1
|
He B, Huang Z, Qin S, Peng P, Duan X, Wang L, Ye Q, Wang K, Jiang J, Li B, Liu R, Huang C. Enhanced SLC35B2/SAV1 sulfation axis promotes tumor growth by inhibiting Hippo signaling in HCC. Hepatology 2024:01515467-990000000-00760. [PMID: 38377452 DOI: 10.1097/hep.0000000000000783] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/16/2023] [Accepted: 12/26/2023] [Indexed: 02/22/2024]
Abstract
BACKGROUND AND AIMS Protein tyrosine sulfation (PTS) is a common posttranslational modification that regulates a variety of physiological and pathological processes. However, the role of PTS in cancer remains poorly understood. The goal of this study was to determine whether and how PTS plays a role in HCC progression. APPROACH AND RESULTS By mass spectrometry and bioinformatics analysis, we identified SAV1 as a novel substrate of PTS in HCC. Oxidative stress upregulates the transcription of SLC35B2, a Golgi-resident transporter of sulfate donor 3'-phosphoadenosine 5'-phosphosulfate, leading to increased sulfation of SAV1. Sulfation of SAV1 disrupts the formation of the SAV1-MST1 complex, resulting in a decrease of MST1 phosphorylation and subsequent inactivation of Hippo signaling. These molecular events ultimately foster the growth of HCC cells both in vivo and in vitro. Moreover, SLC35B2 is a novel transcription target gene of the Hippo pathway, constituting a positive feedback loop that facilitates HCC progression under oxidative stress. CONCLUSIONS Our findings reveal a regulatory mechanism of the SLC35B2/SAV1 sulfation axis in response to oxidative stress, highlighting its potential as a promising therapeutic target for HCC.
Collapse
Affiliation(s)
- Bo He
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, and West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, China
| | - Zhao Huang
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, and West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, China
| | - Siyuan Qin
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, and West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, China
| | - Peilan Peng
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, and West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, China
| | - Xirui Duan
- Department of Oncology, Sichuan Academy of Medical Sciences and Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Longqin Wang
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, and West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, China
| | - Qin Ye
- Department of Oncology, Sichuan Academy of Medical Sciences and Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Kui Wang
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, and West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, China
| | - Jingwen Jiang
- Department of Occupational Health and Environmental Health, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China
| | - Bowen Li
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, and West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, China
| | - Rui Liu
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases & Research Unit of Oral Carcinogenesis and Management & Chinese Academy of Medical Sciences, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Canhua Huang
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, and West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, China
| |
Collapse
|
2
|
Okamoto Y, Shikano S. Emerging roles of a chemoattractant receptor GPR15 and ligands in pathophysiology. Front Immunol 2023; 14:1179456. [PMID: 37457732 PMCID: PMC10348422 DOI: 10.3389/fimmu.2023.1179456] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2023] [Accepted: 06/19/2023] [Indexed: 07/18/2023] Open
Abstract
Chemokine receptors play a central role in the maintenance of immune homeostasis and development of inflammation by directing leukocyte migration to tissues. GPR15 is a G protein-coupled receptor (GPCR) that was initially known as a co-receptor for human immunodeficiency virus (HIV) and simian immunodeficiency virus (SIV), with structural similarity to other members of the chemoattractant receptor family. Since the discovery of its novel function as a colon-homing receptor of T cells in mice a decade ago, GPR15 has been rapidly gaining attention for its involvement in a variety of inflammatory and immune disorders. The recent identification of its natural ligand C10orf99, a chemokine-like polypeptide strongly expressed in gastrointestinal tissues, has established that GPR15-C10orf99 is a novel signaling axis that controls intestinal homeostasis and inflammation through the migration of immune cells. In addition, it has been demonstrated that C10orf99-independent functions of GPR15 and GPR15-independent activities of C10orf99 also play significant roles in the pathophysiology. Therefore, GPR15 and its ligands are potential therapeutic targets. To provide a basis for the future development of GPR15- or GPR15 ligand-targeted therapeutics, we have summarized the latest advances in the role of GPR15 and its ligands in human diseases as well as the molecular mechanisms that regulate GPR15 expression and functions.
Collapse
Affiliation(s)
| | - Sojin Shikano
- Department of Biochemistry and Molecular Genetics, University of Illinois at Chicago, Chicago, IL, United States
| |
Collapse
|
3
|
Huang C, Li Y, Ling Q, Wei C, Fang B, Mao X, Yang R, Zhang L, Huang S, Cheng J, Liao N, Wang F, Mo L, Mo Z, Li L. Establishment of a risk score model for bladder urothelial carcinoma based on energy metabolism-related genes and their relationships with immune infiltration. FEBS Open Bio 2023; 13:736-750. [PMID: 36814419 PMCID: PMC10068335 DOI: 10.1002/2211-5463.13580] [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: 10/20/2022] [Revised: 01/28/2023] [Accepted: 02/21/2023] [Indexed: 02/24/2023] Open
Abstract
Bladder urothelial carcinoma (BLCA) is a common malignant tumor of the human urinary system, and a large proportion of BLCA patients have a poor prognosis. Therefore, there is an urgent need to find more efficient and sensitive biomarkers for the prognosis of BLCA patients in clinical practice. RNA sequencing (RNA-seq) data and clinical information were obtained from The Cancer Genome Atlas, and 584 energy metabolism-related genes (EMRGs) were obtained from the Reactome pathway database. Cox regression analysis and least absolute shrinkage and selection operator analysis were applied to assess prognostic genes and build a risk score model. The estimate and cibersort algorithms were used to explore the immune microenvironment, immune infiltration, and checkpoints in BLCA patients. Furthermore, we used the Human Protein Atlas database and our single-cell RNA-seq datasets of BLCA patients to verify the expression of 13 EMRGs at the protein and single-cell levels. We constructed a risk score model; the area under the curve of the model at 5 years was 0.792. The risk score was significantly correlated with the immune markers M0 macrophages, M2 macrophages, CD8 T cells, follicular helper T cells, regulatory T cells, and dendritic activating cells. Furthermore, eight immune checkpoint genes were significantly upregulated in the high-risk group. The risk score model can accurately predict the prognosis of BLCA patients and has clinical application value. In addition, according to the differences in immune infiltration and checkpoints, BLCA patients with the most significant benefit can be selected for immune checkpoint inhibitor therapy.
