1
|
Ahmed N, Abusalah MAHA, Farzand A, Absar M, Yusof NY, Rabaan AA, AlSaihati H, Alshengeti A, Alwarthan S, Alsuwailem HS, Alrumaih ZA, Alsayyah A, Yean CY. Updates on Epstein-Barr Virus (EBV)-Associated Nasopharyngeal Carcinoma: Emphasis on the Latent Gene Products of EBV. Medicina (B Aires) 2022; 59:medicina59010002. [PMID: 36676626 PMCID: PMC9863520 DOI: 10.3390/medicina59010002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2022] [Revised: 12/19/2022] [Accepted: 12/19/2022] [Indexed: 12/24/2022] Open
Abstract
Nasopharyngeal carcinoma (NPC) is an uncommon type of malignancy/cancer worldwide. However, NPC is an endemic disease in southeast Asia and southern China and the reasons behind the underlying for such changes are unclear. Even though the Epstein-Barr infection (EBV) has been suggested as an important reason for undistinguishable NPC, the EBV itself is not adequate to source this type of cancer. The risk factors, for example, genetic susceptibility, and environmental factors might be associated with EBV to undertake a part in the NPC carcinogenesis. Normal healthy people have a memory B cell pool where the EBV persists, and any disturbance of this connection leads to virus-associated B cell malignancies. Less is known about the relationship between EBV and epithelial cell tumors, especially the EBV-associated nasopharyngeal carcinoma (EBVaNPC) and EBV-associated gastric carcinoma (EBVaGC). Currently, it is believed that premalignant genetic changes in epithelial cells contribute to the aberrant establishment of viral latency in these tumors. The early and late phases of NPC patients' survival rates vary significantly. The presence of EBV in all tumor cells presents prospects for the development of innovative therapeutic and diagnostic techniques, despite the fact that the virus's exact involvement in the carcinogenic process is presently not very well known. EBV research continues to shed light on the carcinogenic process, which is important for a more comprehensive knowledge of tumor etiology and the development of targeted cancer therapeutics. In order to screen for NPC, EBV-related biomarkers have been widely used in a few high-incidence locations because of their close associations with the risks of NPC. The current review highlights the scientific importance of EBV and its possible association with NPC.
Collapse
Affiliation(s)
- Naveed Ahmed
- Department of Medical Microbiology and Parasitology, School of Medical Sciences, Universiti Sains Malaysia, Kubang Kerian 16150, Kelantan, Malaysia
| | | | - Anam Farzand
- Department of Allied Health Science, Superior University, Lahore 54000, Pakistan
| | - Muhammad Absar
- Department of Medical Microbiology and Parasitology, School of Medical Sciences, Universiti Sains Malaysia, Kubang Kerian 16150, Kelantan, Malaysia
| | - Nik Yusnoraini Yusof
- Institute for Research in Molecular Medicine (INFORMM), Health Campus, Universiti Sains Malaysia, Kubang Kerian 16150, Kelantan, Malaysia
| | - Ali A. Rabaan
- Molecular Diagnostic Laboratory, Johns Hopkins Aramco Healthcare, Dhahran 31311, Saudi Arabia
- College of Medicine, Alfaisal University, Riyadh 11533, Saudi Arabia
- Department of Public Health and Nutrition, The University of Haripur, Haripur 22610, Pakistan
| | - Hajir AlSaihati
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, University of Hafr Al Batin, Hafr Al Batin 39831, Saudi Arabia
| | - Amer Alshengeti
- Department of Pediatrics, College of Medicine, Taibah University, Al-Madinah 41491, Saudi Arabia
- Department of Infection Prevention and Control, Prince Mohammad Bin Abdulaziz Hospital, National Guard Health Affairs, Al-Madinah 41491, Saudi Arabia
| | - Sara Alwarthan
- Department of Internal Medicine, College of Medicine, Imam Abdulrahman Bin Faisal University, Dammam 34212, Saudi Arabia
| | - Haifa S. Alsuwailem
- Department of Medicine, College of Medicine, Princess Norah Bint Abdulrahman University, Riyadh 84428, Saudi Arabia
| | - Zainb A. Alrumaih
- College of Medicine, Imam Abdulrahman Bin Faisal University, Dammam 34212, Saudi Arabia
| | - Ahmed Alsayyah
- Department of Pathology, College of Medicine, Imam Abdulrahman Bin Faisal University, Dammam 31441, Saudi Arabia
| | - Chan Yean Yean
- Department of Medical Microbiology and Parasitology, School of Medical Sciences, Universiti Sains Malaysia, Kubang Kerian 16150, Kelantan, Malaysia
- Institute for Research in Molecular Medicine (INFORMM), Health Campus, Universiti Sains Malaysia, Kubang Kerian 16150, Kelantan, Malaysia
- Correspondence:
| |
Collapse
|
2
|
A practical method to screen and identify functioning biomarkers in nasopharyngeal carcinoma. Sci Rep 2021; 11:7294. [PMID: 33790390 PMCID: PMC8012388 DOI: 10.1038/s41598-021-86809-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2020] [Accepted: 03/19/2021] [Indexed: 12/24/2022] Open
Abstract
Nasopharyngeal carcinoma (NPC) is a rare malignancy, with the unique geographical and ethnically characteristics of distribution. Gene chip and bioinformatics have been employed to reveal regulatory mechanisms in current functional genomics. However, a practical solution addressing the unresolved aspects of microarray data processing and analysis have been long pursuit. This study developed a new method to improve the accuracy of identifying key biomarkers, namely Unit Gamma Measurement (UGM), accounting for multiple hypotheses test statistics distribution, which could reduce the dependency problem. Three mRNA expression profile of NPC were selected to feed UGM. Differentially expressed genes (DEGs) were identified with UGM and hub genes were derived from them to explore their association with NPC using functional enrichment and pathway analysis. 47 potential DEGs were identified by UGM from the 3 selected datasets, and affluent in cysteine-type endopeptidase inhibitor activity, cilium movement, extracellular exosome etc. also participate in ECM-receptor interaction, chemical carcinogenesis, TNF signaling pathway, small cell lung cancer and mismatch repair pathway. Down-regulation of CAPS and WFDC2 can prolongation of the overall survival periods in the patients. ARMC4, SERPINB3, MUC4 etc. have a close relationship with NPC. The UGM is a practical method to identify NPC-associated genes and biomarkers.
Collapse
|
3
|
Wang YP, Zhao YJ, Kong XL. A metalloproteinase of the disintegrin and metalloproteinases and the ThromboSpondin Motifs 6 as a novel marker for colon cancer: functional experiments. Genet Mol Biol 2020; 43:e20190266. [PMID: 33063817 PMCID: PMC7564043 DOI: 10.1590/1678-4685-gmb-2019-0266] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2019] [Accepted: 05/05/2020] [Indexed: 12/28/2022] Open
Abstract
Herein, we aimed to investigate the functions of ADAMTS6 in colon cancer and its potential mechanism. Based on the data acquired from TCGA database, we revealed that ADAMTS6 was highly expressed in colon cancer tissues, and high expression of ADAMTS6 predicted worse prognosis in patients with colon cancer. Moreover, qRT-PCR demonstrated that the levels of ADAMTS6 were higher in colon cancer cell lines (NCI-H508, Caco-2, CW-2 and HCT 116) than that in normal control cell line CCD-18Co. Functional experiments displayed that depletion of ADAMTS6 repressed NCI-H508 cell growth, invasion and migration whilst overexpression of ADAMTS6 facilitated Caco-2 cell growth, invasion and migration. Moreover, ADAMTS6 silencing enhanced the protein expression of E-cadherin and reduced the levels of N-cadherin, Vimentin and Snail in NCI-H508 cells, whereas ADAMTS6 overexpression showed the counter effects in Caco-2 cells. The protein levels of p-AKT and p-p65 were decreased by depletion of ADAMTS6 in NCI-H508 cells, while their levels were enhanced by overexpression of ADAMTS6 in Caco-2 cells. These consequences indicated that the accelerating effect of ADAMTS6 on colon cancer cell growth, migration and invasion might be achieved by modulating EMT and AKT/NF-κB signaling pathway, offering important foundations for colon cancer treatment.
