBo Tang
- Cancer Center, Integrated Hospital of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China
| | - Chen Liu
- Cancer Center, Integrated Hospital of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China
| | - Jin Zhou
- Cancer Center, Integrated Hospital of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China
| | - Mengyang Zhao
- Cancer Center, Integrated Hospital of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China
| | - Xin Li
- Shenzhen Key Laboratory of Viral Oncology, the Clinical Innovation & Research Center (CIRC), Shenzhen Hospital, Southern Medical University, Shenzhen, China
| | - Zhao Cheng
- Shenzhen Key Laboratory of Viral Oncology, the Clinical Innovation & Research Center (CIRC), Shenzhen Hospital, Southern Medical University, Shenzhen, China; Department of Pediatric Otorhinolaryngology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Libo Li
- Cancer Center, Integrated Hospital of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China
| | - Yingying Xie
- Cancer Center, Integrated Hospital of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China
| | - Zhen Liu
- Cancer Center, Integrated Hospital of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China; Key Laboratory of Protein Modification and Degradation, School of Basic Medical Sciences, Affiliated Cancer Hospital and Institute of Guangzhou Medical University, Guangzhou, China.
| | - Weiyi Fang
- Cancer Center, Integrated Hospital of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China.
| |
Collapse
|
26
|
Zhang X, Ye Y, Fu M, Zheng B, Qiu Q, Huang Z. Implication of viral microRNAs in the genesis and diagnosis of Epstein-Barr virus-associated tumors. Oncol Lett 2019; 18:3433-3442. [PMID: 31516561 PMCID: PMC6732978 DOI: 10.3892/ol.2019.10713] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2018] [Accepted: 06/26/2019] [Indexed: 12/13/2022] Open
Abstract
The Epstein-Barr virus (EBV) is tightly associated with a variety of human tumors, including Burkitt lymphoma and acquired immune deficiency syndrome-related lymphoma of B-cell origin, as well as nasopharyngeal carcinoma and gastric cancer of epithelial origin. The virus latently infects the host cells and expresses proteins and non-coding RNAs to achieve malignancy. MicroRNAs (miRNAs or miRs) are small RNAs consisting of 19-25 nucleotides, which directly bind to the 3'-untranslated region of mRNAs to promote degradation and inhibit translation of mRNAs. EBV-encoded miRs are generated from two regions of the viral genome, within the apoptosis regulator BHRF1 gene locus and near the BamHI A region in a latency type-dependent manner. In addition, EBV-encoded miRs epigenetically regulate the expression of molecules that are effectors of the cell cycle progression, migration, apoptosis and innate immunity, serving a vital role in supporting viral replication and occurrence of EBV-associated tumors. The feasibility of using such miRs as biomarkers for the diagnosis and prognosis of EBV-associated tumors is currently under investigation.
Collapse
Affiliation(s)
- Xiangning Zhang
- Department of Pathophysiology, Chinese-American Collaborative Cancer Institute, Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, Guangdong Medical University, Dongguan, Guangdong 523808, P.R. China
| | - Yudong Ye
- Department of Otolaryngology, Dongguan City People's Hospital, Dongguan, Guangdong 523059, P.R. China
| | - Ming Fu
- Department of Otolaryngology, Dongguan City People's Hospital, Dongguan, Guangdong 523059, P.R. China
| | - Biying Zheng
- Department of Clinical Microbiology, College of Laboratory Medicine, Guangdong Medical University, Dongguan, Guangdong 523808, P.R. China
| | - Qianhui Qiu
- Department of Otolaryngology, Pearl River Hospital, Southern Medical University, Guangzhou, Guangdong 510280, P.R. China
| | - Zunnan Huang
- Department of Pathophysiology, Chinese-American Collaborative Cancer Institute, Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, Guangdong Medical University, Dongguan, Guangdong 523808, P.R. China
| |
Collapse
|
27
|
Spontaneous lymphoblastoid cell lines from patients with Epstein-Barr virus infection show highly variable proliferation characteristics that correlate with the expression levels of viral microRNAs. PLoS One 2019; 14:e0222847. [PMID: 31568538 PMCID: PMC6768455 DOI: 10.1371/journal.pone.0222847] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2019] [Accepted: 09/07/2019] [Indexed: 12/31/2022] Open
Abstract
The Epstein-Barr virus (EBV) induces B-cell proliferation with high efficiency through expression of latent proteins and microRNAs. This process takes place in vivo soon after infection, presumably to expand the virus reservoir, but can also induce pathologies, e.g. an infectious mononucleosis (IM) syndrome after primary infection or a B-cell lymphoproliferation in immunosuppressed individuals. In this paper, we investigated the growth characteristics of EBV-infected B-cells isolated from transplant recipients or patients with IM. We found that these cells grew and withstood apoptosis at highly variable rates, suggesting that the expansion rate of the infected B-cells widely varies between individuals, thereby influencing the size of the B-cell reservoir and the ability to form tumors in infected individuals. All viruses investigated were type 1 and genetically close to western strains. EBV-infected B-cells expressed the transforming EBV latent genes and microRNAs (miRNAs) at variable levels. We found that the B-cell growth rates positively correlated with the BHRF1 miRNA levels. Comparative studies showed that infected B-cells derived from transplant recipients with iEBVL on average expressed higher levels of EBV miR-BHRF1 miRNAs and grew more rapidly than B-cells from IM patients, suggesting infection by more transforming viruses. Altogether, these findings suggest that EBV infection has a highly variable impact on the B-cell compartment that probably reflects the genetic diversity of both the virus and the host. It also demonstrates the unexpected finding that B-cells from different individuals can grow at different speed under the influence of the same virus infection.
Collapse
|
28
|
Tsang CM, Lui VWY, Bruce JP, Pugh TJ, Lo KW. Translational genomics of nasopharyngeal cancer. Semin Cancer Biol 2019; 61:84-100. [PMID: 31521748 DOI: 10.1016/j.semcancer.2019.09.006] [Citation(s) in RCA: 84] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2019] [Revised: 09/11/2019] [Accepted: 09/11/2019] [Indexed: 12/26/2022]
Abstract
Nasopharyngeal carcinoma (NPC), also named the Cantonese cancer, is a unique cancer with strong etiological association with infection of the Epstein-Barr virus (EBV). With particularly high prevalence in Southeast Asia, the involvement of EBV and genetic aberrations contributive to NPC tumorigenesis have remained unclear for decades. Recently, genomic analysis of NPC has defined it as a genetically homogeneous cancer, driven largely by NF-κB signaling caused by either somatic aberrations of NF-κB negative regulators or by overexpression of the latent membrane protein 1 (LMP1), an EBV viral oncoprotein. This represents a landmark finding of the NPC genome. Exome and RNA sequencing data from new EBV-positive NPC models also highlight the importance of PI3K pathway aberrations in NPC. We also realize for the first time that NPC mutational burden, mutational signatures, MAPK/PI3K aberrations, and MHC Class I gene aberrations, are prognostic for patient outcome. Together, these multiple genomic discoveries begin to shape the focus of NPC therapy development. Given the challenge of NF-κB targeting in human cancers, more innovative drug discovery approaches should be explored to target the unique atypical NF-κB activation feature of NPC. Our next decade of NPC research should focus on further identification of the -omic landscapes of recurrent and metastatic NPC, development of gene-based precision medicines, as well as large-scale drug screening with the newly developed and well-characterized EBV-positive NPC models. Focused preclinical and clinical investigations on these major directions may identify new and effective targeting strategies to further improve survival of NPC patients.