Collapse
Affiliation(s)
- Caihong Huang
- Center for Genomic and Personalized Medicine, Guangxi Key Laboratory for Genomic and Personalized Medicine, Guangxi Collaborative Innovation Center for Genomic and Personalized Medicine, Guangxi Medical University, Nanning, China.,Department of Urology, Institute of Urology and Nephrology, First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Yexin Li
- Center for Genomic and Personalized Medicine, Guangxi Key Laboratory for Genomic and Personalized Medicine, Guangxi Collaborative Innovation Center for Genomic and Personalized Medicine, Guangxi Medical University, Nanning, China.,Department of Urology, Institute of Urology and Nephrology, First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Qiang Ling
- Center for Genomic and Personalized Medicine, Guangxi Key Laboratory for Genomic and Personalized Medicine, Guangxi Collaborative Innovation Center for Genomic and Personalized Medicine, Guangxi Medical University, Nanning, China.,School of Public Health, Guangxi Medical University, Nanning, China
| | - Chunmeng Wei
- Center for Genomic and Personalized Medicine, Guangxi Key Laboratory for Genomic and Personalized Medicine, Guangxi Collaborative Innovation Center for Genomic and Personalized Medicine, Guangxi Medical University, Nanning, China.,Department of Urology, Institute of Urology and Nephrology, First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Bo Fang
- Center for Genomic and Personalized Medicine, Guangxi Key Laboratory for Genomic and Personalized Medicine, Guangxi Collaborative Innovation Center for Genomic and Personalized Medicine, Guangxi Medical University, Nanning, China
| | - Xingning Mao
- Center for Genomic and Personalized Medicine, Guangxi Key Laboratory for Genomic and Personalized Medicine, Guangxi Collaborative Innovation Center for Genomic and Personalized Medicine, Guangxi Medical University, Nanning, China
| | - Rirong Yang
- Center for Genomic and Personalized Medicine, Guangxi Key Laboratory for Genomic and Personalized Medicine, Guangxi Collaborative Innovation Center for Genomic and Personalized Medicine, Guangxi Medical University, Nanning, China
| | - LuLu Zhang
- Center for Genomic and Personalized Medicine, Guangxi Key Laboratory for Genomic and Personalized Medicine, Guangxi Collaborative Innovation Center for Genomic and Personalized Medicine, Guangxi Medical University, Nanning, China
| | - Shengzhu Huang
- Center for Genomic and Personalized Medicine, Guangxi Key Laboratory for Genomic and Personalized Medicine, Guangxi Collaborative Innovation Center for Genomic and Personalized Medicine, Guangxi Medical University, Nanning, China.,Department of Urology, Institute of Urology and Nephrology, First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Jiwen Cheng
- Center for Genomic and Personalized Medicine, Guangxi Key Laboratory for Genomic and Personalized Medicine, Guangxi Collaborative Innovation Center for Genomic and Personalized Medicine, Guangxi Medical University, Nanning, China.,School of Public Health, Guangxi Medical University, Nanning, China
| | - Naikai Liao
- School of Public Health, Guangxi Medical University, Nanning, China
| | - Fubo Wang
- Center for Genomic and Personalized Medicine, Guangxi Key Laboratory for Genomic and Personalized Medicine, Guangxi Collaborative Innovation Center for Genomic and Personalized Medicine, Guangxi Medical University, Nanning, China.,School of Public Health, Guangxi Medical University, Nanning, China
| | - Linjian Mo
- Center for Genomic and Personalized Medicine, Guangxi Key Laboratory for Genomic and Personalized Medicine, Guangxi Collaborative Innovation Center for Genomic and Personalized Medicine, Guangxi Medical University, Nanning, China.,Department of Urology, Institute of Urology and Nephrology, First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Zengnan Mo
- Center for Genomic and Personalized Medicine, Guangxi Key Laboratory for Genomic and Personalized Medicine, Guangxi Collaborative Innovation Center for Genomic and Personalized Medicine, Guangxi Medical University, Nanning, China.,Department of Urology, Institute of Urology and Nephrology, First Affiliated Hospital of Guangxi Medical University, Nanning, China.,School of Public Health, Guangxi Medical University, Nanning, China
| | - Longman Li
- Center for Genomic and Personalized Medicine, Guangxi Key Laboratory for Genomic and Personalized Medicine, Guangxi Collaborative Innovation Center for Genomic and Personalized Medicine, Guangxi Medical University, Nanning, China.,Department of Urology, Institute of Urology and Nephrology, First Affiliated Hospital of Guangxi Medical University, Nanning, China.,School of Public Health, Guangxi Medical University, Nanning, China
| |
Collapse
|
4
|
Maintenance of Epstein-Barr virus latency through interaction of LMP2A with CXCR4. Arch Virol 2022; 167:1947-1959. [PMID: 35752684 DOI: 10.1007/s00705-022-05511-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2021] [Accepted: 05/08/2022] [Indexed: 11/02/2022]
Abstract
Epstein-Barr virus (EBV) belongs to the subfamily Gammaherpesvirinae and was the first human tumor virus to be discovered. The global rate of EBV infection in adults exceeds 90%. EBV can participate in the regulation of multiple genes and signal pathways through its latency genes. Many studies have shown that CXCR4 is involved in the development of gastric cancer, but there have been few studies on the specific mechanisms involved in EBV-associated gastric cancer (EBVaGC). In this study, we explored the mechanism by which EBV-encoded products maintain latent EBV infection through interaction with CXCR4 and investigated the role of CXCR4 in EBV-positive cells. The results show that there is a positive feedback between the EBV-encoded products and CXCR4, and LMP2A can activate CXCR4 through the NF-κB pathway. In addition, CXCR4 can be fed back to LMP2A and EBNA1 through the ERK signaling pathway. At the same time, CXCR4 can promote the proliferation and migration of EBV-positive cells, reduce the expression of the immediate early protein BZLF1, the late protein EBV gp350, and the viral capsid antigen, and play an important role in maintaining the incubation period of EBV infection. These findings are applicable to the further targeted therapy of EBVaGC.
Collapse
|
5
|
He CY, Wang WM, Wan WD, Liang J, Hu JJ, Yuan YX, Jiang CH, Li N. Tyrosine Sulphation of CXCR4 Induces the Migration of Fibroblast in OSF. Oral Dis 2022; 29:1782-1790. [PMID: 35150031 DOI: 10.1111/odi.14150] [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: 11/03/2021] [Revised: 01/12/2022] [Accepted: 02/07/2022] [Indexed: 11/30/2022]
Abstract
Oral submucous fibrosis (OSF) caused by areca nut chewing is a prevalent fibrotic disease in Asia-Pacific countries. Arecoline-induced migration of fibroblasts (FBs) plays a vital role in the development of OSF. However, the specific molecular mechanisms involved remain unclear. Many studies have shown that tyrosine sulphation of chemokines can influence cell migration. Herein, we demonstrated that arecoline stimulates tyrosine sulphation of the chemokine receptor 4 (CXCR4) through the tyrosylprotein sulphotransferase-1 (TPST-1) to enhance the migration ability of FBs. Moreover, by RNA-Seq analysis, we found that the most significantly altered pathway was the EGFR pathway after the arecoline stimulation for FBs. After the knockdown of arecoline-induced EGFR expression, the tyrosine sulphation of CXCR4 was significantly decreased by the inhibition of TPST-1 induction. Finally, in human OSF specimens, TPST-1 expression was directly correlated with the expression of CXCR4. These data indicate that the arecoline-induced tyrosine sulphation of CXCR4, which is regulated by TPST-1, might be a potential mechanism that contributes to FB migration in OSF.