Collapse
Affiliation(s)
- Yun-Peng Wang
- The Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Department of Digestive Endoscopy, Jinan, Shandong, P.R. China
| | - Yu-Jie Zhao
- The Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Department of Digestive Endoscopy, Jinan, Shandong, P.R. China
| | - Xiang-Liang Kong
- The Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Department of Digestive Endoscopy, Jinan, Shandong, P.R. China
| |
Collapse
|
4
|
Chen L, Chan LS, Lung HL, Yip TTC, Ngan RKC, Wong JWC, Lo KW, Ng WT, Lee AWM, Tsao GSW, Lung ML, Mak NK. Crucifera sulforaphane (SFN) inhibits the growth of nasopharyngeal carcinoma through DNA methyltransferase 1 (DNMT1)/Wnt inhibitory factor 1 (WIF1) axis. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2019; 63:153058. [PMID: 31394414 DOI: 10.1016/j.phymed.2019.153058] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/10/2018] [Revised: 07/26/2019] [Accepted: 07/29/2019] [Indexed: 06/10/2023]
Abstract
BACKGROUND Sulforaphane (SFN), a natural compound present in cruciferous vegetable, has been shown to possess anti-cancer activities. Cancer stem cell (CSC) in bulk tumor is generally considered as treatment resistant cell and involved in cancer recurrence. The effects of SFN on nasopharyngeal carcinoma (NPC) CSCs have not yet been explored. PURPOSE The present study aims to examine the anti-tumor activities of SFN on NPC cells with CSC-like properties and the underlying mechanisms. METHODS NPC cells growing in monolayer culture, CSCs-enriched NPC tumor spheres, and also the NPC nude mice xenograft were used to study the anti-tumor activities of SFN on NPC. The population of cells expressing CSC-associated markers was evaluated using flow cytometry and aldehyde dehydrogenase (ALDH) activity assay. The effect of DNA methyltransferase 1 (DNMT1) on the growth of NPC cells was analyzed by using small interfering RNA (siRNA)-mediated silencing method. RESULTS SFN was found to inhibit the formation of CSC-enriched NPC tumor spheres and reduce the population of cells with CSC-associated properties (SRY (Sex determining Region Y)-box 2 (SOX2) and ALDH). In the functional study, SFN was found to restore the expression of Wnt inhibitory factor 1 (WIF1) and the effect was accompanied with the downregulation of DNMT1. The functional activities of WIF1 and DNMT1 were confirmed using exogenously added recombinant WIF1 and siRNA knockdown of DNMT1. Moreover, SFN was found to inhibit the in vivo growth of C666-1 cells and enhance the anti-tumor effects of cisplatin. CONCLUSION Taken together, we demonstrated that SFN could suppress the growth of NPC cells via the DNMT1/WIF1 axis.
Collapse
Affiliation(s)
- Luo Chen
- Department of Biology, Hong Kong Baptist University, Kowloon, Hong Kong, China
| | - Lai Sheung Chan
- Department of Biology, Hong Kong Baptist University, Kowloon, Hong Kong, China
| | - Hong Lok Lung
- Department of Biology, Hong Kong Baptist University, Kowloon, Hong Kong, China
| | - Timothy Tak Chun Yip
- Department of Clinical Oncology, Queen Elizabeth Hospital Hong Kong, Kowloon, Hong Kong, China; Center for Nasopharyngeal Carcinoma Research, University of Hong Kong, Pokfulam, Hong Kong, China
| | - Roger Kai Cheong Ngan
- Department of Clinical Oncology, Queen Elizabeth Hospital Hong Kong, Kowloon, Hong Kong, China; Center for Nasopharyngeal Carcinoma Research, University of Hong Kong, Pokfulam, Hong Kong, China
| | | | - Kwok Wai Lo
- Department of Anatomical and Cellular Pathology and State Key Laboratory of Translational Oncology, The Chinese University of Hong Kong, Shatin, NT, Hong Kong, China
| | - Wai Tong Ng
- Clinical Oncology, Pamela Youde Nethersole Eastern Hospital, Chai Wan, Hong Kong, China
| | - Anne Wing Mui Lee
- Department of Clinical Oncology, Center for Nasopharyngeal Carcinoma Research, University of Hong Kong, Pokfulam, Hong Kong, China
| | - George Sai Wah Tsao
- Department of Anatomy, Center for Nasopharyngeal Carcinoma Research, University of Hong Kong, Pokfulam, Hong Kong, China
| | - Maria Li Lung
- Department of Clinical Oncology, Center for Nasopharyngeal Carcinoma Research, University of Hong Kong, Pokfulam, Hong Kong, China
| | - Nai Ki Mak
- Department of Biology, Hong Kong Baptist University, Kowloon, Hong Kong, China.
| |
Collapse
|
5
|
Li HY, Zhou T, Lin W, Lin S, Zhong H. Association of hypoxia-inducible factor-1α (HIF1α) 1790G/A gene polymorphism with renal cell carcinoma and prostate cancer susceptibility: a meta-analysis. BMC MEDICAL GENETICS 2019; 20:141. [PMID: 31419966 PMCID: PMC6698016 DOI: 10.1186/s12881-019-0874-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/09/2019] [Accepted: 08/08/2019] [Indexed: 02/07/2023]
Abstract
BACKGROUND This meta-analysis was performed to evaluate the relationship between hypoxia-inducible factor-1α (HIF1α) 1790G/A gene polymorphism and the susceptibility to renal cell carcinoma (RCC) and prostate cancer (PCa). METHODS Association investigations were identified and included from the Embase, Cochrane Library and PubMed databases on March 1, 2018, and eligible investigations were analyzed by meta-analysis. Odds ratios (OR) were used to express the dichotomous data, and the 95% confidence intervals (CI) were also calculated. RESULTS In this meta-analysis, we found that the AA genotype of HIF1α 1790G/A was positively associated with the risk of RCC in overall populations, Caucasians, but not for Asians. G allele and GG genotype were not associated with the susceptibility of RCC in overall populations, Caucasians, and Asians. The G allele was negatively associated with PCa susceptibility in overall populations, Asians, but not for Caucasians. GG genotype was negatively associated with PCa susceptibility in Asians, but not for overall populations and Caucasians. HIF1α 1790G/A AA genotype was not associated with PCa susceptibility in overall populations of Caucasians or Asians. CONCLUSION AA genotype of HIF1α 1790G/A was positively associated with RCC risk in overall populations and Caucasians. Furthermore, the G allele was negatively associated with prostate cancer susceptibility in overall populations, Asians, and GG genotype was negatively associated with PCa susceptibility in Asians.