Collapse
Affiliation(s)
- Chi Man Tsang
- Department of Anatomical and cellular Pathology and State Key Laboratory of Translational Oncology, The Chinese University of Hong Kong, Hong Kong Special Administrative Region
| | - Vivian Wai Yan Lui
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong Special Administrative Region
| | - Jeffrey P Bruce
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, M5G 1L7, Canada
| | - Trevor J Pugh
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, M5G 1L7, Canada; Department of Medical Biophysics, University of Toronto, Toronto, ON, M5G 1L7, Canada; Ontario Institute for Cancer Research, Toronto, ON, M5G 1L7, Canada
| | - Kwok Wai Lo
- Department of Anatomical and cellular Pathology and State Key Laboratory of Translational Oncology, The Chinese University of Hong Kong, Hong Kong Special Administrative Region.
| |
Collapse
|
29
|
Shannon-Lowe C, Rickinson A. The Global Landscape of EBV-Associated Tumors. Front Oncol 2019; 9:713. [PMID: 31448229 PMCID: PMC6691157 DOI: 10.3389/fonc.2019.00713] [Citation(s) in RCA: 288] [Impact Index Per Article: 48.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2019] [Accepted: 07/18/2019] [Indexed: 12/12/2022] Open
Abstract
Epstein-Barr virus (EBV), a gamma-1 herpesvirus, is carried as a life-long asymptomatic infection by the great majority of individuals in all human populations. Yet this seemingly innocent virus is aetiologically linked to two pre-malignant lymphoproliferative diseases (LPDs) and up to nine distinct human tumors; collectively these have a huge global impact, being responsible for some 200,000 new cases of cancer arising worldwide each year. EBV replicates in oral epithelium but persists as a latent infection within the B cell system and several of its diseases are indeed of B cell origin; these include B-LPD of the immunocompromised, Hodgkin Lymphoma (HL), Burkitt Lymphoma (BL), Diffuse Large B cell Lymphoma (DLBCL) and two rarer tumors associated with profound immune impairment, plasmablastic lymphoma (PBL) and primary effusion lymphoma (PEL). Surprisingly, the virus is also linked to tumors arising in other cellular niches which, rather than being essential reservoirs of virus persistence in vivo, appear to represent rare cul-de-sacs of latent infection. These non-B cell tumors include LPDs and malignant lymphomas of T or NK cells, nasopharyngeal carcinoma (NPC) and gastric carcinoma of epithelial origin, and leiomyosarcoma, a rare smooth muscle cell tumor of the immunocompromised. Here we describe the main characteristics of these tumors, their distinct epidemiologies, histological features and degrees of EBV association, then consider how their different patterns of EBV latency may reflect the alternative latency programmes through which the virus first colonizes and then persists in immunocompetent host. For each tumor, we discuss current understanding of EBV's role in the oncogenic process, the identity (where known) of host genetic and environmental factors predisposing tumor development, and the recent evidence from cancer genomics identifying somatic changes that either complement or in some cases replace the contribution of the virus. Thereafter we look for possible connections between the pathogenesis of these apparently different malignancies and point to new research areas where insights may be gained.
Collapse
Affiliation(s)
- Claire Shannon-Lowe
- Institute for Immunology and Immunotherapy, The University of Birmingham, Birmingham, United Kingdom
| | - Alan Rickinson
- Institute for Immunology and Immunotherapy, The University of Birmingham, Birmingham, United Kingdom
| |
Collapse
|
30
|
Wang M, Gu B, Chen X, Wang Y, Li P, Wang K. The Function and Therapeutic Potential of Epstein-Barr Virus-Encoded MicroRNAs in Cancer. MOLECULAR THERAPY. NUCLEIC ACIDS 2019; 17:657-668. [PMID: 31400608 PMCID: PMC6698931 DOI: 10.1016/j.omtn.2019.07.002] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/27/2018] [Revised: 05/14/2019] [Accepted: 07/06/2019] [Indexed: 02/06/2023]
Abstract
Epstein-Barr virus (EBV) is a ubiquitous human γ-herpesvirus that infects over 90% of the global population. EBV is considered a contributory factor in a variety of malignancies including nasopharyngeal carcinoma, gastric carcinoma, Burkitt lymphoma, and Hodgkin’s lymphoma. Notably, EBV was the first virus found to encode microRNAs (miRNAs). Increasing evidence indicates that EBV-encoded miRNAs contribute to the carcinogenesis and development of EBV-associated malignancies. EBV miRNAs have been shown to inhibit the expression of genes involved in cell proliferation, apoptosis, invasion, and immune signaling pathways. Therefore, EBV miRNAs perform a significant function in the complex host-virus interaction and EBV-driven carcinogenesis. However, the integrated mechanisms underlying the roles of EBV miRNAs in carcinogenesis remain to be further explored. In this review, we describe recent advances regarding the involvement of EBV miRNAs in the pathogenesis of EBV-associated malignancies and discuss their potential utility as cancer biomarkers. An in-depth investigation into the pro-carcinogenic role of EBV miRNAs will expand our knowledge of the biological processes associated with virus-driven tumors and contribute to the development of novel therapeutic strategies for the treatment of EBV-associated malignancies.
Collapse
Affiliation(s)
- Man Wang
- Institute for Translational Medicine, Medical College of Qingdao University, Dengzhou Road 38, Qingdao 266021, China.
| | - Bianli Gu
- Henan Key Laboratory of Cancer Epigenetics, Cancer Institute, The First Affiliated Hospital and College of Clinical Medicine of Henan University of Science and Technology, Luoyang 471003, China
| | - Xinzhe Chen
- Institute for Translational Medicine, Medical College of Qingdao University, Dengzhou Road 38, Qingdao 266021, China
| | - Yefu Wang
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan 430072, China
| | - Peifeng Li
- Institute for Translational Medicine, Medical College of Qingdao University, Dengzhou Road 38, Qingdao 266021, China
| | - Kun Wang
- Institute for Translational Medicine, Medical College of Qingdao University, Dengzhou Road 38, Qingdao 266021, China.
| |
Collapse
|
31
|
Hussein HAM, Alfhili MA, Pakala P, Simon S, Hussain J, McCubrey JA, Akula SM. miRNAs and their roles in KSHV pathogenesis. Virus Res 2019; 266:15-24. [PMID: 30951791 DOI: 10.1016/j.virusres.2019.03.024] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2019] [Revised: 03/26/2019] [Accepted: 03/26/2019] [Indexed: 12/12/2022]
Abstract
Kaposi's sarcoma-associated herpesvirus (KSHV) is the etiological agent of Kaposi's sarcoma (KS), primary effusion lymphoma (PEL), and multicentric Castleman Disease (MCD). Recent mechanistic advances have discerned the importance of microRNAs in the virus-host relationship. KSHV has two modes of replication: lytic and latent phase. KSHV entry into permissive cells, establishment of infection, and maintenance of latency are contingent upon successful modulation of the host miRNA transcriptome. Apart from host cell miRNAs, KSHV also encodes viral miRNAs. Among various cellular and molecular targets, miRNAs are appearing to be key players in regulating viral pathogenesis. Therefore, the use of miRNAs as novel therapeutics has gained considerable attention as of late. This innovative approach relies on either mimicking miRNA species by identical oligonucleotides, or selective silencing of miRNA with specific oligonucleotide inhibitors. Here, we provide an overview of KSHV pathogenesis at the molecular level with special emphasis on the various roles miRNAs play during virus infection.
Collapse
Affiliation(s)
- Hosni A M Hussein
- Department of Microbiology & Immunology, Brody School of Medicine, East Carolina University, Greenville, NC 27834, United States; Faculty of Science, Al Azhar University, Assiut Branch, Assiut 71524, Egypt
| | - Mohammad A Alfhili
- Department of Medicine (Division of Hematology/Oncology), Brody School of Medicine, East Carolina University, Greenville, NC 27834, United States; Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, King Saud University, Riyadh 11433, Saudi Arabia
| | - Pranaya Pakala
- Department of Microbiology & Immunology, Brody School of Medicine, East Carolina University, Greenville, NC 27834, United States
| | - Sandra Simon
- Department of Microbiology & Immunology, Brody School of Medicine, East Carolina University, Greenville, NC 27834, United States
| | - Jaffer Hussain
- Department of Internal Medicine, Brody School of Medicine, East Carolina University, Greenville, NC 27834, United States
| | - James A McCubrey
- Department of Microbiology & Immunology, Brody School of Medicine, East Carolina University, Greenville, NC 27834, United States
| | - Shaw M Akula
- Department of Microbiology & Immunology, Brody School of Medicine, East Carolina University, Greenville, NC 27834, United States.
| |
Collapse
|
32
|
Hartung A, Makarewicz O, Egerer R, Karrasch M, Klink A, Sauerbrei A, Kentouche K, Pletz MW. EBV miRNA expression profiles in different infection stages: A prospective cohort study. PLoS One 2019; 14:e0212027. [PMID: 30759142 PMCID: PMC6373943 DOI: 10.1371/journal.pone.0212027] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2018] [Accepted: 01/25/2019] [Indexed: 01/15/2023] Open
Abstract
The Epstein-Barr virus (EBV) produces different microRNAs (miRNA) with distinct regulatory functions within the infectious cycle. These viral miRNAs regulate the expression of viral and host genes and have been discussed as potential diagnostic markers or even therapeutic targets, provided that the expression profile can be unambiguously correlated to a specific stage of infection or a specific EBV-induced disorder. In this context, miRNA profiling becomes more important since the roles of these miRNAs in the pathogenesis of infections and malignancies are not fully understood. Studies of EBV miRNA expression profiles are sparse and have mainly focused on associated malignancies. This study is the first to examine the miRNA profiles of EBV reactivation and to use a correction step with seronegative patients as a reference. Between 2012 and 2017, we examined the expression profiles of 11 selected EBV miRNAs in 129 whole blood samples from primary infection, reactivation, healthy carriers and EBV seronegative patients. Three of the miRNAs could not be detected in any sample. Other miRNAs showed significantly higher expression levels and prevalence during primary infection than in other stages; miR-BHRF1-1 was the most abundant. The expression profiles from reactivation differed slightly but not significantly from those of healthy carriers, but a specific marker miRNA for each stage could not be identified within the selected EBV miRNA targets.