Collapse
Affiliation(s)
- C Y He
- Department of Oral and Maxillofacial Surgery, Center of Stomatology, Xiangya Hospital, Central South University, Changsha, China
| | - W M Wang
- Department of Oral and Maxillofacial Surgery, Center of Stomatology, Xiangya Hospital, Central South University, Changsha, China.,Institute of Oral Precancerous Lesions, Central South University, Changsha, China.,Research Center of Oral and Maxillofacial Tumor, Xiangya Hospital, Central South University, Changsha, China
| | - W D Wan
- Department of Oral and Maxillofacial Surgery, Center of Stomatology, Xiangya Hospital, Central South University, Changsha, China
| | - J Liang
- Department of Oral and Maxillofacial Surgery, Center of Stomatology, Xiangya Hospital, Central South University, Changsha, China
| | - J J Hu
- Department of Oral and Maxillofacial Surgery, Center of Stomatology, Xiangya Hospital, Central South University, Changsha, China
| | - Y X Yuan
- Department of Oral and Maxillofacial Surgery, Center of Stomatology, Xiangya Hospital, Central South University, Changsha, China.,Institute of Oral Precancerous Lesions, Central South University, Changsha, China.,Research Center of Oral and Maxillofacial Tumor, Xiangya Hospital, Central South University, Changsha, China
| | - C H Jiang
- Department of Oral and Maxillofacial Surgery, Center of Stomatology, Xiangya Hospital, Central South University, Changsha, China.,Institute of Oral Precancerous Lesions, Central South University, Changsha, China.,Research Center of Oral and Maxillofacial Tumor, Xiangya Hospital, Central South University, Changsha, China
| | - N Li
- Department of Oral and Maxillofacial Surgery, Center of Stomatology, Xiangya Hospital, Central South University, Changsha, China.,Institute of Oral Precancerous Lesions, Central South University, Changsha, China.,Research Center of Oral and Maxillofacial Tumor, Xiangya Hospital, Central South University, Changsha, China.,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
| |
Collapse
|
6
|
Wang E, Wang Y, Zhou S, Xia X, Han R, Fei G, Zeng D, Wang R. Identification of three hub genes related to the prognosis of idiopathic pulmonary fibrosis using bioinformatics analysis. Int J Med Sci 2022; 19:1417-1429. [PMID: 36035368 PMCID: PMC9413564 DOI: 10.7150/ijms.73305] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Accepted: 08/09/2022] [Indexed: 12/02/2022] Open
Abstract
Background: Idiopathic pulmonary fibrosis (IPF) is a chronic respiratory disease characterized by peripheral distribution of bilateral pulmonary fibrosis that is more pronounced at the base. IPF has a short median survival time and a poor prognosis. Therefore, it is necessary to identify effective prognostic indicators to guide the treatment of patients with IPF. Methods: We downloaded microarray data of bronchoalveolar lavage cells from the Gene Expression Omnibus (GEO), containing 176 IPF patients and 20 controls. The top 5,000 genes in the median absolute deviation were classified into different color modules using weighted gene co-expression network analysis (WGCNA), and the modules significantly associated with both survival time and survival status were identified as prognostic modules. We used Lasso Cox regression and multivariate Cox regression to search for hub genes related to prognosis from the differentially expressed genes (DEGs) in the prognostic modules and constructed a risk model and nomogram accordingly. Moreover, based on the risk model, we divided IPF patients into high-risk and low-risk groups to determine the biological functions and immune cell subtypes associated with the prognosis of IPF using gene set enrichment analysis and immune cell infiltration analysis. Results: A total of 153 DEGs located in the prognostic modules, three (TPST1, MRVI1, and TM4SF1) of which were eventually defined as prognostic hub genes. A risk model was constructed based on the expression levels of the three hub genes, and the accuracy of the model was evaluated using time-dependent receiver operating characteristic (ROC) curves. The areas under the curve for 1-, 2-, and 3-year survival rates were 0.862, 0.885, and 0.833, respectively. The results of enrichment analysis showed that inflammation and immune processes significantly affected the prognosis of patients with IPF. The degree of mast and natural killer (NK) cell infiltration also increases the prognostic risk of IPF. Conclusions: We identified three hub genes as independent molecular markers to predict the prognosis of patients with IPF and constructed a prognostic model that may be helpful in promoting therapeutic gains for IPF patients.
Collapse
Affiliation(s)
- Enze Wang
- Department of respiratory and critical care medicine, the first affiliated hospital of Anhui medical university, Hefei 230022, China
| | - Yue Wang
- Department of Infectious Diseases, Hefei second people's hospital, Hefei 230001, China
| | - Sijing Zhou
- Department of occupational medicine, Hefei third clinical college of Anhui Medical University, Hefei 230022, China
| | - Xingyuan Xia
- Department of respiratory and critical care medicine, the first affiliated hospital of Anhui medical university, Hefei 230022, China
| | - Rui Han
- Department of respiratory and critical care medicine, the first affiliated hospital of Anhui medical university, Hefei 230022, China
| | - Guanghe Fei
- Department of respiratory and critical care medicine, the first affiliated hospital of Anhui medical university, Hefei 230022, China
| | - Daxiong Zeng
- Department of pulmonary and critical care medicine, Suzhou Dushu Lake Hospital, Suzhou, 215006, China.,Department of pulmonary and critical care medicine, Dushu Lake Hospital Affiliated to Soochow University, Medical Center of Soochow University, Suzhou, 215006, China
| | - Ran Wang
- Department of respiratory and critical care medicine, the first affiliated hospital of Anhui medical university, Hefei 230022, China
| |
Collapse
|
7
|
Lawrie J, Waldrop S, Morozov A, Niu W, Guo J. Engineering of a Small Protein Scaffold To Recognize Sulfotyrosine with High Specificity. ACS Chem Biol 2021; 16:1508-1517. [PMID: 34251168 DOI: 10.1021/acschembio.1c00382] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Protein tyrosine O-sulfation is an essential post-translational modification required for effective biological processes such as hemostasis, inflammatory response, and visual phototransduction. Because of its unstable nature under mass spectrometry conditions and residing on low-abundance cell surface proteins, sulfated tyrosine (sulfotyrosine) residues are difficult to detect or analyze. Enrichment of sulfotyrosine-containing proteins (sulfoproteins) from complex biological samples are typically required before analysis. In this work, we seek to engineer the phosphotyrosine binding pocket of a Src Homology 2 (SH2) domain to act as an antisulfotyrosine antibody mimic. Using tailored selection schemes, several SH2 mutants are identified with high affinity and specificity to sulfotyrosine. Further molecular docking simulations highlight potential mechanisms supporting observed characteristics of these SH2 mutants. Utilities of the evolved SH2 mutants were demonstrated by the detection and enrichment of sulfoproteins.