Collapse
Affiliation(s)
- Hong-Yan Li
- Department of Nephrology, Huadu District People’s Hospital of Guangzhou, Southern Medical University, Guangzhou, 510800 China
| | - Tianbiao Zhou
- Department of Nephrology, the Second Affiliated Hospital of Shantou University Medical College, 515041, No 69 Dongxia Road, Shantou, China
| | - Wenshan Lin
- Department of Nephrology, the Second Affiliated Hospital of Shantou University Medical College, 515041, No 69 Dongxia Road, Shantou, China
| | - Shujun Lin
- Department of Nephrology, the Second Affiliated Hospital of Shantou University Medical College, 515041, No 69 Dongxia Road, Shantou, China
| | - Hongzhen Zhong
- Department of Nephrology, the Second Affiliated Hospital of Shantou University Medical College, 515041, No 69 Dongxia Road, Shantou, China
| |
Collapse
|
6
|
Peng S, Huang GL, Xu N, Lu Y, Zeng L, Li X, Luo S, Lyu X, Jiang Q, Li T, He Z. The human ATF1 rs11169571 polymorphism associated with risk of nasopharyngeal carcinoma in Southern Chinese populations. Cancer Med 2019; 8:1893-1898. [PMID: 30905073 PMCID: PMC6488110 DOI: 10.1002/cam4.2022] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2018] [Revised: 01/08/2019] [Accepted: 01/16/2019] [Indexed: 12/12/2022] Open
Abstract
Our previous work reported activating transcription factor 1 (ATF1) is a promotive factor of nasopharyngeal carcinoma (NPC) tumorigenesis. This study is to further explore the association between the human ATF1 rs11169571 polymorphism and the risk of NPC occurrence. The association between ATF1 rs11169571 and risk of NPC occurrence was investigated in clinical samples of 560 patients and 661 controls obtained from southern China with high incidence of NPC. The genotypes were detected by PCR‐RFLP. The differential expression activity of alleles ‐T and ‐C was analyzed with CNE‐2 and C666‐1 cells by luciferase reporter assay. Our data suggested that the allelic frequency and genotypes were significantly different between patients and controls. Compared to the TT homozygote, the TC and CC genotypes have been shown to be significantly decreased in NPC patients (OR = 0.494, 95% CI = 0.387‐0.629, P < 0.001 and OR = 0.556, 95% CI = 0.364‐0.851, P = 0.007, respectively). Compared to the ‐T allele, the ‐C allele is a factor of decreased risk in NPC (OR = 0.642, 95% CI = 0.537‐0.767, P < 0.001). Luciferase reporter activity revealed that the ‐T allele confers a higher expression activity than the ‐C allele in CNE2 cells and C666‐1 cells. In conclusion, ATF1 rs11169571 which could affect the expression of ATF1 is associated with NPC risk.
Collapse
Affiliation(s)
- Shutang Peng
- China-American Cancer Research Institute, Dongguan Scientific Research Center, Guangdong Medical University, Dongguan, China.,Key Laboratory for Epigenetics of Dongguan City, Dongguan, China.,Key Laboratory for Medical Molecular Diagnostics of Guangdong Province, Dongguan, China
| | - Guo-Liang Huang
- China-American Cancer Research Institute, Dongguan Scientific Research Center, Guangdong Medical University, Dongguan, China.,Key Laboratory for Epigenetics of Dongguan City, Dongguan, China.,Key Laboratory for Medical Molecular Diagnostics of Guangdong Province, Dongguan, China
| | - Nansong Xu
- China-American Cancer Research Institute, Dongguan Scientific Research Center, Guangdong Medical University, Dongguan, China.,Key Laboratory for Epigenetics of Dongguan City, Dongguan, China.,Key Laboratory for Medical Molecular Diagnostics of Guangdong Province, Dongguan, China
| | - Yan Lu
- China-American Cancer Research Institute, Dongguan Scientific Research Center, Guangdong Medical University, Dongguan, China.,Key Laboratory for Epigenetics of Dongguan City, Dongguan, China.,Key Laboratory for Medical Molecular Diagnostics of Guangdong Province, Dongguan, China
| | - Liuyan Zeng
- Department of Health Management Center, The Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, China
| | - Xin Li
- Cancer Research Institute, Southern Medical University, Guangzhou, China
| | - Shengqun Luo
- China-American Cancer Research Institute, Dongguan Scientific Research Center, Guangdong Medical University, Dongguan, China.,Key Laboratory for Epigenetics of Dongguan City, Dongguan, China.,Key Laboratory for Medical Molecular Diagnostics of Guangdong Province, Dongguan, China
| | - Xiaoming Lyu
- Department of Laboratory Medicine, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China
| | - Qiang Jiang
- Cancer Research Institute, Southern Medical University, Guangzhou, China
| | - Tong Li
- China-American Cancer Research Institute, Dongguan Scientific Research Center, Guangdong Medical University, Dongguan, China.,Key Laboratory for Epigenetics of Dongguan City, Dongguan, China.,Key Laboratory for Medical Molecular Diagnostics of Guangdong Province, Dongguan, China
| | - Zhiwei He
- China-American Cancer Research Institute, Dongguan Scientific Research Center, Guangdong Medical University, Dongguan, China.,Key Laboratory for Epigenetics of Dongguan City, Dongguan, China.,Key Laboratory for Medical Molecular Diagnostics of Guangdong Province, Dongguan, China
| |
Collapse
|
7
|
Tsang C, Lo K, Nicholls JM, Huang S, Tsao S. Pathogenesis of Nasopharyngeal Carcinoma. NASOPHARYNGEAL CARCINOMA 2019:45-64. [DOI: 10.1016/b978-0-12-814936-2.00003-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
8
|
Lin W, Yip YL, Jia L, Deng W, Zheng H, Dai W, Ko JMY, Lo KW, Chung GTY, Yip KY, Lee SD, Kwan JSH, Zhang J, Liu T, Chan JYW, Kwong DLW, Lee VHF, Nicholls JM, Busson P, Liu X, Chiang AKS, Hui KF, Kwok H, Cheung ST, Cheung YC, Chan CK, Li B, Cheung ALM, Hau PM, Zhou Y, Tsang CM, Middeldorp J, Chen H, Lung ML, Tsao SW. Establishment and characterization of new tumor xenografts and cancer cell lines from EBV-positive nasopharyngeal carcinoma. Nat Commun 2018; 9:4663. [PMID: 30405107 PMCID: PMC6220246 DOI: 10.1038/s41467-018-06889-5] [Citation(s) in RCA: 87] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2017] [Accepted: 09/25/2018] [Indexed: 12/30/2022] Open
Abstract
The lack of representative nasopharyngeal carcinoma (NPC) models has seriously hampered research on EBV carcinogenesis and preclinical studies in NPC. Here we report the successful growth of five NPC patient-derived xenografts (PDXs) from fifty-eight attempts of transplantation of NPC specimens into NOD/SCID mice. The take rates for primary and recurrent NPC are 4.9% and 17.6%, respectively. Successful establishment of a new EBV-positive NPC cell line, NPC43, is achieved directly from patient NPC tissues by including Rho-associated coiled-coil containing kinases inhibitor (Y-27632) in culture medium. Spontaneous lytic reactivation of EBV can be observed in NPC43 upon withdrawal of Y-27632. Whole-exome sequencing (WES) reveals a close similarity in mutational profiles of these NPC PDXs with their corresponding patient NPC. Whole-genome sequencing (WGS) further delineates the genomic landscape and sequences of EBV genomes in these newly established NPC models, which supports their potential use in future studies of NPC. The lack of appropriate models restricts pre-clinical research for nasopharyngeal carcinoma (NPC). Here the authors report the development and characterization of NPC patient-derived xenografts (PDXs), and EBV positive NPC cell line from patient tumor, and suggest their potential use in future NPC research.