Collapse
Affiliation(s)
- Anita Hartung
- Institute of Infectious Diseases and Infection Control, Jena University Hospital, Jena, Germany
- * E-mail:
| | - Oliwia Makarewicz
- Institute of Infectious Diseases and Infection Control, Jena University Hospital, Jena, Germany
| | - Renate Egerer
- Institute of Medical Microbiology, Jena University Hospital, Jena, Germany
| | - Matthias Karrasch
- Institute of Medical Microbiology, Jena University Hospital, Jena, Germany
| | - Anne Klink
- Department of Haematology and Medical Oncology, Jena University Hospital, Jena, Germany
| | - Andreas Sauerbrei
- Institute of Virology and Antiviral Therapy, Jena University Hospital, Jena, Germany
| | - Karim Kentouche
- Clinic for Children and Youth Medicine, Jena University Hospital, Jena, Germany
| | - Mathias W. Pletz
- Institute of Infectious Diseases and Infection Control, Jena University Hospital, Jena, Germany
| |
Collapse
|
33
|
Dong M, Chen JN, Huang JT, Gong LP, Shao CK. The roles of EBV-encoded microRNAs in EBV-associated tumors. Crit Rev Oncol Hematol 2019; 135:30-38. [PMID: 30819444 DOI: 10.1016/j.critrevonc.2019.01.014] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2018] [Revised: 01/22/2019] [Accepted: 01/22/2019] [Indexed: 12/19/2022] Open
Abstract
Epstein-Barr virus (EBV) is believed to be a pathogen causing a number of human cancers, but the pathogenic mechanisms remain unclear. An increasing number of studies have indicated that EBV-encoded microRNAs (EBV miRNAs) are expressed in a latency type- and tumor type-dependent manner, playing important roles in the development and progression of EBV-associated tumors. By targeting one or more genes of the virus and the host, EBV miRNAs are responsible for the deregulation of a variety of viral and host cell biological processes, including viral replication, latency maintenance, immune evasion, cell apoptosis and metabolism, and tumor proliferation and metastasis. In addition, some EBV miRNAs can be used as excellent diagnostic, prognostic and treatment efficacy predictive biomarkers for EBV-associated tumors. More importantly, EBV miRNA-targeting therapeutics have emerged and have been developing rapidly, which may open a new era in the treatment of EBV-associated tumors in the near future.
Collapse
Affiliation(s)
- Min Dong
- Department of Medical Oncology, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510630, China
| | - Jian-Ning Chen
- Department of Pathology, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510630, China
| | - Jun-Ting Huang
- Department of Pathology, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510630, China
| | - Li-Ping Gong
- Department of Pathology, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510630, China
| | - Chun-Kui Shao
- Department of Pathology, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510630, China.
| |
Collapse
|
34
|
Epstein-Barr virus-coded miR-BART13 promotes nasopharyngeal carcinoma cell growth and metastasis via targeting of the NKIRAS2/NF-κB pathway. Cancer Lett 2019; 447:33-40. [PMID: 30684592 DOI: 10.1016/j.canlet.2019.01.022] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2018] [Revised: 01/05/2019] [Accepted: 01/17/2019] [Indexed: 12/27/2022]
Abstract
Based on analysis of Epstein-Barr virus (EBV) BART microRNA expression profiles, we previously reported that EBV-encoded miR-BART13 is upregulated in nasopharyngeal carcinoma (NPC) plasma specimens. However, the effects and molecular mechanisms of miR-BART13 in NPC remain largely unknown. We found that miR-BART13 was significantly upregulated in NPC tissue specimens. Ectopic expression of miR-BART13 promoted NPC cell proliferation, epithelial mesenchymal transition, and metastasis in vitro, and facilitated xenograft tumor growth and lung metastasis in vivo. Molecularly, NF-κB inhibitor interacting Ras-like 2 (NKIRAS2), a negative regulator of the NF-κB signaling, was identified to be a direct target of miR-BART13 in NPC cells, and NKIRAS2 mRNA and protein expression was inversely correlated with miR-BART13 in NPC tissues, respecitvely. Furthermore, the NF-κB signaling pathway was activated by miR-BART13. By rescued experiments, reconstitution of NKIRAS2 expression abrogated all the phenotypes upregulated by miR-BART13, and attenuated activity of NF-κB signaling pathway activated by miR-BART13 in NPC cells. Our findings indicated the newly identified miR-BART13/NKIRAS2/NF-κB signaling axis may provide further insights into better understanding of NPC initiation and development, and targeting of this pathway could be further studied as a therapeutic strategy for NPC patients.
Collapse
|
35
|
Chen H, Ji M, Zong JF, Ko JMY, Dai W, Lung ML. Conventional and Novel Diagnostic Biomarkers and Approaches for Detection of Nasopharyngeal Carcinoma. NASOPHARYNGEAL CARCINOMA 2019:129-153. [DOI: 10.1016/b978-0-12-814936-2.00007-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
36
|
Gao W, Wong TS, Lv KX, Zhang MJ, Tsang RKY, Chan JYW. Detection of Epstein-Barr virus (EBV)-encoded microRNAs in plasma of patients with nasopharyngeal carcinoma. Head Neck 2018; 41:780-792. [PMID: 30548946 DOI: 10.1002/hed.25544] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2017] [Revised: 08/03/2018] [Accepted: 09/28/2018] [Indexed: 02/02/2023] Open
Abstract
BACKGROUND Nasopharyngeal carcinoma (NPC) latently infected by Epstein-Barr virus (EBV) expresses 40 EBV BART microRNAs (miRNAs). Difference in diagnostic efficacy of these miRNAs on NPC detection was observed. Here, we performed a comprehensive evaluation on the efficacy of these miRNAs. METHODS Quantitative polymerase chain reaction was performed on plasma nucleic acid isolated from patients with NPC and noncancer donors. RESULTS For primary NPC, BART2-5P, BART6-3P, BART7-3P, BART7-5P, BART9-5P, BART11-3P, BART17-5P, and BART19-5P were significantly elevated. For recurrent NPC, plasma levels of BART2-3P, BART2-5P, BART5-3P, BART5-5P, BART6-3P, BART8-3P, BART9-5P, BART17-5P, BART19-3P, and BART20-3P were significantly increased. Area under curve (AUC) analysis showed that BART19-5P had the best performance to identify NPC which was serologically EBV DNA undetectable. For recurrent NPC, BART8-3P and BART10-3P had highest AUC value for identifying cancer in EBV DNA undetectable plasma. CONCLUSION Our data supported the use of circulating EBV miRNAs in NPC and recurrent NPC detection.