Collapse
Affiliation(s)
- Justin Lawrie
- Department of Chemistry, University of Nebraska−Lincoln, Lincoln, Nebraska 68588, United States
| | - Sean Waldrop
- Department of Chemistry, University of Nebraska−Lincoln, Lincoln, Nebraska 68588, United States
| | - Anya Morozov
- Department of Chemistry, University of Nebraska−Lincoln, Lincoln, Nebraska 68588, United States
| | - Wei Niu
- Department of Chemical & Biomolecular Engineering, University of Nebraska−Lincoln, Lincoln, Nebraska 68588, United States
- The Nebraska Center for Integrated Biomolecular Communication (NCIBC), University of Nebraska−Lincoln, Lincoln, Nebraska 68588, United States
| | - Jiantao Guo
- Department of Chemistry, University of Nebraska−Lincoln, Lincoln, Nebraska 68588, United States
- The Nebraska Center for Integrated Biomolecular Communication (NCIBC), University of Nebraska−Lincoln, Lincoln, Nebraska 68588, United States
| |
Collapse
|
8
|
Guo XY, Gao XD, Fujita M. Sulfation of a FLAG tag mediated by SLC35B2 and TPST2 affects antibody recognition. PLoS One 2021; 16:e0250805. [PMID: 33951064 PMCID: PMC8099120 DOI: 10.1371/journal.pone.0250805] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2021] [Accepted: 04/14/2021] [Indexed: 11/30/2022] Open
Abstract
A FLAG tag consisting of DYKDDDDK is an epitope tag that is frequently and widely used to detect recombinant proteins of interest. In this study, we performed a CRISPR-based genetic screening to identify factors involved in the detection of a FLAG-tagged misfolded model protein at the cell surface. In the screening, SLC35B2, which encodes 3’-phosphoadenosine-5’-phosphosulfate transporter 1, was identified as the candidate gene. The detection of FLAG-tagged misfolded proteins at the cell surface was significantly increased in SLC35B2-knockout cells. Furthermore, protein tyrosine sulfation mediated by tyrosyl-protein sulfotransferase 2 (TPST2) suppressed FLAG-tagged protein detection. Localization analysis of the FLAG-tagged misfolded proteins confirmed that defects in tyrosine sulfation are only responsible for enhancing anti-FLAG staining on the plasma membrane but not inducing the localization change of misfolded proteins on the plasma membrane. These results suggest that a FLAG tag on the misfolded protein would be sulfated, causing a reduced detection by the M2 anti-FLAG antibody. Attention should be required when quantifying the FLAG-tagged proteins in the secretory pathway.
Collapse
Affiliation(s)
- Xin-Yu Guo
- Key Laboratory of Carbohydrate Chemistry and Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, Wuxi, Jiangsu, China
| | - Xiao-Dong Gao
- Key Laboratory of Carbohydrate Chemistry and Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, Wuxi, Jiangsu, China
| | - Morihisa Fujita
- Key Laboratory of Carbohydrate Chemistry and Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, Wuxi, Jiangsu, China
- * E-mail:
| |
Collapse
|
9
|
Okamoto Y, Shikano S. Tyrosine sulfation and O-glycosylation of chemoattractant receptor GPR15 differentially regulate interaction with GPR15L. J Cell Sci 2021; 134:237784. [PMID: 33758080 DOI: 10.1242/jcs.247833] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2020] [Accepted: 03/15/2021] [Indexed: 12/23/2022] Open
Abstract
GPR15 is a G-protein-coupled receptor (GPCR) that directs lymphocyte homing to the colon and skin. Recent studies have identified a chemokine-like protein GPR15L (also known as C10orf99) as a functional ligand of GPR15. In this study, we examined the structural elements that regulate the GPR15-GPR15L interaction with primary focus on post-translational modifications (PTMs) of receptor N-terminus and on the C-terminus of the ligand. Our findings reveal that the GPR15 receptor is sulfated on the N-terminal tyrosine residue(s) and disruption of tyrosine sulfation inhibits binding of GPR15L. In contrast, the disruption of O-glycosylation on the N-terminal threonine or serine residues, or the removal of α2,3-linked sialic acids from O-glycans, enhances the GPR15L binding. Thus, GPR15 represents a unique chemoattractant receptor in which different N-terminal PTMs regulate its ligand binding in a contrasting manner. We further demonstrate that, unlike canonical chemokines, GPR15L activity critically requires its extreme C-terminal residue and that its hydrophobicity may be a key attribute that facilitates an optimal interaction with the receptor. Our results reveal novel insights into chemoattractant receptor-ligand interaction and provide a valid footing for potential intervention targeting the GPR15-GPR15L axis.
Collapse
Affiliation(s)
- Yukari Okamoto
- Department of Biochemistry and Molecular Genetics, University of Illinois at Chicago, Chicago, IL 60607-7170, USA
| | - Sojin Shikano
- Department of Biochemistry and Molecular Genetics, University of Illinois at Chicago, Chicago, IL 60607-7170, USA
| |
Collapse
|
10
|
Cao Y, Xie L, Shi F, Tang M, Li Y, Hu J, Zhao L, Zhao L, Yu X, Luo X, Liao W, Bode AM. Targeting the signaling in Epstein-Barr virus-associated diseases: mechanism, regulation, and clinical study. Signal Transduct Target Ther 2021; 6:15. [PMID: 33436584 PMCID: PMC7801793 DOI: 10.1038/s41392-020-00376-4] [Citation(s) in RCA: 42] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2020] [Revised: 09/30/2020] [Accepted: 10/15/2020] [Indexed: 12/11/2022] Open
Abstract
Epstein–Barr virus-associated diseases are important global health concerns. As a group I carcinogen, EBV accounts for 1.5% of human malignances, including both epithelial- and lymphatic-originated tumors. Moreover, EBV plays an etiological and pathogenic role in a number of non-neoplastic diseases, and is even involved in multiple autoimmune diseases (SADs). In this review, we summarize and discuss some recent exciting discoveries in EBV research area, which including DNA methylation alterations, metabolic reprogramming, the changes of mitochondria and ubiquitin-proteasome system (UPS), oxidative stress and EBV lytic reactivation, variations in non-coding RNA (ncRNA), radiochemotherapy and immunotherapy. Understanding and learning from this advancement will further confirm the far-reaching and future value of therapeutic strategies in EBV-associated diseases.