Collapse
Affiliation(s)
- Weitao Lin
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Yim Ling Yip
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Lin Jia
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Wen Deng
- School of Nursing, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Hong Zheng
- Department of Clinical Oncology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China.,Center for Biomedical Informatics Research, Stanford University, Stanford, 94305, CA, USA
| | - Wei Dai
- Department of Clinical Oncology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Josephine Mun Yee Ko
- Department of Clinical Oncology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Kwok Wai Lo
- Department of Anatomical and Cellular Pathology and State Key Laboratory of Translational Oncology, The Chinese University of Hong Kong, Hong Kong, China
| | - Grace Tin Yun Chung
- Department of Anatomical and Cellular Pathology and State Key Laboratory of Translational Oncology, The Chinese University of Hong Kong, Hong Kong, China
| | - Kevin Y Yip
- Department of Computer Science and Engineering, The Chinese University of Hong Kong, Hong Kong, China
| | - Sau-Dan Lee
- Department of Computer Science and Engineering, The Chinese University of Hong Kong, Hong Kong, China
| | - Johnny Sheung-Him Kwan
- Department of Anatomical and Cellular Pathology and State Key Laboratory of Translational Oncology, The Chinese University of Hong Kong, Hong Kong, China
| | - Jun Zhang
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Tengfei Liu
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Jimmy Yu-Wai Chan
- Department of Surgery, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Dora Lai-Wan Kwong
- Department of Clinical Oncology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Victor Ho-Fun Lee
- Department of Clinical Oncology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - John Malcolm Nicholls
- Department of Pathology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Pierre Busson
- Gustave Roussy, Paris-Saclay University, CNRS, UMR8126, Villejuif, F-94805, France
| | - Xuefeng Liu
- Center for Cell Reprogramming, Department of Pathology, Georgetown University Medical Center, Washington, 20057, DC, USA.,Department of Endocrinology and Metabolism, The Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan, China.,Affiliated Cancer Hospital & Institute, Guangzhou Medical University, Guangzhou, 510095, Guangdong, China
| | - Alan Kwok Shing Chiang
- Department of Paediatrics and Adolescent Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Kwai Fung Hui
- Department of Paediatrics and Adolescent Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Hin Kwok
- Center for Genomic Sciences, The University of Hong Kong, Hong Kong, China
| | - Siu Tim Cheung
- Department of Surgery and Li Ka Shing Institute of Health Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China
| | - Yuk Chun Cheung
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Chi Keung Chan
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Bin Li
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China.,College of Life Science and Technology, Jinan University, Guangzhou, 510632, Guangdong, China
| | - Annie Lai-Man Cheung
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Pok Man Hau
- Department of Anatomical and Cellular Pathology and State Key Laboratory of Translational Oncology, The Chinese University of Hong Kong, Hong Kong, China
| | - Yuan Zhou
- Department of Anatomical and Cellular Pathology and State Key Laboratory of Translational Oncology, The Chinese University of Hong Kong, Hong Kong, China
| | - Chi Man Tsang
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China.,Department of Anatomical and Cellular Pathology and State Key Laboratory of Translational Oncology, The Chinese University of Hong Kong, Hong Kong, China
| | - Jaap Middeldorp
- VU University Medical Center, Department of Pathology, Cancer Center Amsterdam, de Boelelaan 1117, 1081 HV, Amsterdam, The Netherlands
| | - Honglin Chen
- Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Maria Li Lung
- Department of Clinical Oncology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China.
| | - Sai Wah Tsao
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China.
| |
Collapse
|
9
|
Chai AWY, Cheung AKL, Dai W, Ko JMY, Ip JCY, Chan KW, Kwong DLW, Ng WT, Lee AWM, Ngan RKC, Yau CC, Tung SY, Lee VHF, Lam AKY, Pillai S, Law S, Lung ML. Metastasis-suppressing NID2, an epigenetically-silenced gene, in the pathogenesis of nasopharyngeal carcinoma and esophageal squamous cell carcinoma. Oncotarget 2018; 7:78859-78871. [PMID: 27793011 PMCID: PMC5346683 DOI: 10.18632/oncotarget.12889] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2016] [Accepted: 10/19/2016] [Indexed: 12/13/2022] Open
Abstract
Nidogen-2 (NID2) is a key component of the basement membrane that stabilizes the extracellular matrix (ECM) network. The aim of the study is to analyze the functional roles of NID2 in the pathogenesis of nasopharyngeal carcinoma (NPC) and esophageal squamous cell carcinoma (ESCC). We performed genome-wide methylation profiling of NPC and ESCC and validated our findings using the methylation-sensitive high-resolution melting (MS-HRM) assay. Results showed that promoter methylation of NID2 was significantly higher in NPC and ESCC samples than in their adjacent non-cancer counterparts. Consistently, down-regulation of NID2 was observed in the clinical samples and cell lines of both NPC and ESCC. Re-expression of NID2 suppresses clonogenic survival and migration abilities of transduced NPC and ESCC cells. We showed that NID2 significantly inhibits liver metastasis. Mechanistic studies of signaling pathways also confirm that NID2 suppresses the EGFR/Akt and integrin/FAK/PLCγ metastasis-related pathways. This study provides novel insights into the crucial tumor metastasis suppression roles of NID2 in cancers.
Collapse
Affiliation(s)
- Annie Wai Yeeng Chai
- Department of Clinical Oncology, The University of Hong Kong, Hong Kong (SAR), People's Republic of China
| | - Arthur Kwok Leung Cheung
- Department of Clinical Oncology, The University of Hong Kong, Hong Kong (SAR), People's Republic of China
| | - Wei Dai
- Department of Clinical Oncology, The University of Hong Kong, Hong Kong (SAR), People's Republic of China
| | - Josephine Mun Yee Ko
- Department of Clinical Oncology, The University of Hong Kong, Hong Kong (SAR), People's Republic of China
| | - Joseph Chok Yan Ip
- Department of Clinical Oncology, The University of Hong Kong, Hong Kong (SAR), People's Republic of China
| | - Kwok Wah Chan
- Center for Cancer Research, The University of Hong Kong, Hong Kong (SAR), People's Republic of China.,Department of Pathology, The University of Hong Kong, Hong Kong (SAR), People's Republic of China
| | - Dora Lai-Wan Kwong
- Department of Clinical Oncology, The University of Hong Kong, Hong Kong (SAR), People's Republic of China.,Center for Nasopharyngeal Carcinoma Research, The University of Hong Kong, Hong Kong (SAR), People's Republic of China
| | - Wai Tong Ng
- Center for Nasopharyngeal Carcinoma Research, The University of Hong Kong, Hong Kong (SAR), People's Republic of China.,Department of Clinical Oncology, Pamela Youde Nethersole Eastern Hospital, Hong Kong (SAR), People's Republic of China
| | - Anne Wing Mui Lee
- Department of Clinical Oncology, The University of Hong Kong, Hong Kong (SAR), People's Republic of China.,Center for Nasopharyngeal Carcinoma Research, The University of Hong Kong, Hong Kong (SAR), People's Republic of China
| | - Roger Kai Cheong Ngan
- Center for Nasopharyngeal Carcinoma Research, The University of Hong Kong, Hong Kong (SAR), People's Republic of China.,Department of Clinical Oncology, Queen Elizabeth Hospital, Hong Kong (SAR), People's Republic of China
| | - Chun Chung Yau
- Center for Nasopharyngeal Carcinoma Research, The University of Hong Kong, Hong Kong (SAR), People's Republic of China.,Department of Oncology, Princess Margaret Hospital, Hong Kong (SAR), People's Republic of China
| | - Stewart Yuk Tung
- Center for Nasopharyngeal Carcinoma Research, The University of Hong Kong, Hong Kong (SAR), People's Republic of China.,Department of Clinical Oncology, Tuen Mun Hospital, Hong Kong (SAR), People's Republic of China
| | - Victor Ho Fun Lee
- Department of Clinical Oncology, The University of Hong Kong, Hong Kong (SAR), People's Republic of China.,Center for Nasopharyngeal Carcinoma Research, The University of Hong Kong, Hong Kong (SAR), People's Republic of China
| | - Alfred King-Yin Lam
- Department of Cancer Molecular Pathology, Griffith Medical School and Menzies Health Institute Queensland, Griffith University, Gold Coast, Australia
| | - Suja Pillai
- Department of Cancer Molecular Pathology, Griffith Medical School and Menzies Health Institute Queensland, Griffith University, Gold Coast, Australia
| | - Simon Law
- Center for Cancer Research, The University of Hong Kong, Hong Kong (SAR), People's Republic of China.,Department of Surgery, The University of Hong Kong, Hong Kong (SAR), People's Republic of China
| | - Maria Li Lung
- Department of Clinical Oncology, The University of Hong Kong, Hong Kong (SAR), People's Republic of China.,Center for Cancer Research, The University of Hong Kong, Hong Kong (SAR), People's Republic of China.,Center for Nasopharyngeal Carcinoma Research, The University of Hong Kong, Hong Kong (SAR), People's Republic of China
| |
Collapse
|
10
|
Ni H, Jiang B, Zhou Z, Yuan X, Cao X, Huang G, Li Y. Inactivation of MSH3 by promoter methylation correlates with primary tumor stage in nasopharyngeal carcinoma. Int J Mol Med 2017; 40:673-678. [PMID: 28656302 PMCID: PMC5547962 DOI: 10.3892/ijmm.2017.3044] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2015] [Accepted: 06/22/2017] [Indexed: 11/06/2022] Open
Abstract
The aim of this study was to investigate the inactivation of the MutS homolog human 3 (MSH3) gene by promoter methylation in nasopharyngeal carcinoma (NPC). Methylation-specific PCR, semi-quantitative reverse transcription PCR and immunohistochemical analysis were used to detect methylation and the mRNA and protein expression levels of MSH3 in 54 cases of NPC tissues and 16 cases of normal nasopharyngeal epithelial (NNE) tissues. The association between promoter methylation and mRNA expression, and the mRNA and protein expression of the gene and clinical factors was analyzed. The promoter methylation of MSH3 was detected in 50% (27/54) of the primary tumors, but not in the 16 NNE tissues. The mRNA and protein expression levels were significantly decreased in the 54 cases of human NPC as compared to the 16 NNE tissues (P<0.05). The MSH3-methylated cases exhibited significantly lower mRNA and protein expression levels than the unmethylated cases (P<0.05). The MSH3 mRNA and protein expression levels were significantly associated with the variable T stage (P<0.05); however, they did not correlate with the age and sex of the patients, or with the N stage, TNM classification or histopathological subtype (P>0.05). On the whole, MSH3 was frequently inactivated by promoter methylation and its mRNA and protein expression correlated with the primary tumor stage in NPC.