Collapse
Affiliation(s)
- Wei Gao
- Division of Head and Neck, Department of Surgery, The University of Hong Kong, Hong Kong SAR, China.,Shenzhen Institute of Research and Innovation, The University of Hong Kong, China
| | - Thian-Sze Wong
- Division of Head and Neck, Department of Surgery, The University of Hong Kong, Hong Kong SAR, China.,Shenzhen Institute of Research and Innovation, The University of Hong Kong, China
| | - Ke-Xing Lv
- Division of Head and Neck, Department of Surgery, The University of Hong Kong, Hong Kong SAR, China
| | - Min-Juan Zhang
- Division of Head and Neck, Department of Surgery, The University of Hong Kong, Hong Kong SAR, China.,Shenzhen Institute of Research and Innovation, The University of Hong Kong, China
| | - Raymond King-Yin Tsang
- Division of Head and Neck, Department of Surgery, The University of Hong Kong, Hong Kong SAR, China.,Shenzhen Institute of Research and Innovation, The University of Hong Kong, China
| | - Jimmy Yu-Wai Chan
- Division of Head and Neck, Department of Surgery, The University of Hong Kong, Hong Kong SAR, China.,Shenzhen Institute of Research and Innovation, The University of Hong Kong, China
| |
Collapse
|
37
|
Ramayanti O, Verkuijlen SAWM, Novianti P, Scheepbouwer C, Misovic B, Koppers-Lalic D, van Weering J, Beckers L, Adham M, Martorelli D, Middeldorp JM, Pegtel DM. Vesicle-bound EBV-BART13-3p miRNA in circulation distinguishes nasopharyngeal from other head and neck cancer and asymptomatic EBV-infections. Int J Cancer 2018; 144:2555-2566. [PMID: 30411781 PMCID: PMC6587801 DOI: 10.1002/ijc.31967] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2018] [Revised: 10/04/2018] [Accepted: 10/17/2018] [Indexed: 01/05/2023]
Abstract
Cell‐free microRNA (miRNA) in biofluids released by tumors in either protein or vesicle‐bound form, represent promising minimally‐invasive cancer biomarkers. However, a highly abundant non‐tumor background in human plasma and serum complicates the discovery and detection of tumor‐selective circulating miRNAs. We performed small RNA sequencing on serum and plasma RNA from Nasopharyngeal Carcinoma (NPC) patients. Collectively, Epstein Barr virus‐encoded miRNAs, more so than endogenous miRNAs, signify presence of NPC. However, RNAseq‐based EBV miRNA profiles differ between NPC patients, suggesting inter‐tumor heterogeneity or divergent secretory characteristics. We determined with sensitive qRT‐PCR assays that EBV miRNAs BART7‐3p, BART9‐3p and BART13‐3p are actively secreted by C666.1 NPC cells bound to extracellular vesicles (EVs) and soluble ribonucleoprotein complexes. Importantly, these miRNAs are expressed in all primary NPC tumor biopsies and readily detected in nasopharyngeal brushings from both early and late‐stage NPC patients. Increased levels of BART7‐3p, BART9‐3p and particularly BART13‐3p, distinguish NPC patient sera from healthy controls. Receiver operating characteristic curve analysis using sera from endemic NPC patients, other head and neck cancers and individuals with asymptomatic EBV‐infections reveals a superior diagnostic performance of EBV miRNAs over anti‐EBNA1 IgA serology and EBV‐DNA load (AUC 0.87–0.96 vs 0.86 and 0.66 respectively). The high specificity of circulating EBV‐BART13‐3p (97%) for NPC detection is in agreement with active secretion from NPC tumor cells. We conclude EV‐bound BART13‐3p in circulation is a promising, NPC‐selective, biomarker that should be considered as part of a screening strategy to identify NPC in endemic regions. What's new? Analysis of DNA from human tumor viruses in patient blood is a non‐invasive screening method for individuals at risk for developing cancer. A drawback is over‐diagnosis as these sensitive methods also detect non‐cancer‐related infections. Here the authors show by RNA sequencing and PCR amplification that a microRNA (BART13‐3p) encoded by the Epstein–Barr Virus (EBV) is associated with circulating vesicles in patients with nasopharyngeal carcinoma, thus distinguishing between cancer and non‐cancer‐related EBV infections.
Collapse
Affiliation(s)
- Octavia Ramayanti
- Amsterdam UMC, Vrije Universiteit Amsterdam, Department of Pathology, Cancer Center Amsterdam, Amsterdam, The Netherlands
| | - Sandra A W M Verkuijlen
- Amsterdam UMC, Vrije Universiteit Amsterdam, Department of Pathology, Cancer Center Amsterdam, Amsterdam, The Netherlands
| | - Putri Novianti
- Amsterdam UMC, Vrije Universiteit Amsterdam, Department of Pathology, Cancer Center Amsterdam, Amsterdam, The Netherlands.,Amsterdam UMC, Department of Epidemiology and Biostatistics, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Chantal Scheepbouwer
- Amsterdam UMC, Department of Neurosurgery, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Branislav Misovic
- Amsterdam UMC, Department of Neurosurgery, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Danijela Koppers-Lalic
- Amsterdam UMC, Department of Neurosurgery, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Jan van Weering
- Amsterdam UMC, Clinical Genetics, Center for Neurogenomics and Cognitive Research, Neuroscience Campus Amsterdam, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - Lisa Beckers
- Amsterdam UMC, Vrije Universiteit Amsterdam, Department of Pathology, Cancer Center Amsterdam, Amsterdam, The Netherlands
| | - Marlinda Adham
- Department of Ear, Nose and Throat Surgery, dr. Cipto Mangunkusumo Hospital, University of Indonesia, Jakarta, Indonesia
| | - Debora Martorelli
- National Cancer Institute, Centro di Riferimento Oncologico, Aviano, Italy
| | - Jaap M Middeldorp
- Amsterdam UMC, Vrije Universiteit Amsterdam, Department of Pathology, Cancer Center Amsterdam, Amsterdam, The Netherlands
| | - Dirk Michiel Pegtel
- Amsterdam UMC, Vrije Universiteit Amsterdam, Department of Pathology, Cancer Center Amsterdam, Amsterdam, The Netherlands
| |
Collapse
|
38
|
Zheng X, Wang J, Wei L, Peng Q, Gao Y, Fu Y, Lu Y, Qin Z, Zhang X, Lu J, Ou C, Li Z, Zhang X, Liu P, Xiong W, Li G, Yan Q, Ma J. Epstein-Barr Virus MicroRNA miR-BART5-3p Inhibits p53 Expression. J Virol 2018; 92:e01022-18. [PMID: 30209170 PMCID: PMC6232473 DOI: 10.1128/jvi.01022-18] [Citation(s) in RCA: 75] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2018] [Accepted: 09/05/2018] [Indexed: 12/26/2022] Open
Abstract
Epstein-Barr virus (EBV) is the first human virus found to encode many microRNAs. It is etiologically linked to nasopharyngeal carcinoma and EBV-associated gastric carcinoma. During the latent infection period, there are only a few EBV proteins expressed, whereas EBV microRNAs, such as the BamHI-A region rightward transcript (BART) microRNAs, are highly expressed. However, how these BART miRNAs precisely regulate the tumor growth in nasopharyngeal carcinoma and gastric carcinoma remains obscure. Here, we report that upregulation of EBV-miR-BART5-3p promotes the growth of nasopharyngeal carcinoma and gastric carcinoma cells. BART5-3p directly targets the tumor suppressor gene TP53 on its 3'-untranslated region (3'-UTR) and consequently downregulates CDKN1A, BAX, and FAS expression, leading to acceleration of the cell cycle progress and inhibition of cell apoptosis. BART5-3p contributes to the resistance to chemotherapeutic drugs and ionizing irradiation-induced p53 increase. Moreover, BART5-3p also facilitates degradation of p53 proteins. BART5-3p is the first EBV-microRNA to be identified as inhibiting p53 expression and function, which suggests a novel mechanism underlying the strategies employed by EBV to maintain latent infection and promote the development of EBV-associated carcinomas.IMPORTANCE EBV encodes 44 mature microRNAs, which have been proven to promote EBV-associated diseases by targeting host genes and self-viral genes. In EBV-associated carcinomas, the expression of viral protein is limited but the expression of BART microRNAs is extremely high, suggesting that they could be major factors in the contribution of EBV-associated tumorigenesis. p53 is a critical tumor suppressor. Unlike in most human solid tumors, TP53 mutations are rare in nasopharyngeal carcinoma and EBV-associated gastric carcinoma tissues, suggesting a possibility that some EBV-encoded products suppress the functions of p53. This study provides the first evidence that a BART microRNA can suppress p53 expression by directly targeting its 3'-UTR. This study implies that EBV can use its BART microRNAs to modulate the expression of p53, thus maintaining its latency and contributing to tumorigenesis.