Collapse
Affiliation(s)
- Ya Cao
- Key Laboratory of Carcinogenesis and Invasion, Chinese Ministry of Education, Department of Radiology, Xiangya Hospital, Central South University, 410078, Changsha, China. .,Cancer Research Institute and School of Basic Medical Science, Xiangya School of Medicine, Central South University, 410078, Changsha, China. .,Key Laboratory of Carcinogenesis, Chinese Ministry of Health, 410078, Changsha, China. .,Research Center for Technologies of Nucleic Acid-Based Diagnostics and Therapeutics Hunan Province, 410078, Changsha, China. .,Molecular Imaging Research Center of Central South University, 410008, Changsha, Hunan, China. .,National Joint Engineering Research Center for Genetic Diagnostics of Infectious Diseases and Cancer, 410078, Changsha, China. .,Department of Radiology, Xiangya Hospital, Central South University, 410078, Changsha, China.
| | - Longlong Xie
- Key Laboratory of Carcinogenesis and Invasion, Chinese Ministry of Education, Department of Radiology, Xiangya Hospital, Central South University, 410078, Changsha, China.,Cancer Research Institute and School of Basic Medical Science, Xiangya School of Medicine, Central South University, 410078, Changsha, China.,Key Laboratory of Carcinogenesis, Chinese Ministry of Health, 410078, Changsha, China
| | - Feng Shi
- Key Laboratory of Carcinogenesis and Invasion, Chinese Ministry of Education, Department of Radiology, Xiangya Hospital, Central South University, 410078, Changsha, China.,Cancer Research Institute and School of Basic Medical Science, Xiangya School of Medicine, Central South University, 410078, Changsha, China.,Key Laboratory of Carcinogenesis, Chinese Ministry of Health, 410078, Changsha, China
| | - Min Tang
- Key Laboratory of Carcinogenesis and Invasion, Chinese Ministry of Education, Department of Radiology, Xiangya Hospital, Central South University, 410078, Changsha, China.,Cancer Research Institute and School of Basic Medical Science, Xiangya School of Medicine, Central South University, 410078, Changsha, China.,Key Laboratory of Carcinogenesis, Chinese Ministry of Health, 410078, Changsha, China.,Molecular Imaging Research Center of Central South University, 410008, Changsha, Hunan, China
| | - Yueshuo Li
- Key Laboratory of Carcinogenesis and Invasion, Chinese Ministry of Education, Department of Radiology, Xiangya Hospital, Central South University, 410078, Changsha, China.,Cancer Research Institute and School of Basic Medical Science, Xiangya School of Medicine, Central South University, 410078, Changsha, China.,Key Laboratory of Carcinogenesis, Chinese Ministry of Health, 410078, Changsha, China
| | - Jianmin Hu
- Key Laboratory of Carcinogenesis and Invasion, Chinese Ministry of Education, Department of Radiology, Xiangya Hospital, Central South University, 410078, Changsha, China.,Cancer Research Institute and School of Basic Medical Science, Xiangya School of Medicine, Central South University, 410078, Changsha, China.,Key Laboratory of Carcinogenesis, Chinese Ministry of Health, 410078, Changsha, China
| | - Lin Zhao
- Key Laboratory of Carcinogenesis and Invasion, Chinese Ministry of Education, Department of Radiology, Xiangya Hospital, Central South University, 410078, Changsha, China.,Cancer Research Institute and School of Basic Medical Science, Xiangya School of Medicine, Central South University, 410078, Changsha, China.,Key Laboratory of Carcinogenesis, Chinese Ministry of Health, 410078, Changsha, China
| | - Luqing Zhao
- Key Laboratory of Carcinogenesis and Invasion, Chinese Ministry of Education, Department of Radiology, Xiangya Hospital, Central South University, 410078, Changsha, China
| | - Xinfang Yu
- Key Laboratory of Carcinogenesis and Invasion, Chinese Ministry of Education, Department of Radiology, Xiangya Hospital, Central South University, 410078, Changsha, China.,Cancer Research Institute and School of Basic Medical Science, Xiangya School of Medicine, Central South University, 410078, Changsha, China.,Key Laboratory of Carcinogenesis, Chinese Ministry of Health, 410078, Changsha, China
| | - Xiangjian Luo
- Key Laboratory of Carcinogenesis and Invasion, Chinese Ministry of Education, Department of Radiology, Xiangya Hospital, Central South University, 410078, Changsha, China.,Cancer Research Institute and School of Basic Medical Science, Xiangya School of Medicine, Central South University, 410078, Changsha, China.,Key Laboratory of Carcinogenesis, Chinese Ministry of Health, 410078, Changsha, China.,Molecular Imaging Research Center of Central South University, 410008, Changsha, Hunan, China
| | - Weihua Liao
- Department of Radiology, Xiangya Hospital, Central South University, 410078, Changsha, China
| | - Ann M Bode
- The Hormel Institute, University of Minnesota, Austin, MN, 55912, USA
| |
Collapse
|
11
|
Liu D, Zhou B, Liu R. An RNA-sequencing-based transcriptome for a significantly prognostic novel driver signature identification in bladder urothelial carcinoma. PeerJ 2020; 8:e9422. [PMID: 32742772 PMCID: PMC7380276 DOI: 10.7717/peerj.9422] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2019] [Accepted: 06/04/2020] [Indexed: 12/21/2022] Open
Abstract
Bladder cancer (BC) is the ninth most common malignancy worldwide. Bladder urothelial carcinoma (BLCA) constitutes more than 90% of bladder cancer (BC). The five-year survival rate is 5–70%, and patients with BLCA have a poor clinical outcome. The identification of novel clinical molecular markers in BLCA is still urgent to allow for predicting clinical outcomes. This study aimed to identify a novel signature integrating the three-dimension transcriptome of protein coding genes, long non-coding RNAs, microRNAs that is related to the overall survival of patients with BLCA, contributing to earlier prediction and effective treatment selection, as well as to the verification of the established model in the subtypes identified. Gene expression profiling and the clinical information of 400 patients diagnosed with BLCA were retrieved from The Cancer Genome Atlas (TCGA) database. A univariate Cox regression analysis, robust likelihood-based survival modelling analysis and random forests for survival regression and classification algorithms were used to identify the critical biomarkers. A multivariate Cox regression analysis was utilized to construct a risk score formula with a maximum area under the curve (AUC = 0.7669 in the training set). The significant signature could classify patients into high-risk and low-risk groups with significant differences in overall survival time. Similar results were confirmed in the test set (AUC = 0.645) and in the entire set (AUC = 0.710). The multivariate Cox regression analysis indicated that the five-RNA signature was an independent predictive factor for patients with BLCA. Non-negative matrix factorization and a similarity network fusion algorithm were applied for identifying three molecular subtypes. The signature could separate patients in every subtype into high- and low- groups with a distinct difference. Gene set variation analysis of protein-coding genes associated with the five prognostic RNAs demonstrated that the co-expressed protein-coding genes were involved in the pathways and biological process of tumourigenesis. The five-RNA signature could serve as to some degree a reliable independent signature for predicting outcome in patients with BLCA.