Collapse
Affiliation(s)
- Haifeng Ni
- Department of Otolaryngology, Affiliated Hangzhou First People's Hospital of Nanjing Medical University, Hangzhou, Zhejiang 310006, P.R. China
| | - Bo Jiang
- Department of Otolaryngology, Affiliated Hangzhou First People's Hospital of Nanjing Medical University, Hangzhou, Zhejiang 310006, P.R. China
| | - Zhen Zhou
- Department of Otolaryngology, Affiliated Hangzhou First People's Hospital of Nanjing Medical University, Hangzhou, Zhejiang 310006, P.R. China
| | - Xiaoyang Yuan
- Department of Otolaryngology, Affiliated Hangzhou First People's Hospital of Nanjing Medical University, Hangzhou, Zhejiang 310006, P.R. China
| | - Xiaolin Cao
- Department of Otolaryngology, Affiliated Hangzhou First People's Hospital of Nanjing Medical University, Hangzhou, Zhejiang 310006, P.R. China
| | - Guangwu Huang
- Department of Otolaryngology, Affiliated Hangzhou First People's Hospital of Nanjing Medical University, Hangzhou, Zhejiang 310006, P.R. China
| | - Yong Li
- Department of Otolaryngology, Affiliated Hangzhou First People's Hospital of Nanjing Medical University, Hangzhou, Zhejiang 310006, P.R. China
| |
Collapse
|
11
|
Ni H, Zhou Z, Jiang B, Yuan X, Cao X, Huang G, Li Y. Inactivation of parkin by promoter methylation correlated with lymph node metastasis and genomic instability in nasopharyngeal carcinoma. Tumour Biol 2017; 39:1010428317695025. [PMID: 28351314 DOI: 10.1177/1010428317695025] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
This study aimed to investigate the inactivation of the parkin gene by promoter methylation and its relationship with genome instability in nasopharyngeal carcinoma. Parkin was considered as a tumor suppressor gene in various types of cancers. However, its role in nasopharyngeal carcinoma is unexplored. Genomic instabilities were detected in nasopharyngeal carcinoma tissues by the random amplified polymorphic DNA. The methylation-specific polymerase chain reaction, semi-quantitative reverse transcription polymerase chain reaction, and immunohistochemical analysis were used to detect methylation and mRNA and protein expression of parkin in 54 cases of nasopharyngeal carcinoma tissues and 16 cases of normal nasopharyngeal epithelia tissues, and in 5 nasopharyngeal carcinoma cell lines (CNE1, CNE2, TWO3, C666, and HONE1) and 1 normal nasopharyngeal epithelia cell line (NP69). mRNA expression of parkin in CNE1 and CNE2 was analyzed before and after methyltransferase inhibitor 5-aza-2-deoxycytidine treatment. The relationship between promoter methylation and mRNA expression, demethylation and mRNA expression, and mRNA and protein expression of the gene and clinical factors and genomic instabilities were analyzed. The mRNA and protein expression levels were significantly reduced in 54 cases of human nasopharyngeal carcinoma compared with 16 cases of normal nasopharyngeal epithelia. Parkin-methylated cases showed significantly lower mRNA and protein expression levels compared with unmethylated cases. After 5-aza-2-deoxycytidine treatment, parkin mRNA expression was restored in CNE1 and CNE2; 92.59% (50/54) of nasopharyngeal carcinoma demonstrated genomic instability. Parkin is frequently inactivated by promoter methylation, and its mRNA and protein expression correlate with lymph node metastasis and genomic instability. Parkin deficiency probably promotes tumorigenesis in nasopharyngeal carcinoma.
Collapse
Affiliation(s)
- Haifeng Ni
- Department of Otolaryngology, Hangzhou First People’s Hospital, Nanjing Medical University, Hangzhou, China
| | - Zhen Zhou
- Department of Otolaryngology, Hangzhou First People’s Hospital, Nanjing Medical University, Hangzhou, China
| | - Bo Jiang
- Department of Otolaryngology, Hangzhou First People’s Hospital, Nanjing Medical University, Hangzhou, China
| | - Xiaoyang Yuan
- Department of Otolaryngology, Hangzhou First People’s Hospital, Nanjing Medical University, Hangzhou, China
| | - Xiaolin Cao
- Department of Otolaryngology, Hangzhou First People’s Hospital, Nanjing Medical University, Hangzhou, China
| | - Guangwu Huang
- Department of Otolaryngology, First Affiliated Hospital, Guangxi Medical University, Nanning, China
| | - Yong Li
- Department of Otolaryngology, Hangzhou First People’s Hospital, Nanjing Medical University, Hangzhou, China
| |
Collapse
|
12
|
Guo Y, Luo W, Hu Z, Li J, Li X, Cao H, Li J, Wen B, Zhang J, Cheng H, Guo W, Tan T, Luo D. Low expression of Aldo-keto reductase 1B10 is a novel independent prognostic indicator for nasopharyngeal carcinoma. Cell Biosci 2016; 6:18. [PMID: 26949513 PMCID: PMC4779195 DOI: 10.1186/s13578-016-0082-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2015] [Accepted: 02/14/2016] [Indexed: 01/19/2023] Open
Abstract
Background Nasopharyngeal carcinoma (NPC) is one of the most common human head and neck cancers with high incidence in Southern China, Southeast Asia and North Africa. Because of its nonspecific symptoms, the early diagnosis of NPC is very difficult. The 5-year survival rate is not ideal in spite of great innovations in radiation and chemotherapy treatments. Highly sensitive and specific prognostic biomarkers are eager for NPC clinical diagnosis. To find specific target molecules is very important for individualized treatment. Aldo–keto reductase B10 (AKR1B10) is closely related to tumorigenesis and tumor development, and however, its expression level in NPC tissues is not clear. Results AKR1B10 expression levels were validated in benign, para-cancerous nasopharyngeal and NPC tissues by immunohistochemical evaluation. AKR1B10 was positively expressed in 42 (82.4 %) of 51 benign specimens, and 235 (98.7 %) of 238 para-carcinoma specimens. This percentage was significantly higher than 44.5 % (133/299) in nasopharyngeal carcinoma tissue (p < 0.01). AKR1B10 mRNA quantitative levels detected by real-time quantitative RT-PCR in 90 NPC tissue samples (0.10 ± 0.21) were significantly lower than that in 15 benign tissue samples (1.03 ± 1.12) (p < 0.01). AKR1B10 expression levels in NPC were correlated negatively with T-classification, lymph node metastasis (p < 0.05). We established nasopharyngeal cancer monoclonal cells CNE-2/AKR1B10 with AKR1B10 stable expression and CNE-2/vector cells without AKR1B10 expression by using a modified lentivirus-mediated method, and found that AKR1B10 inhibited the proliferation of CNE-2/AKR1B10 cells by using MTT assay and flow cytometry, and cell migration by in vitro scratch test. Conclusion Taken together, our data suggest that low expression of AKR1B10 is an independent prognostic indicator in nasopharyngeal carcinoma, and that AKR1B10 may be involved in regulating the proliferation and migration of nasopharyngeal cancer cells.