Collapse
Affiliation(s)
- Xiang Zheng
- Xiangya Hospital, Cancer Research Institute, Central South University, Changsha, China
- Clinical Anatomy & Reproductive Medicine Application Institute, Department of Histology and Embryology, School of Medicine, University of South China, Hengyang, China
- Hunan Key Laboratory of Translational Radiation Oncology, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China
- Hunan Key Laboratory of Nonresolving Inflammation and Cancer, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Jia Wang
- Xiangya Hospital, Cancer Research Institute, Central South University, Changsha, China
| | - Lingyu Wei
- Xiangya Hospital, Cancer Research Institute, Central South University, Changsha, China
- Hunan Key Laboratory of Nonresolving Inflammation and Cancer, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Qiu Peng
- Xiangya Hospital, Cancer Research Institute, Central South University, Changsha, China
| | - Yingxue Gao
- Xiangya Hospital, Cancer Research Institute, Central South University, Changsha, China
| | - Yuxin Fu
- Xiangya Hospital, Cancer Research Institute, Central South University, Changsha, China
| | - Yuanjun Lu
- Xiangya Hospital, Cancer Research Institute, Central South University, Changsha, China
| | - Zailong Qin
- Xiangya Hospital, Cancer Research Institute, Central South University, Changsha, China
| | - Xuemei Zhang
- Xiangya Hospital, Cancer Research Institute, Central South University, Changsha, China
| | - Jianhong Lu
- Xiangya Hospital, Cancer Research Institute, Central South University, Changsha, China
| | - Chunlin Ou
- Xiangya Hospital, Cancer Research Institute, Central South University, Changsha, China
| | - Zhengshuo Li
- Xiangya Hospital, Cancer Research Institute, Central South University, Changsha, China
| | - Xiaoyue Zhang
- Xiangya Hospital, Cancer Research Institute, Central South University, Changsha, China
| | - Peishan Liu
- Xiangya Hospital, Cancer Research Institute, Central South University, Changsha, China
| | - Wei Xiong
- Xiangya Hospital, Cancer Research Institute, Central South University, Changsha, China
- Hunan Key Laboratory of Translational Radiation Oncology, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China
- Hunan Key Laboratory of Nonresolving Inflammation and Cancer, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Guiyuan Li
- Xiangya Hospital, Cancer Research Institute, Central South University, Changsha, China
| | - Qun Yan
- Xiangya Hospital, Cancer Research Institute, Central South University, Changsha, China
| | - Jian Ma
- Xiangya Hospital, Cancer Research Institute, Central South University, Changsha, China
- Hunan Key Laboratory of Translational Radiation Oncology, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China
- Hunan Key Laboratory of Nonresolving Inflammation and Cancer, The Third Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
39
|
Lin C, Zong J, Lin W, Wang M, Xu Y, Zhou R, Lin S, Guo Q, Chen H, Ye Y, Zhang B, Pan J. EBV-miR-BART8-3p induces epithelial-mesenchymal transition and promotes metastasis of nasopharyngeal carcinoma cells through activating NF-κB and Erk1/2 pathways. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2018; 37:283. [PMID: 30477559 PMCID: PMC6257964 DOI: 10.1186/s13046-018-0953-6] [Citation(s) in RCA: 69] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/26/2018] [Accepted: 11/07/2018] [Indexed: 12/22/2022]
Abstract
Background Epstein-Barr virus (EBV) is ubiquitously associated with nasopharyngeal carcinoma (NPC). EBV encodes two groups of microRNAs (miRNAs) which are divided into BamHI fragment H rightward open reading frame 1 (BHRF1) and BamHI-A rightward transcripts (BART) microRNAs. EBV miR-BART has been found to be involved in the development and progression of NPC. However, so far the role of EBV-miR-BART8-3p in NPC progression remains unknown. This study aimed to investigate the role of EBV-miR-BART8-3p in NPC and explore the underlying mechanisms. Methods miRNA expression was profiled in NPC and normal nasopharyngeal mucosal specimens using miRNA sequencing. EBV-miR-BART8-3p and RNF38 expression was quantified with qPCR assay. The migration, invasion and metastasis of NPC cells were evaluated using CCK-8, colony-forming, wound-healing, and migration and invasion assays. The expression levels of epithelial-mesenchymal transition (EMT)-related markers,metastasis-related markers and NF-κB and Erk1/2 signaling proteins were determined using Western blotting. Tumorigenic assay was performed to evaluate the pulmonary metastatic ability of NPC cells in vivo. Results EBV BART miRNAs were highly over-expressed and co-expressed in NPC and might be associated with deactivated immune response in NPC according to the sequencing analysis. EBV-miR-BART8-3p expression was significantly higher in human NPC specimens than in normal nasopharyngeal mucosal specimens. EBV-miR-BART8-3p was found to promote NPC migration, invasion and metastasis, drove an EMT process and upregulated expression of metastasis-related proteins expression in NPC cells. Our data showed EBV-miR-BART8-3p directly targeted RNF38 in NPC cells. Conclusion The present study demonstrates that EBV-miR-BART8-3p plays a significant role in inducing EMT and promoting metastasis through directly targeting RNF38 in NPC cells via the activation of NF-κB and Erk1/2 signaling pathways. Our findings suggest that EBV-miR-BART8-3p is a potential therapeutic target for NPC. Electronic supplementary material The online version of this article (10.1186/s13046-018-0953-6) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Cheng Lin
- Fujian Medical University, Fuzhou, 350108, Fujian Province, China.,Department of Radiation Oncology, Fujian Cancer Hospital & Fujian Medical University Cancer Hospital, Fuzhou, 350011, Fujian Province, China
| | - Jingfeng Zong
- Department of Radiation Oncology, Fujian Cancer Hospital & Fujian Medical University Cancer Hospital, Fuzhou, 350011, Fujian Province, China
| | - Wansong Lin
- Laboratory of Immuno-Oncology, Fujian Cancer Hospital & Fujian Medical University Cancer Hospital, Fuzhou, 350011, Fujian Province, China
| | - Minghui Wang
- Department of Genetics and Genomic Sciences, Icahn Institute of Genomics and Multiscale Biology, Mount Sinai Center for Transformative Disease Modeling, Icahn School of Medicine at Mount Sinai, 1470 Madison Avenue, New York, NY, 10029, USA
| | - Yuanji Xu
- Department of Radiation Oncology, Fujian Cancer Hospital & Fujian Medical University Cancer Hospital, Fuzhou, 350011, Fujian Province, China
| | - Rui Zhou
- Fujian Medical University, Fuzhou, 350108, Fujian Province, China
| | - Shaojun Lin
- Department of Radiation Oncology, Fujian Cancer Hospital & Fujian Medical University Cancer Hospital, Fuzhou, 350011, Fujian Province, China
| | - Qiaojuan Guo
- Department of Radiation Oncology, Fujian Cancer Hospital & Fujian Medical University Cancer Hospital, Fuzhou, 350011, Fujian Province, China
| | - Honglin Chen
- State Key Laboratory for Emerging Infectious Diseases, Department of Microbiology and the Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The University of Hong Kong, Hong Kong, SAR, China
| | - Yunbin Ye
- Laboratory of Immuno-Oncology, Fujian Cancer Hospital & Fujian Medical University Cancer Hospital, Fuzhou, 350011, Fujian Province, China.,Fujian Provincial Key Laboratory of Translational Cancer Medicine, Fuzhou, 350014, Fujian, China
| | - Bin Zhang
- Department of Genetics and Genomic Sciences, Icahn Institute of Genomics and Multiscale Biology, Mount Sinai Center for Transformative Disease Modeling, Icahn School of Medicine at Mount Sinai, 1470 Madison Avenue, New York, NY, 10029, USA.
| | - Jianji Pan
- Department of Radiation Oncology, Fujian Cancer Hospital & Fujian Medical University Cancer Hospital, Fuzhou, 350011, Fujian Province, China.