Collapse
Affiliation(s)
- Danqi Liu
- Department of Pharmacy, Xiangya Hospital, Central South University, Changsha, Hunan, People's Republic of China.,Institute for Rational and Safe Medication Practices, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, People's Republic of China.,The Hunan Institute of Pharmacy Practice and Clinical Research, Xiangya Hospital, Central South University, Changsha, Hunan, People's Republic of China
| | - Boting Zhou
- Department of Pharmacy, Xiangya Hospital, Central South University, Changsha, Hunan, People's Republic of China.,Institute for Rational and Safe Medication Practices, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, People's Republic of China.,The Hunan Institute of Pharmacy Practice and Clinical Research, Xiangya Hospital, Central South University, Changsha, Hunan, People's Republic of China
| | - Rangru Liu
- Hainan Province Key Laboratory for Drug Preclinical Study of Pharmacology and Toxicology Research, Hainan Medical University, Haikou, Hainan, People's Republic of China
| |
Collapse
|
12
|
Semisynthesis of an evasin from tick saliva reveals a critical role of tyrosine sulfation for chemokine binding and inhibition. Proc Natl Acad Sci U S A 2020; 117:12657-12664. [PMID: 32461364 DOI: 10.1073/pnas.2000605117] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Blood-feeding arthropods produce antiinflammatory salivary proteins called evasins that function through inhibition of chemokine-receptor signaling in the host. Herein, we show that the evasin ACA-01 from the Amblyomma cajennense tick can be posttranslationally sulfated at two tyrosine residues, albeit as a mixture of sulfated variants. Homogenously sulfated variants of the proteins were efficiently assembled via a semisynthetic native chemical ligation strategy. Sulfation significantly improved the binding affinity of ACA-01 for a range of proinflammatory chemokines and enhanced the ability of ACA-01 to inhibit chemokine signaling through cognate receptors. Comparisons of evasin sequences and structural data suggest that tyrosine sulfation serves as a receptor mimetic strategy for recognizing and suppressing the proinflammatory activity of a wide variety of mammalian chemokines. As such, the incorporation of this posttranslational modification (PTM) or mimics thereof into evasins may provide a strategy to optimize tick salivary proteins for antiinflammatory applications.
Collapse
|
13
|
Cancer-Associated Fibroblasts in Undifferentiated Nasopharyngeal Carcinoma: A Putative Role for the EBV-Encoded Oncoprotein, LMP1. Pathogens 2019; 9:pathogens9010008. [PMID: 31861782 PMCID: PMC7168608 DOI: 10.3390/pathogens9010008] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2019] [Revised: 12/17/2019] [Accepted: 12/18/2019] [Indexed: 12/13/2022] Open
Abstract
Undifferentiated nasopharyngeal carcinoma (NPC) is 100% associated with Epstein–Barr virus (EBV) infection, and biopsies display variable levels of expression of the viral oncoprotein, latent membrane protein 1 (LMP1). Emerging evidence suggests an important role for cancer-associated fibroblasts (CAFs) in the NPC tumour microenvironment, yet the interaction between the virus, its latent gene products and the recruitment and activation of CAFs in the NPC tumour stroma remains unclear. This short review will discuss the current evidence for the importance of CAFs in NPC pathogenesis and outline a putative role for the EBV-encoded oncoprotein, LMP1, in governing tumour–stromal interactions.
Collapse
|
14
|
Chen Z, Liu G, Hossain A, Danilova IG, Bolkov MA, Liu G, Tuzankina IA, Tan W. A co-expression network for differentially expressed genes in bladder cancer and a risk score model for predicting survival. Hereditas 2019; 156:24. [PMID: 31333338 PMCID: PMC6617625 DOI: 10.1186/s41065-019-0100-1] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2019] [Accepted: 06/27/2019] [Indexed: 02/07/2023] Open
Abstract
Background Urothelial bladder cancer (BLCA) is one of the most common internal malignancies worldwide with poor prognosis. This study aims to explore effective prognostic biomarkers and construct a prognostic risk score model for patients with BLCA. Methods Weighted gene co-expression network analysis (WGCNA) was used for identifying the co-expression module related to the pathological stage of BLCA based on the RNA-Seq data retrieved from The Cancer Genome Atlas database. Prognostic biomarkers screened by Cox proportional hazard regression model and random forest were used to construct a risk score model that can predict the prognosis of patients with BLCA. The GSE13507 dataset was used as the independent testing dataset to test the performance of the risk score model in predicting the prognosis of patients with BLCA. Results WGCNA identified seven co-expression modules, in which the brown module consisted of 77 genes was most significantly correlated with the pathological stage of BLCA. Cox proportional hazard regression model and random forest identified TPST1 and P3H4 as prognostic biomarkers. Elevated TPST1 and P3H4 expressions were associated with the high pathological stage and worse survival. The risk score model based on the expression level of TPST1 and P3H4 outperformed pathological stage indicators and previously proposed prognostic models. Conclusion The gene co-expression network-based study could provide additional insight into the tumorigenesis and progression of BLCA, and our proposed risk score model may aid physicians in the assessment of the prognosis of patients with BLCA.