Collapse
Affiliation(s)
- Yuanwei Guo
- Translational Medicine Institute, National and Local Joint Engineering Laboratory for High-through Molecular Diagnosis Technology, Collaborative Research Center for Post-doctoral Mobile Stations of Central South University, Affiliated The First People's Hospital of Chenzhou, University of South China, 432000 Chenzhou, People's Republic of China.,Center for Clinical Pathology, Affiliated The First People's Hospital of Chenzhou, University of South China, 432000 Chenzhou, People's Republic of China
| | - Weihao Luo
- Translational Medicine Institute, National and Local Joint Engineering Laboratory for High-through Molecular Diagnosis Technology, Collaborative Research Center for Post-doctoral Mobile Stations of Central South University, Affiliated The First People's Hospital of Chenzhou, University of South China, 432000 Chenzhou, People's Republic of China
| | - Zheng Hu
- Translational Medicine Institute, National and Local Joint Engineering Laboratory for High-through Molecular Diagnosis Technology, Collaborative Research Center for Post-doctoral Mobile Stations of Central South University, Affiliated The First People's Hospital of Chenzhou, University of South China, 432000 Chenzhou, People's Republic of China.,Department of Clinical Pharmacology, Xiangya Hospital and Institute of Clinical Pharmacology, Central South University and Hunan Key Laboratory of Pharmacogenetics, 410078 Changsha, Hunan People's Republic of China
| | - Jia Li
- Translational Medicine Institute, National and Local Joint Engineering Laboratory for High-through Molecular Diagnosis Technology, Collaborative Research Center for Post-doctoral Mobile Stations of Central South University, Affiliated The First People's Hospital of Chenzhou, University of South China, 432000 Chenzhou, People's Republic of China
| | - Xiaojie Li
- Center for Clinical Pathology, Affiliated The First People's Hospital of Chenzhou, University of South China, 432000 Chenzhou, People's Republic of China
| | - Huiqiu Cao
- Center for Clinical Pathology, Affiliated The First People's Hospital of Chenzhou, University of South China, 432000 Chenzhou, People's Republic of China
| | - Jun Li
- E.N.T. Department, The First People's Hospital of Chenzhou, 432000 Chenzhou, People's Republic of China
| | - Bo Wen
- Translational Medicine Institute, National and Local Joint Engineering Laboratory for High-through Molecular Diagnosis Technology, Collaborative Research Center for Post-doctoral Mobile Stations of Central South University, Affiliated The First People's Hospital of Chenzhou, University of South China, 432000 Chenzhou, People's Republic of China
| | - Jian Zhang
- Translational Medicine Institute, National and Local Joint Engineering Laboratory for High-through Molecular Diagnosis Technology, Collaborative Research Center for Post-doctoral Mobile Stations of Central South University, Affiliated The First People's Hospital of Chenzhou, University of South China, 432000 Chenzhou, People's Republic of China
| | - Hao Cheng
- E.N.T. Department, The First People's Hospital of Chenzhou, 432000 Chenzhou, People's Republic of China
| | - Wangyuan Guo
- Translational Medicine Institute, National and Local Joint Engineering Laboratory for High-through Molecular Diagnosis Technology, Collaborative Research Center for Post-doctoral Mobile Stations of Central South University, Affiliated The First People's Hospital of Chenzhou, University of South China, 432000 Chenzhou, People's Republic of China
| | - Tan Tan
- Translational Medicine Institute, National and Local Joint Engineering Laboratory for High-through Molecular Diagnosis Technology, Collaborative Research Center for Post-doctoral Mobile Stations of Central South University, Affiliated The First People's Hospital of Chenzhou, University of South China, 432000 Chenzhou, People's Republic of China.,Center for Clinical Pathology, Affiliated The First People's Hospital of Chenzhou, University of South China, 432000 Chenzhou, People's Republic of China
| | - Dixian Luo
- Translational Medicine Institute, National and Local Joint Engineering Laboratory for High-through Molecular Diagnosis Technology, Collaborative Research Center for Post-doctoral Mobile Stations of Central South University, Affiliated The First People's Hospital of Chenzhou, University of South China, 432000 Chenzhou, People's Republic of China.,Center for Clinical Pathology, Affiliated The First People's Hospital of Chenzhou, University of South China, 432000 Chenzhou, People's Republic of China
| |
Collapse
|
13
|
Dai W, Cheung AKL, Ko JMY, Cheng Y, Zheng H, Ngan RKC, Ng WT, Lee AWM, Yau CC, Lee VHF, Lung ML. Comparative methylome analysis in solid tumors reveals aberrant methylation at chromosome 6p in nasopharyngeal carcinoma. Cancer Med 2015; 4:1079-90. [PMID: 25924914 PMCID: PMC4529346 DOI: 10.1002/cam4.451] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2015] [Revised: 02/23/2015] [Accepted: 02/23/2015] [Indexed: 01/01/2023] Open
Abstract
Altered patterns of DNA methylation are key features of cancer. Nasopharyngeal carcinoma (NPC) has the highest incidence in Southern China. Aberrant methylation at the promoter region of tumor suppressors is frequently reported in NPC; however, genome-wide methylation changes have not been comprehensively investigated. Therefore, we systematically analyzed methylome data in 25 primary NPC tumors and nontumor counterparts using a high-throughput approach with the Illumina HumanMethylation450 BeadChip. Comparatively, we examined the methylome data of 11 types of solid tumors collected by The Cancer Genome Atlas (TCGA). In NPC, the hypermethylation pattern was more dominant than hypomethylation and the majority of de novo methylated loci were within or close to CpG islands in tumors. The comparative methylome analysis reveals hypermethylation at chromosome 6p21.3 frequently occurred in NPC (false discovery rate; FDR=1.33 × 10−9), but was less obvious in other types of solid tumors except for prostate and Epstein–Barr virus (EBV)-positive gastric cancer (FDR<10−3). Bisulfite pyrosequencing results further confirmed the aberrant methylation at 6p in an additional patient cohort. Evident enrichment of the repressive mark H3K27me3 and active mark H3K4me3 derived from human embryonic stem cells were found at these regions, indicating both DNA methylation and histone modification function together, leading to epigenetic deregulation in NPC. Our study highlights the importance of epigenetic deregulation in NPC. Polycomb Complex 2 (PRC2), responsible for H3K27 trimethylation, is a promising therapeutic target. A key genomic region on 6p with aberrant methylation was identified. This region contains several important genes having potential use as biomarkers for NPC detection.