| |
Collapse
|
40
|
Jiang C, Chen J, Xie S, Zhang L, Xiang Y, Lung M, Kam NW, Kwong DLW, Cao S, Guan XY. Evaluation of circulating EBV microRNA BART2-5p in facilitating early detection and screening of nasopharyngeal carcinoma. Int J Cancer 2018; 143:3209-3217. [PMID: 29971780 DOI: 10.1002/ijc.31642] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2017] [Revised: 03/19/2018] [Accepted: 04/17/2018] [Indexed: 12/27/2022]
Affiliation(s)
- Chen Jiang
- Department of Clinical Oncology; The University of Hong Kong; Pokfulam, Hong Kong
- State Key Laboratory of Oncology in Southern China; Collaborative Innovation Center for Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Sun Yat-sen University Cancer Center; Guangzhou China
| | - Jinna Chen
- Department of Clinical Oncology; The University of Hong Kong; Pokfulam, Hong Kong
| | - Shanghang Xie
- Department of Epidemiology, Cancer Prevention Center; State Key Laboratory of Oncology in Southern China, Sun Yat-sen University Cancer Center; Guangzhou China
| | - Lifang Zhang
- Department of Epidemiology, Cancer Prevention Center; State Key Laboratory of Oncology in Southern China, Sun Yat-sen University Cancer Center; Guangzhou China
| | - Yanqun Xiang
- Department of Nasopharyngeal Carcinoma; Sun Yat-Sen University Cancer Center; Guangzhou China
| | - Maria Lung
- Department of Clinical Oncology; The University of Hong Kong; Pokfulam, Hong Kong
| | - Ngar-Woon Kam
- Department of Clinical Oncology; The University of Hong Kong; Pokfulam, Hong Kong
| | - Dora Lai-wan Kwong
- Department of Clinical Oncology; The University of Hong Kong; Pokfulam, Hong Kong
| | - Sumei Cao
- Department of Epidemiology, Cancer Prevention Center; State Key Laboratory of Oncology in Southern China, Sun Yat-sen University Cancer Center; Guangzhou China
| | - Xin-Yuan Guan
- Department of Clinical Oncology; The University of Hong Kong; Pokfulam, Hong Kong
- State Key Laboratory of Oncology in Southern China; Collaborative Innovation Center for Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Sun Yat-sen University Cancer Center; Guangzhou China
| |
Collapse
|
41
|
Sun L, Meckes DG. Methodological Approaches to Study Extracellular Vesicle miRNAs in Epstein⁻Barr Virus-Associated Cancers. Int J Mol Sci 2018; 19:ijms19092810. [PMID: 30231493 PMCID: PMC6164614 DOI: 10.3390/ijms19092810] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2018] [Revised: 09/14/2018] [Accepted: 09/14/2018] [Indexed: 02/07/2023] Open
Abstract
Epstein Barr-virus (EBV) was the first virus identified to be associated with human cancer in 1964 and is found ubiquitously throughout the world's population. It is now established that EBV contributes to the development and progression of multiple human cancers of both lymphoid and epithelial cell origins. EBV encoded miRNAs play an important role in tumor proliferation, angiogenesis, immune escape, tissue invasion, and metastasis. Recently, EBV miRNAs have been found to be released from infected cancer cells in extracellular vesicles (EVs) and regulate gene expression in neighboring uninfected cells present in the tumor microenvironment and possibly at distal sites. As EVs are abundant in many biological fluids, the viral and cellular miRNAs present within EBV-modified EVs may serve as noninvasion markers for cancer diagnosis and prognosis. In this review, we discuss recent advances in EV isolation and miRNA detection, and provide a complete workflow for EV purification from plasma and deep-sequencing for biomarker discovery.
Collapse
Affiliation(s)
- Li Sun
- Department of Biomedical Sciences, Florida State University College of Medicine, Tallahassee, FL 32306, USA.
| | - David G Meckes
- Department of Biomedical Sciences, Florida State University College of Medicine, Tallahassee, FL 32306, USA.
| |
Collapse
|
42
|
El-Sharkawy A, Al Zaidan L, Malki A. Epstein-Barr Virus-Associated Malignancies: Roles of Viral Oncoproteins in Carcinogenesis. Front Oncol 2018; 8:265. [PMID: 30116721 PMCID: PMC6082928 DOI: 10.3389/fonc.2018.00265] [Citation(s) in RCA: 68] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2018] [Accepted: 06/27/2018] [Indexed: 12/19/2022] Open
Abstract
The Epstein–Barr virus (EBV) is the first herpesvirus identified to be associated with human cancers known to infect the majority of the world population. EBV-associated malignancies are associated with a latent form of infection, and several of the EBV-encoded latent proteins are known to mediate cellular transformation. These include six nuclear antigens and three latent membrane proteins (LMPs). In lymphoid and epithelial tumors, viral latent gene expressions have distinct pattern. In both primary and metastatic tumors, the constant expression of latent membrane protein 2A (LMP2A) at the RNA level suggests that this protein is the key player in the EBV-associated tumorigenesis. While LMP2A contributing to the malignant transformation possibly by cooperating with the aberrant host genome. This can be done in part by dysregulating signaling pathways at multiple points, notably in the cell cycle and apoptotic pathways. Recent studies also have confirmed that LMP1 and LMP2 contribute to carcinoma progression and that this may reflect the combined effects of these proteins on activation of multiple signaling pathways. This review article aims to investigate the aforementioned EBV-encoded proteins that reveal established roles in tumor formation, with a greater emphasis on the oncogenic LMPs (LMP1 and LMP2A) and their roles in dysregulating signaling pathways. It also aims to provide a quick look on the six members of the EBV nuclear antigens and their roles in dysregulating apoptosis.
Collapse
Affiliation(s)
- Ahmed El-Sharkawy
- Human Molecular Genetics Laboratory, Institute of Genetics and Biophysics "A. Buzzati-Traverso" (IGB)-CNR, Naples, Italy.,Biomolecular Science Programme, Università Degli Studi Della Campania "Luigi Vanvitelli", Naples, Italy
| | - Lobna Al Zaidan
- Biomedical Science Department, College of Health Sciences, Qatar University, Doha, Qatar
| | - Ahmed Malki
- Biomedical Science Department, College of Health Sciences, Qatar University, Doha, Qatar
| |
Collapse
|
43
|
Infection of Epstein⁻Barr Virus in Type III Latency Modulates Biogenesis of Exosomes and the Expression Profile of Exosomal miRNAs in the Burkitt Lymphoma Mutu Cell Lines. Cancers (Basel) 2018; 10:cancers10070237. [PMID: 30029522 PMCID: PMC6071279 DOI: 10.3390/cancers10070237] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2018] [Accepted: 07/17/2018] [Indexed: 12/15/2022] Open
Abstract
Infection of Epstein–Barr virus (EBV), a ubiquitous human gamma herpesvirus, is associated with various malignancies in B lymphocytes and epithelial cells. EBV encodes 49 microRNAs in two separated regions, termed the BART and BHRF1 loci. Although accumulating evidence demonstrates that EBV infection regulates the profile of microRNAs in the cells, little is known about the microRNAs in exosomes released from infected cells. Here, we characterized the expression profile of intracellular and exosomal microRNAs in EBV-negative, and two related EBV-infected Burkitt lymphoma cell lines having type I and type III latency by next-generation sequencing. We found that the biogenesis of exosomes is upregulated in type III latently infected cells compared with EBV-negative and type I latently infected cells. We also observed that viral and several specific host microRNAs were predominantly incorporated in the exosomes released from the cells in type III latency. We confirmed that multiple viral microRNAs were transferred to the epithelial cells cocultured with EBV-infected B cells. Our findings indicate that EBV infection, in particular in type III latency, modulates the biogenesis of exosomes and the profile of exosomal microRNAs, potentially contributing to phenotypic changes in cells receiving these exosomes.
Collapse
|
44
|
Polakovicova I, Jerez S, Wichmann IA, Sandoval-Bórquez A, Carrasco-Véliz N, Corvalán AH. Role of microRNAs and Exosomes in Helicobacter pylori and Epstein-Barr Virus Associated Gastric Cancers. Front Microbiol 2018; 9:636. [PMID: 29675003 PMCID: PMC5895734 DOI: 10.3389/fmicb.2018.00636] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2017] [Accepted: 03/19/2018] [Indexed: 12/17/2022] Open
Abstract
Emerging evidence suggests that chronic inflammation caused by pathogen infection is connected to the development of various types of cancer. It is estimated that up to 20% of all cancer deaths is linked to infections and inflammation. In gastric cancer, such triggers can be infection of the gastric epithelium by either Helicobacter pylori (H. pylori), a bacterium present in half of the world population; or by Epstein-Barr virus (EBV), a double-stranded DNA virus which has recently been associated with gastric cancer. Both agents can establish lifelong inflammation by evolving to escape immune surveillance and, under certain conditions, contribute to the development of gastric cancer. Non-coding RNAs, mainly microRNAs (miRNAs), influence the host innate and adaptive immune responses, though long non-coding RNAs and viral miRNAs also alter these processes. Reports suggest that chronic infection results in altered expression of host miRNAs. In turn, dysregulated miRNAs modulate the host inflammatory immune response, favoring bacterial survival and persistence within the gastric mucosa. Given the established roles of miRNAs in tumorigenesis and innate immunity, they may serve as an important link between H. pylori- and EBV-associated inflammation and carcinogenesis. Example of this is up-regulation of miR-155 in H. pylori and EBV infection. The tumor environment contains a variety of cells that need to communicate with each other. Extracellular vesicles, especially exosomes, allow these cells to deliver certain type of information to other cells promoting cancer growth and metastasis. Exosomes have been shown to deliver not only various types of genetic information, mainly miRNAs, but also cytotoxin-associated gene A (CagA), a major H. pylori virulence factor. In addition, a growing body of evidence demonstrates that exosomes contain genetic material of viruses and viral miRNAs and proteins such as EBV latent membrane protein 1 (LMP1) which are delivered into recipient cells. In this review, we focus on the dysregulated H. pylori- and EBV-associated miRNAs while trying to unveil possible causal mechanisms. Moreover, we discuss the role of exosomes as vehicles for miRNA delivery in H. pylori- and EBV-related carcinogenesis.