Collapse
Affiliation(s)
- Zihao Chen
- 1Department of Urology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515 China
| | - Guojun Liu
- 2Institute of Natural Sciences and Mathematics, Ural Federal University, Ekaterinburg, 620000 Russia
| | - Aslam Hossain
- 2Institute of Natural Sciences and Mathematics, Ural Federal University, Ekaterinburg, 620000 Russia
| | - Irina G Danilova
- 2Institute of Natural Sciences and Mathematics, Ural Federal University, Ekaterinburg, 620000 Russia.,4Institute of Immunology and Physiology, Ural Branch of the Russian Academy of Sciences, Ekaterinburg, 620000 Russia
| | - Mikhail A Bolkov
- 3Institute of Chemical Engineering, Ural Federal University, Ekaterinburg, 620000 Russia.,4Institute of Immunology and Physiology, Ural Branch of the Russian Academy of Sciences, Ekaterinburg, 620000 Russia
| | - Guoqing Liu
- 5School of Life Science and Technology, Inner Mongolia University of Science and Technology, Baotou, 014010 China
| | - Irina A Tuzankina
- 3Institute of Chemical Engineering, Ural Federal University, Ekaterinburg, 620000 Russia.,4Institute of Immunology and Physiology, Ural Branch of the Russian Academy of Sciences, Ekaterinburg, 620000 Russia
| | - Wanlong Tan
- 1Department of Urology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515 China
| |
Collapse
|
15
|
Xiong J, Guo S, Bing Z, Su Y, Guo L. A Comprehensive RNA Expression Signature for Cervical Squamous Cell Carcinoma Prognosis. Front Genet 2019; 9:696. [PMID: 30662454 PMCID: PMC6328499 DOI: 10.3389/fgene.2018.00696] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2018] [Accepted: 12/12/2018] [Indexed: 01/08/2023] Open
Abstract
Clinicopathological characteristics alone are not enough to predict the survival of patients with cervical squamous cell carcinoma (CESC) due to clinical heterogeneity. In recent years, many genes and non-coding RNAs have been shown to be oncogenes or tumor-suppressors in CESC cells. This study aimed to develop a comprehensive transcriptomic signature for CESC patient prognosis. Univariate, multivariate, and Least Absolute Shrinkage and Selection Operator penalized Cox regression were used to identify prognostic signatures for CESC patients from transcriptomic data of The Cancer Genome Atlas. A normalized prognostic index (NPI) was formulated as a synthetical index for CESC prognosis. Time-dependent receiver operating characteristic curve analysis was used to compare prognostic signatures. A prognostic transcriptomic signature was identified, including 1 microRNA, 1 long non-coding RNA, and 6 messenger RNAs. Decreased survival was associated with CESC patients being in the high-risk group stratified by NPI. The NPI was an independent predictor for CESC patient prognosis and it outperformed the known clinicopathological characteristics, microRNA-only signature, gene-only signature, and previously identified microRNA and gene signatures. Function and pathway enrichment analysis revealed that the identified prognostic RNAs were mainly involved in angiogenesis. In conclusion, we proposed a transcriptomic signature for CESC prognosis and it may be useful for effective clinical risk management of CESC patients. Moreover, RNAs in the transcriptomic signature provided clues for downstream experimental validation and mechanism exploration.
Collapse
Affiliation(s)
- Jie Xiong
- Department of Epidemiology and Health Statistics, Xiangya School of Public Health, Central South University, Changsha, China
| | - Shengyu Guo
- Department of Epidemiology and Health Statistics, Xiangya School of Public Health, Central South University, Changsha, China
| | - Zhitong Bing
- Department of Computational Physics, Institute of Modern Physics of Chinese Academy of Sciences, Lanzhou, China
| | - Yanlin Su
- Department of Gynaecology and Obstetrics, Changsha Central Hospital, Changsha, China
| | - Le Guo
- The First Department of Operation, Hunan Provincial People's Hospital, Changsha, China
| |
Collapse
|
16
|
Zhao CX, Zhu W, Ba ZQ, Xu HJ, Liu WD, Zhu B, Wang L, Song YJ, Yuan S, Ren CP. The regulatory network of nasopharyngeal carcinoma metastasis with a focus on EBV, lncRNAs and miRNAs. Am J Cancer Res 2018; 8:2185-2209. [PMID: 30555738 PMCID: PMC6291648] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2018] [Accepted: 10/16/2018] [Indexed: 06/09/2023] Open
Abstract
Metastasis of nasopharyngeal carcinoma (NPC) remains a main cause of death for NPC patients even though great advances have been made in therapeutic approaches. An in-depth study into the molecular mechanisms of NPC metastasis will help us combat NPC. Epstein-Barr virus (EBV) infection is an evident feature of nonkeratinizing NPC and is strongly associated with tumor metastasis. Recently, long noncoding RNAs (lncRNAs) and microRNAs (miRNAs) have become a hot topic of research due to their epigenetic regulatory roles in NPC metastasis. The EBV products, lncRNAs and miRNAs can target each other and share several common signaling pathways, which form an interconnected, complex molecular regulatory network. In this review, we discuss the features of this regulatory network and summarize the molecular mechanisms of NPC metastasis, focusing on EBV, lncRNAs and miRNAs with updated knowledge.
Collapse
Affiliation(s)
- Chen-Xuan Zhao
- The Key Laboratory of Carcinogenesis of The Chinese Ministry of Health and The Key Laboratory of Carcinogenesis and Cancer Invasion of The Chinese Ministry of Education, Xiangya Hospital, Central South UniversityChangsha 410008, Hunan, P. R. China
- Cancer Research Institute, Collaborative Innovation Center for Cancer Medicine, School of Basic Medical Science, Central South UniversityChangsha 410078, Hunan, P. R. China
| | - Wei Zhu
- The Key Laboratory of Carcinogenesis of The Chinese Ministry of Health and The Key Laboratory of Carcinogenesis and Cancer Invasion of The Chinese Ministry of Education, Xiangya Hospital, Central South UniversityChangsha 410008, Hunan, P. R. China
- Cancer Research Institute, Collaborative Innovation Center for Cancer Medicine, School of Basic Medical Science, Central South UniversityChangsha 410078, Hunan, P. R. China
| | - Zheng-Qing Ba
- The Key Laboratory of Carcinogenesis of The Chinese Ministry of Health and The Key Laboratory of Carcinogenesis and Cancer Invasion of The Chinese Ministry of Education, Xiangya Hospital, Central South UniversityChangsha 410008, Hunan, P. R. China
- Cancer Research Institute, Collaborative Innovation Center for Cancer Medicine, School of Basic Medical Science, Central South UniversityChangsha 410078, Hunan, P. R. China
| | - Hong-Juan Xu
- The Key Laboratory of Carcinogenesis of The Chinese Ministry of Health and The Key Laboratory of Carcinogenesis and Cancer Invasion of The Chinese Ministry of Education, Xiangya Hospital, Central South UniversityChangsha 410008, Hunan, P. R. China
- Cancer Research Institute, Collaborative Innovation Center for Cancer Medicine, School of Basic Medical Science, Central South UniversityChangsha 410078, Hunan, P. R. China
| | - Wei-Dong Liu
- The Key Laboratory of Carcinogenesis of The Chinese Ministry of Health and The Key Laboratory of Carcinogenesis and Cancer Invasion of The Chinese Ministry of Education, Xiangya Hospital, Central South UniversityChangsha 410008, Hunan, P. R. China