Collapse
Affiliation(s)
- Wei Dai
- Department of Clinical Oncology, University of Hong Kong, Hong Kong (SAR), China
| | | | - Josephine Mun Yee Ko
- Department of Clinical Oncology, University of Hong Kong, Hong Kong (SAR), China
| | - Yue Cheng
- Department of Clinical Oncology, University of Hong Kong, Hong Kong (SAR), China
| | - Hong Zheng
- Department of Clinical Oncology, University of Hong Kong, Hong Kong (SAR), China
| | - Roger Kai Cheong Ngan
- Center for Nasopharyngeal Carcinoma Research, University of Hong Kong, Hong Kong (SAR), China.,Department of Clinical Oncology, Queen Elizabeth Hospital, Hong Kong (SAR), China
| | - Wai Tong Ng
- Center for Nasopharyngeal Carcinoma Research, University of Hong Kong, Hong Kong (SAR), China.,Department of Clinical Oncology, Pamela Youde Nethersole Eastern Hospital, Hong Kong (SAR), China
| | - Anne Wing Mui Lee
- Center for Nasopharyngeal Carcinoma Research, University of Hong Kong, Hong Kong (SAR), China.,Department of Clinical Oncology, University of Hong Kong-Shenzhen Hospital, Shenzhen, China
| | - Chun Chung Yau
- Center for Nasopharyngeal Carcinoma Research, University of Hong Kong, Hong Kong (SAR), China.,Department of Oncology, Princess Margaret Hospital, Hong Kong (SAR), China
| | - Victor Ho Fu Lee
- Department of Clinical Oncology, University of Hong Kong, Hong Kong (SAR), China
| | - Maria Li Lung
- Department of Clinical Oncology, University of Hong Kong, Hong Kong (SAR), China.,Center for Nasopharyngeal Carcinoma Research, University of Hong Kong, Hong Kong (SAR), China
| |
Collapse
|
14
|
Abstract
Since its discovery 50 years ago, Epstein-Barr virus (EBV) has been linked to the development of cancers originating from both lymphoid and epithelial cells. Approximately 95% of the world's population sustains an asymptomatic, life-long infection with EBV. The virus persists in the memory B-cell pool of normal healthy individuals, and any disruption of this interaction results in virus-associated B-cell tumors. The association of EBV with epithelial cell tumors, specifically nasopharyngeal carcinoma (NPC) and EBV-positive gastric carcinoma (EBV-GC), is less clear and is currently thought to be caused by the aberrant establishment of virus latency in epithelial cells that display premalignant genetic changes. Although the precise role of EBV in the carcinogenic process is currently poorly understood, the presence of the virus in all tumor cells provides opportunities for developing novel therapeutic and diagnostic approaches. The study of EBV and its role in carcinomas continues to provide insight into the carcinogenic process that is relevant to a broader understanding of tumor pathogenesis and to the development of targeted cancer therapies.
Collapse
Affiliation(s)
- Lawrence S Young
- Warwick Medical School, The University of Warwick, Coventry, CV4 7AL, UK.
| | | |
Collapse
|
15
|
Cheng Y, Ho RLKY, Chan KC, Kan R, Tung E, Lung HL, Yau WL, Cheung AKL, Ko JMY, Zhang ZF, Luo DZ, Feng ZB, Chen S, Guan XY, Kwong D, Stanbridge EJ, Lung ML. Anti-angiogenic pathway associations of the 3p21.3 mapped BLU gene in nasopharyngeal carcinoma. Oncogene 2014; 34:4219-28. [PMID: 25347745 PMCID: PMC4761643 DOI: 10.1038/onc.2014.353] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2014] [Revised: 08/01/2014] [Accepted: 09/02/2014] [Indexed: 12/31/2022]
Abstract
Zinc-finger, MYND-type containing 10 (ZMYND10), or more commonly called BLU, expression is frequently downregulated in nasopharyngeal carcinoma (NPC) and many other tumors due to promoter hypermethylation. Functional evidence shows that the BLU gene inhibits tumor growth in animal assays, but the detailed molecular mechanism responsible for this is still not well understood. In current studies, we find that 93.5% of early-stage primary NPC tumors show downregulated BLU expression. Using a PCR array, overexpression of the BLU gene was correlated to the angiogenesis network in NPC cells. Moreover, expression changes of the MMP family, VEGF and TSP1, were often detected in different stages of NPC, suggesting the possibility that BLU may be directly involved in the microenvironment and anti-angiogenic activity in NPC development. Compared with vector-alone control cells, BLU stable transfectants, derived from poorly-differentiated NPC HONE1 cells, suppress VEGF165, VEGF189 and TSP1 expression at both the RNA and protein levels, and significantly reduce the secreted VEGF protein in these cells, reflecting an unknown regulatory mechanism mediated by the BLU gene in NPC. Cells expressing BLU inhibited cellular invasion, migration and tube formation. These in vitro results were further confirmed by in vivo tumor suppression and a matrigel plug angiogenesis assay in nude mice. Tube-forming ability was clearly inhibited, when the BLU gene is expressed in these cells. Up to 70-90% of injected tumor cells expressing increased exogenous BLU underwent cell death in animal assays. Overexpressed BLU only inhibited VEGF165 expression in differentiated squamous NPC HK1 cells, but also showed an anti-angiogenic effect in the animal assay, revealing a complicated mechanism regulating angiogenesis and the microenvironment in different NPC cell lines. Results of these studies indicate that alteration of BLU gene expression influences anti-angiogenesis pathways and is important for the development of NPC.