Collapse
Affiliation(s)
- Iva Polakovicova
- Advanced Center for Chronic Diseases, Pontificia Universidad Católica de Chile, Santiago, Chile.,UC Center for Investigational Oncology, Pontificia Universidad Católica de Chile, Santiago, Chile.,Department of Hematology-Oncology, Faculty of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Sofia Jerez
- Department of Hematology-Oncology, Faculty of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Ignacio A Wichmann
- Advanced Center for Chronic Diseases, Pontificia Universidad Católica de Chile, Santiago, Chile.,UC Center for Investigational Oncology, Pontificia Universidad Católica de Chile, Santiago, Chile.,Department of Hematology-Oncology, Faculty of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile
| | | | - Nicolás Carrasco-Véliz
- Advanced Center for Chronic Diseases, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Alejandro H Corvalán
- Advanced Center for Chronic Diseases, Pontificia Universidad Católica de Chile, Santiago, Chile.,UC Center for Investigational Oncology, Pontificia Universidad Católica de Chile, Santiago, Chile.,Department of Hematology-Oncology, Faculty of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile
| |
Collapse
|
45
|
Wang M, Yu F, Wu W, Wang Y, Ding H, Qian L. Epstein-Barr virus-encoded microRNAs as regulators in host immune responses. Int J Biol Sci 2018; 14:565-576. [PMID: 29805308 PMCID: PMC5968849 DOI: 10.7150/ijbs.24562] [Citation(s) in RCA: 68] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2017] [Accepted: 03/06/2018] [Indexed: 12/12/2022] Open
Abstract
Epstein-Barr virus (EBV) is an oncogenic virus that infects over 90% of the world's adult population. EBV can establish life-long latent infection in host due to the balance between EBV and host immune system. EBV latency is associated with various malignancies such as nasopharyngeal carcinoma, gastric carcinoma and Burkitt's lymphoma. EBV is the first human virus that has the capability to encode microRNAs (miRNAs). Remarkably, EBV-encoded miRNAs are abundantly expressed in latently-infected cells and serve important function in viral infection and pathogenesis. Increasing evidence indicates that EBV miRNAs target the host mRNAs involved in cell proliferation, apoptosis and transformation. EBV miRNAs also inhibit the expression of viral antigens, thereby enabling infected cells to escape immune recognition. Intriguingly, EBV miRNAs directly suppress host antiviral immunity by interfering with antigen presentation and immune cell activation. This review will update the current knowledge about EBV miRNAs implicated in host immune responses. An in-depth understanding of the functions of EBV miRNAs in host antiviral immunity will shed light on the EBV-host interactions and provide potential therapeutic targets for the treatment of EBV-associated malignancies.
Collapse
Affiliation(s)
- Man Wang
- Institute for Translational Medicine, Medical College of Qingdao University, Dengzhou Road 38, Qingdao 266021, China
| | - Fei Yu
- Institute for Translational Medicine, Medical College of Qingdao University, Dengzhou Road 38, Qingdao 266021, China
| | - Wei Wu
- Institute for Translational Medicine, Medical College of Qingdao University, Dengzhou Road 38, Qingdao 266021, China
| | - Yu Wang
- Institute for Translational Medicine, Medical College of Qingdao University, Dengzhou Road 38, Qingdao 266021, China
| | - Han Ding
- Institute for Translational Medicine, Medical College of Qingdao University, Dengzhou Road 38, Qingdao 266021, China
| | - Lili Qian
- Institute for Translational Medicine, Medical College of Qingdao University, Dengzhou Road 38, Qingdao 266021, China
| |
Collapse
|
46
|
Huang SCM, Tsao SW, Tsang CM. Interplay of Viral Infection, Host Cell Factors and Tumor Microenvironment in the Pathogenesis of Nasopharyngeal Carcinoma. Cancers (Basel) 2018; 10:E106. [PMID: 29617291 PMCID: PMC5923361 DOI: 10.3390/cancers10040106] [Citation(s) in RCA: 50] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2018] [Revised: 03/29/2018] [Accepted: 03/30/2018] [Indexed: 12/15/2022] Open
Abstract
Undifferentiated nasopharyngeal carcinoma (NPC) is strongly associated with Epstein-Barr virus (EBV) infection. In addition, heavy infiltration of leukocytes is a common characteristic of EBV-associated NPC. It has long been suggested that substantial and interactive impacts between cancer and stromal cells create a tumor microenvironment (TME) to promote tumorigenesis. The coexistence of tumor-infiltrating lymphocytes with EBV-infected NPC cells represents a distinct TME which supports immune evasion and cancer development from the early phase of EBV infection. Intracellularly, EBV-encoded viral products alter host cell signaling to facilitate tumor development and progression. Intercellularly, EBV-infected cancer cells communicate with stromal cells through secretion of cytokines and chemokines, or via release of tumor exosomes, to repress immune surveillance and enhance metastasis. Although high expression of miR-BARTs has been detected in NPC patients, contributions of these more recently discovered viral products to the establishment of TME are still vaguely defined. Further investigations are needed to delineate the mechanistic linkage of the interplay between viral and host factors, especially in relation to TME, which can be harnessed in future therapeutic strategies.
Collapse
Affiliation(s)
| | - Sai Wah Tsao
- School of Biomedical Sciences, University of Hong Kong, Hong Kong SAR, HK, China.
| | - Chi Man Tsang
- School of Biomedical Sciences, University of Hong Kong, Hong Kong SAR, HK, China.
| |
Collapse
|
47
|
Mukohyama J, Iwakiri D, Zen Y, Mukohara T, Minami H, Kakeji Y, Shimono Y. Evaluation of the risk of lymphomagenesis in xenografts by the PCR-based detection of EBV BamHI W region in patient cancer specimens. Oncotarget 2018; 7:50150-50160. [PMID: 27367028 PMCID: PMC5226574 DOI: 10.18632/oncotarget.10322] [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: 04/26/2016] [Accepted: 06/13/2016] [Indexed: 01/27/2023] Open
Abstract
Establishment of patient-derived tumor xenografts (PDXs) is hampered by lymphomagenesis mostly caused by the latently-infected Epstein-Barr virus (EBV) contained in patient cancer tissues. However, the character of patient tissues that result in lymphomagenesis after xenotransplantation is not elucidated. In this study, we analyzed the patient colorectal cancer (CRC) tissues and the PDXs established by their xenotransplantation. We found that 2 of 9 (22%) PDX tumors were EBV-associated human diffuse large B cell lymphoma which was formed by clonal proliferation of human B-cell lymphocytes, were strongly positive for EBER-ISH, and were classified as type III latency. Expression of EBV genes and RNAs, such as EBNAs, LMP1, EBER and EBV-associated microRNAs in patient CRC tissues were unlikely to be associated with lymphomagenesis in PDXs. In contrast, the positive PCR-based amplification of BamHI W region, a major internal repeat in EBV genome, in the patient CRC tissues was correlated with lymphomagenesis in PDXs. These results suggest that the detection of the EBV BamHI W region in the patient surgical specimens will be an effective way to predict the risk of lymphomagenesis in PDXs before xenotransplantation.