Collapse
Affiliation(s)
- Y Cheng
- Department of Clinical Oncology/Center for Nasopharyngeal Carcinoma Research, University of Hong Kong, Hong Kong, SAR, Hong Kong
| | - R L K Y Ho
- Department of Clinical Oncology/Center for Nasopharyngeal Carcinoma Research, University of Hong Kong, Hong Kong, SAR, Hong Kong
| | - K C Chan
- Department of Clinical Oncology/Center for Nasopharyngeal Carcinoma Research, University of Hong Kong, Hong Kong, SAR, Hong Kong
| | - R Kan
- Department of Clinical Oncology/Center for Nasopharyngeal Carcinoma Research, University of Hong Kong, Hong Kong, SAR, Hong Kong
| | - E Tung
- Department of Clinical Oncology/Center for Nasopharyngeal Carcinoma Research, University of Hong Kong, Hong Kong, SAR, Hong Kong
| | - H L Lung
- Department of Clinical Oncology/Center for Nasopharyngeal Carcinoma Research, University of Hong Kong, Hong Kong, SAR, Hong Kong
| | - W L Yau
- Department of Anatomical and Cellular Pathology, Chinese University of Hong Kong, Hong Kong, SAR, Hong Kong
| | - A K L Cheung
- Department of Clinical Oncology/Center for Nasopharyngeal Carcinoma Research, University of Hong Kong, Hong Kong, SAR, Hong Kong
| | - J M Y Ko
- Department of Clinical Oncology/Center for Nasopharyngeal Carcinoma Research, University of Hong Kong, Hong Kong, SAR, Hong Kong
| | - Z F Zhang
- Department of Pathology, Guangxi Medical University, Guangxi, People's Republic of China
| | - D Z Luo
- Department of Pathology, Guangxi Medical University, Guangxi, People's Republic of China
| | - Z B Feng
- Department of Pathology, Guangxi Medical University, Guangxi, People's Republic of China
| | - S Chen
- Department of Pathology, Guangxi Medical University, Guangxi, People's Republic of China
| | - X Y Guan
- Department of Clinical Oncology/Center for Nasopharyngeal Carcinoma Research, University of Hong Kong, Hong Kong, SAR, Hong Kong
| | - D Kwong
- Department of Clinical Oncology/Center for Nasopharyngeal Carcinoma Research, University of Hong Kong, Hong Kong, SAR, Hong Kong
| | - E J Stanbridge
- Department of Microbiology and Molecular Genetics, University of California, Irvine, CA, USA
| | - M L Lung
- Department of Clinical Oncology/Center for Nasopharyngeal Carcinoma Research, University of Hong Kong, Hong Kong, SAR, Hong Kong
| |
Collapse
|
16
|
Xiao X, Zhao W, Tian F, Zhou X, Zhang J, Huang T, Hou B, Du C, Wang S, Mo Y, Yu N, Zhou S, You J, Zhang Z, Huang G, Zeng X. Cytochrome b5 reductase 2 is a novel candidate tumor suppressor gene frequently inactivated by promoter hypermethylation in human nasopharyngeal carcinoma. Tumour Biol 2013; 35:3755-63. [PMID: 24338690 PMCID: PMC3980038 DOI: 10.1007/s13277-013-1497-1] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2013] [Accepted: 11/28/2013] [Indexed: 01/30/2023] Open
Abstract
Cytochrome b5 reductase 2 (CYB5R2), a member of the flavoprotein pyridine nucleotide cytochrome reductase family, is associated with a number of physiological reactions. However, its role in cancer, especially nasopharyngeal carcinoma (NPC), has not been addressed. Here, we investigate the transcript levels and promoter methylation status of CYB5R2 in NPC derived cell lines and tumor biopsies and experimentally address its role as a tumor suppressor gene. We find that CYB5R2 transcript levels are decreased in NPC cell lines and tumor biopsies. Promoter hypermethylation of CYB5R2 was detected in all six tested NPC cell lines and in 84 % of primary NPC tumor biopsies but not in normal nasopharyngeal epithelium. Clinically, CYB5R2 methylation was associated with lymph node metastasis in NPC patients (P < 0.05). The endogenous expression of CYB5R2 could be restored in vitro by the methyltransferase inhibitor 5-aza-2′-deoxycytidine in NPC cell lines. Ectopic expression of CYB5R2 had an inhibitory effect on proliferation, clonogenicity and migration of NPC cells. Moreover, in vivo tests in nude mice indicated that ectopic expression of CYB5R2 reduces the tumorigenicity of CYB5R2-negative NPC cells. Collectively, these findings suggest that CYB5R2 may be a functional tumor suppressor gene, frequently inactivated by hypermethylation of its promoter in NPC. We report here the first instance of epigenetic downregulation in NPC tumor biopsies of a key enzyme, CYB5R2, which is responsible for the detoxification of environmental carcinogens. We propose the possibility of utilizing CYB5R2 promoter methylation as a diagnostic biomarker of NPC in the future.
Collapse
Affiliation(s)
- Xue Xiao
- Department of Otolaryngology-Head and Neck Surgery, First Affiliated Hospital of Guangxi Medical University, 6# Shuangyong Road, Nanning, 530021, China
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
17
|
Wang LJ, Chou YF, Chen PR, Su B, Hsu YC, Chang CH, Lee JW. Differential miRNA expression in repeated recurrence of nasopharyngeal carcinoma. Cancer Lett 2013; 344:188-94. [PMID: 24183849 DOI: 10.1016/j.canlet.2013.10.023] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2013] [Revised: 10/22/2013] [Accepted: 10/24/2013] [Indexed: 12/12/2022]
Abstract
Deregulated microRNAs (miRNAs) are known to be involved in the tumorigenesis of nasopharyngeal carcinoma (NPC). However, the role of miRNA expression in tumor recurrence is not yet understood. We found distinctive miRNA expression in repeated recurrent tumors using miRNA microarray, and verified this using quantitative real-time RT-PCR and miRNA in situ hybridization analysis. Computational analysis and immunohistochemistry further revealed that differentially expressed miRNAs may work in concert to modulate a multitude of biological pathways. The results not only indicate differential miRNA expression during tumor relapse, but imply the potential use of miRNAs to classify repeated recurrence of NPC beyond the histological approach.
Collapse
Affiliation(s)
- Ling-Jung Wang
- Institute of Medical Sciences, Tzu-Chi University, Hualien, Taiwan
| | - Yu-Fu Chou
- Department of Otolaryngology, Buddhist Tzu Chi General Hospital, Hualien, Taiwan
| | - Peir-Rong Chen
- Department of Otolaryngology, Buddhist Tzu Chi General Hospital, Hualien, Taiwan
| | - Borcherng Su
- Department of Pathology, Buddhist Tzu Chi General Hospital, Hualien, Taiwan
| | - Yih-Chih Hsu
- Department of Bioscience Technology, Chung Yuan Christian University, Taoyuan, Taiwan; Center for Nanotechnology, Chung Yuan Christian University, Taoyuan, Taiwan
| | - Chung-Hsing Chang
- Research Center of Excellence for Environment Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan; Department of Dermatology, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Jeng-Woei Lee
- Institute of Medical Sciences, Tzu-Chi University, Hualien, Taiwan; Department of Life Sciences, Tzu-Chi University, Hualien, Taiwan.
| |
Collapse
|
18
|
Physiological β-catenin signaling controls self-renewal networks and generation of stem-like cells from nasopharyngeal carcinoma. BMC Cell Biol 2013; 14:44. [PMID: 24073846 PMCID: PMC3819748 DOI: 10.1186/1471-2121-14-44] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2013] [Accepted: 09/25/2013] [Indexed: 12/30/2022] Open
Abstract
Background A few reports suggested that low levels of Wnt signaling might drive cell reprogramming, but these studies could not establish a clear relationship between Wnt signaling and self-renewal networks. There are ongoing debates as to whether and how the Wnt/β-catenin signaling is involved in the control of pluripotency gene networks. Additionally, whether physiological β-catenin signaling generates stem-like cells through interactions with other pathways is as yet unclear. The nasopharyngeal carcinoma HONE1 cells have low expression of β-catenin and wild-type expression of p53, which provided a possibility to study regulatory mechanism of stemness networks induced by physiological levels of Wnt signaling in these cells. Results Introduction of increased β-catenin signaling, haploid expression of β-catenin under control by its natural regulators in transferred chromosome 3, resulted in activation of Wnt/β-catenin networks and dedifferentiation in HONE1 hybrid cell lines, but not in esophageal carcinoma SLMT1 hybrid cells that had high levels of endogenous β-catenin expression. HONE1 hybrid cells displayed stem cell-like properties, including enhancement of CD24+ and CD44+ populations and generation of spheres that were not observed in parental HONE1 cells. Signaling cascades were detected in HONE1 hybrid cells, including activation of p53- and RB1-mediated tumor suppressor pathways, up-regulation of Nanog-, Oct4-, Sox2-, and Klf4-mediated pluripotency networks, and altered E-cadherin expression in both in vitro and in vivo assays. qPCR array analyses further revealed interactions of physiological Wnt/β-catenin signaling with other pathways such as epithelial-mesenchymal transition, TGF-β, Activin, BMPR, FGFR2, and LIFR- and IL6ST-mediated cell self-renewal networks. Using β-catenin shRNA inhibitory assays, a dominant role for β-catenin in these cellular network activities was observed. The expression of cell surface markers such as CD9, CD24, CD44, CD90, and CD133 in generated spheres was progressively up-regulated compared to HONE1 hybrid cells. Thirty-four up-regulated components of the Wnt pathway were identified in these spheres. Conclusions Wnt/β-catenin signaling regulates self-renewal networks and plays a central role in the control of pluripotency genes, tumor suppressive pathways and expression of cancer stem cell markers. This current study provides a novel platform to investigate the interaction of physiological Wnt/β-catenin signaling with stemness transition networks.
Collapse
|