Collapse
Affiliation(s)
- Junko Mukohyama
- Division of Molecular and Cellular Biology, Kobe University Graduate School of Medicine, Kobe, Japan.,Division of Gastrointestinal Surgery, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Dai Iwakiri
- Division of Clinical Virology, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Yoh Zen
- Department of Diagnostic Pathology, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Toru Mukohara
- Division of Medical Oncology/Hematology, Kobe University Graduate School of Medicine, Kobe, Japan.,Cancer Center, Kobe University Hospital, Kobe, Japan
| | - Hironobu Minami
- Division of Medical Oncology/Hematology, Kobe University Graduate School of Medicine, Kobe, Japan.,Cancer Center, Kobe University Hospital, Kobe, Japan
| | - Yoshihiro Kakeji
- Division of Gastrointestinal Surgery, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Yohei Shimono
- Division of Molecular and Cellular Biology, Kobe University Graduate School of Medicine, Kobe, Japan.,Division of Medical Oncology/Hematology, Kobe University Graduate School of Medicine, Kobe, Japan
| |
Collapse
|
48
|
Hassan J, Dean J, De Gascun CF, Riordan M, Sweeney C, Connell J, Awan A. Plasma EBV microRNAs in paediatric renal transplant recipients. J Nephrol 2017; 31:445-451. [PMID: 29185211 DOI: 10.1007/s40620-017-0462-2] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2017] [Accepted: 11/13/2017] [Indexed: 11/26/2022]
Abstract
BACKGROUND Epstein-Barr virus (EBV) was the first human virus identified to express microRNA (miRNA). To date, 44 mature miRNAs are encoded for within the EBV genome. EBV miRNAs have not been profiled in paediatric renal transplant recipients. In this study, we investigated circulating EBV miRNA profiles as novel biomarkers in paediatric renal transplant patients. METHODS Forty-two microRNAs encoded within 2 EBV open reading frames (BART and BHRF) were examined in renal transplant recipients who resolved EBV infection (REI) or maintained chronic high viral loads (CHL), and in non-transplant patients with acute infectious mononucleosis (IM). RESULTS Plasma EBV-miR-BART2-5p was present in higher numbers of IM (7/8) and CHL (7/10) compared to REI (7/12) patients. A trend was observed between the numbers of plasma EBV miRNAs expressed and EBV viral load (p < 0.07). Several EBV-miRs including BART7-3p, 15, 9-3p, 11-3p, 1-3p and 3-3p were detected in IM and CHL patients only. The lytic EBV-miRs, BHRF1-2-3p and 1-1, indicating active viral replication, were detected in IM patients only. One CHL patient developed post-transplant lymphoproliferative disease (PTLD) after several years and analysis of 10 samples over a 30-month period showed an average 24-fold higher change in plasma EBV-miR-BART2-5p compared to the CHL group and 110-fold higher change compared to the REI group. CONCLUSIONS Our results suggest that EBV-miR-BART2-5p, which targets the stress-induced immune ligand MICB to escape recognition and elimination by NK cells, may have a role in sustaining high EBV viral loads in CHL paediatric kidney transplant recipients.
Collapse
Affiliation(s)
- Jaythoon Hassan
- National Virus Reference Laboratory, University College Dublin, Belfield, Dublin 4, Ireland.
| | - Jonathan Dean
- National Virus Reference Laboratory, University College Dublin, Belfield, Dublin 4, Ireland
| | - Cillian F De Gascun
- National Virus Reference Laboratory, University College Dublin, Belfield, Dublin 4, Ireland
| | - Michael Riordan
- Department of Nephrology, Children's University Hospital, Temple Street, Dublin 1, Ireland
| | - Clodagh Sweeney
- Department of Nephrology, Children's University Hospital, Temple Street, Dublin 1, Ireland
| | - Jeff Connell
- National Virus Reference Laboratory, University College Dublin, Belfield, Dublin 4, Ireland
| | - Atif Awan
- Department of Nephrology, Children's University Hospital, Temple Street, Dublin 1, Ireland
| |
Collapse
|
49
|
Zhang J, Huang T, Zhou Y, Cheng ASL, Yu J, To KF, Kang W. The oncogenic role of Epstein-Barr virus-encoded microRNAs in Epstein-Barr virus-associated gastric carcinoma. J Cell Mol Med 2017; 22:38-45. [PMID: 28990284 PMCID: PMC5742672 DOI: 10.1111/jcmm.13354] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2017] [Accepted: 07/15/2017] [Indexed: 12/13/2022] Open
Abstract
Epstein–Barr virus (EBV) infection is detected in various epithelial malignancies, such as nasopharyngeal carcinoma (NPC) and gastric cancer (GC). EBV comprises some unique molecular features and encodes viral genes and microRNAs (miRNAs) by its own DNA sequence. EBV genes are required to maintain latency and contribute to oncogenic property. miRNAs encoded by EBV have been shown to contribute to initiation and progression of EBV‐related malignancies. By a number of genomic profiling studies, some EBV miRNAs were confirmed to be highly expressed in EBV‐associated gastric cancer (EBVaGC) samples and cell lines. The majority host targets of the EBV miRNAs are important for promoting cell growth and inhibiting apoptosis, facilitating cell survival and immune evasion. However, the integrated molecular mechanisms related to EBV miRNAs remain to be investigated. In this review, we summarized the crucial role of EBV miRNAs in epithelial malignancies, especially in EBVaGC. Collectively, EBV miRNAs play a significant role in the viral and host gene regulation network. Understanding the comprehensive potential targets and relevant functions of EBV miRNAs in gastric carcinogenesis might provide better clinical translation.
Collapse
Affiliation(s)
- Jinglin Zhang
- Department of Anatomical and Cellular Pathology, State Key Laboratory of Oncology in South China, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong SAR, China.,Institute of Digestive Disease, Partner State Key Laboratory of Digestive Disease, The Chinese University of Hong Kong, Hong Kong SAR, China.,Li Ka Shing Institute of Health Science, Sir Y.K. Pao Cancer Center, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Tingting Huang
- Department of Anatomical and Cellular Pathology, State Key Laboratory of Oncology in South China, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong SAR, China.,Institute of Digestive Disease, Partner State Key Laboratory of Digestive Disease, The Chinese University of Hong Kong, Hong Kong SAR, China.,Li Ka Shing Institute of Health Science, Sir Y.K. Pao Cancer Center, The Chinese University of Hong Kong, Hong Kong SAR, China.,Shenzhen Research Institute, The Chinese University of Hong Kong, Shenzhen, China
| | - Yuhang Zhou
- Department of Anatomical and Cellular Pathology, State Key Laboratory of Oncology in South China, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong SAR, China.,Institute of Digestive Disease, Partner State Key Laboratory of Digestive Disease, The Chinese University of Hong Kong, Hong Kong SAR, China.,Li Ka Shing Institute of Health Science, Sir Y.K. Pao Cancer Center, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Alfred S L Cheng
- Institute of Digestive Disease, Partner State Key Laboratory of Digestive Disease, The Chinese University of Hong Kong, Hong Kong SAR, China.,Shenzhen Research Institute, The Chinese University of Hong Kong, Shenzhen, China.,School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong, China
| | - Jun Yu
- Institute of Digestive Disease, Partner State Key Laboratory of Digestive Disease, The Chinese University of Hong Kong, Hong Kong SAR, China.,Shenzhen Research Institute, The Chinese University of Hong Kong, Shenzhen, China.,Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Hong Kong, China
| | - Ka Fai To
- Department of Anatomical and Cellular Pathology, State Key Laboratory of Oncology in South China, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong SAR, China.,Institute of Digestive Disease, Partner State Key Laboratory of Digestive Disease, The Chinese University of Hong Kong, Hong Kong SAR, China.,Li Ka Shing Institute of Health Science, Sir Y.K. Pao Cancer Center, The Chinese University of Hong Kong, Hong Kong SAR, China.,Shenzhen Research Institute, The Chinese University of Hong Kong, Shenzhen, China
| | - Wei Kang
- Department of Anatomical and Cellular Pathology, State Key Laboratory of Oncology in South China, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong SAR, China.,Institute of Digestive Disease, Partner State Key Laboratory of Digestive Disease, The Chinese University of Hong Kong, Hong Kong SAR, China.,Li Ka Shing Institute of Health Science, Sir Y.K. Pao Cancer Center, The Chinese University of Hong Kong, Hong Kong SAR, China.,Shenzhen Research Institute, The Chinese University of Hong Kong, Shenzhen, China
| |
Collapse
|
50
|
Bergallo M, Gambarino S, Pinon M, Barat V, Montanari P, Daprà V, Galliano I, Calvo PL. EBV-encoded microRNAs profile evaluation in pediatric liver transplant recipients. J Clin Virol 2017; 91:36-41. [DOI: 10.1016/j.jcv.2017.04.002] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2017] [Revised: 03/21/2017] [Accepted: 04/02/2017] [Indexed: 01/08/2023]
|