- Cancer Research Institute, Collaborative Innovation Center for Cancer Medicine, School of Basic Medical Science, Central South UniversityChangsha 410078, Hunan, P. R. China
| | - Bin Zhu
- The Key Laboratory of Carcinogenesis of The Chinese Ministry of Health and The Key Laboratory of Carcinogenesis and Cancer Invasion of The Chinese Ministry of Education, Xiangya Hospital, Central South UniversityChangsha 410008, Hunan, P. R. China
- Cancer Research Institute, Collaborative Innovation Center for Cancer Medicine, School of Basic Medical Science, Central South UniversityChangsha 410078, Hunan, P. R. China
| | - Lei Wang
- The Key Laboratory of Carcinogenesis of The Chinese Ministry of Health and The Key Laboratory of Carcinogenesis and Cancer Invasion of The Chinese Ministry of Education, Xiangya Hospital, Central South UniversityChangsha 410008, Hunan, P. R. China
- Cancer Research Institute, Collaborative Innovation Center for Cancer Medicine, School of Basic Medical Science, Central South UniversityChangsha 410078, Hunan, P. R. China
| | - Yu-Jia Song
- The Key Laboratory of Carcinogenesis of The Chinese Ministry of Health and The Key Laboratory of Carcinogenesis and Cancer Invasion of The Chinese Ministry of Education, Xiangya Hospital, Central South UniversityChangsha 410008, Hunan, P. R. China
- Cancer Research Institute, Collaborative Innovation Center for Cancer Medicine, School of Basic Medical Science, Central South UniversityChangsha 410078, Hunan, P. R. China
| | - Shuai Yuan
- The Key Laboratory of Carcinogenesis of The Chinese Ministry of Health and The Key Laboratory of Carcinogenesis and Cancer Invasion of The Chinese Ministry of Education, Xiangya Hospital, Central South UniversityChangsha 410008, Hunan, P. R. China
- Cancer Research Institute, Collaborative Innovation Center for Cancer Medicine, School of Basic Medical Science, Central South UniversityChangsha 410078, Hunan, P. R. China
| | - Cai-Ping Ren
- The Key Laboratory of Carcinogenesis of The Chinese Ministry of Health and The Key Laboratory of Carcinogenesis and Cancer Invasion of The Chinese Ministry of Education, Xiangya Hospital, Central South UniversityChangsha 410008, Hunan, P. R. China
- Cancer Research Institute, Collaborative Innovation Center for Cancer Medicine, School of Basic Medical Science, Central South UniversityChangsha 410078, Hunan, P. R. China
| |
Collapse
|
17
|
Shi Y, Liu N, Lai W, Yan B, Chen L, Liu S, Liu S, Wang X, Xiao D, Liu X, Mao C, Jiang Y, Jia J, Liu Y, Yang R, Cao Y, Tao Y. Nuclear EGFR-PKM2 axis induces cancer stem cell-like characteristics in irradiation-resistant cells. Cancer Lett 2018; 422:81-93. [PMID: 29477380 DOI: 10.1016/j.canlet.2018.02.028] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2017] [Revised: 01/31/2018] [Accepted: 02/17/2018] [Indexed: 12/17/2022]
Abstract
Radiation therapy has become an important tool in the treatment of cancer patients, but most patients relapse within 5 years. Relapse is due to the presence of cancer stem cells (CSCs), but the molecular mechanism of radioresistance in CSCs remains largely elusive. Here, we found that irradiation-resistant (IR) cells exhibited increased stem cell-like properties together with elevated anchorage-independent growth and metastasis ability. EGFR not only leads to increased acquisition of endometrial cancer stem cell markers in radioresistant sublines but is critical for the cancer stem-cell phenotype and tumorigenicity. Moreover, PKM2 functions as an interacting partner of EGFR, which induces the EMT phenotype and stem cell-like properties in IR cells. Finally, we found that the regulatory function of the EGFR-PKM2 axis is dependent on nuclear EGFR. In sum, our study indicated that EGFR and PKM2 directly interact and bind with each other to regulate the transcription of stemness-related genes and promote the stem-like phenotype, thus promoting invasion and metastasis.
Collapse
Affiliation(s)
- Ying Shi
- Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, Hunan, 410008 China; Cancer Research Institute, Key Laboratory of Carcinogenesis, Ministry of Health, Central South University, 110 Xiangya Road, Changsha, Hunan, 410078 China
| | - Na Liu
- Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, Hunan, 410008 China; Cancer Research Institute, Key Laboratory of Carcinogenesis, Ministry of Health, Central South University, 110 Xiangya Road, Changsha, Hunan, 410078 China
| | - Weiwei Lai
- Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, Hunan, 410008 China; Cancer Research Institute, Key Laboratory of Carcinogenesis, Ministry of Health, Central South University, 110 Xiangya Road, Changsha, Hunan, 410078 China
| | - Bin Yan
- Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, Hunan, 410008 China; Cancer Research Institute, Key Laboratory of Carcinogenesis, Ministry of Health, Central South University, 110 Xiangya Road, Changsha, Hunan, 410078 China
| | - Ling Chen
- Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, Hunan, 410008 China; Cancer Research Institute, Key Laboratory of Carcinogenesis, Ministry of Health, Central South University, 110 Xiangya Road, Changsha, Hunan, 410078 China
| | - Shouping Liu
- Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, Hunan, 410008 China; Cancer Research Institute, Key Laboratory of Carcinogenesis, Ministry of Health, Central South University, 110 Xiangya Road, Changsha, Hunan, 410078 China
| | - Shuang Liu
- Institute of Medical Sciences, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, Hunan, 410008 China
| | - Xiang Wang
- Department of Thoracic Surgery, Second Xiangya Hospital, Central South University, Changsha, China
| | - Desheng Xiao
- Department of Pathology, Xiangya Hospital, Central South University, Changsha, Hunan, 410008 China
| | - Xiaoli Liu
- Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, Hunan, 410008 China; Cancer Research Institute, Key Laboratory of Carcinogenesis, Ministry of Health, Central South University, 110 Xiangya Road, Changsha, Hunan, 410078 China
| | - Chao Mao
- Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, Hunan, 410008 China; Cancer Research Institute, Key Laboratory of Carcinogenesis, Ministry of Health, Central South University, 110 Xiangya Road, Changsha, Hunan, 410078 China
| | - Yiqun Jiang
- Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, Hunan, 410008 China; Cancer Research Institute, Key Laboratory of Carcinogenesis, Ministry of Health, Central South University, 110 Xiangya Road, Changsha, Hunan, 410078 China
| | - Jiantao Jia
- Department of Pathophysiology, Changzhi Medical College, Changzhi, Shanxi, 046000 China
| | - Yating Liu
- Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, Hunan, 410008 China; Cancer Research Institute, Key Laboratory of Carcinogenesis, Ministry of Health, Central South University, 110 Xiangya Road, Changsha, Hunan, 410078 China
| | - Rui Yang
- Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, Hunan, 410008 China; Cancer Research Institute, Key Laboratory of Carcinogenesis, Ministry of Health, Central South University, 110 Xiangya Road, Changsha, Hunan, 410078 China
| | - Ya Cao
- Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, Hunan, 410008 China; Cancer Research Institute, Key Laboratory of Carcinogenesis, Ministry of Health, Central South University, 110 Xiangya Road, Changsha, Hunan, 410078 China
| | - Yongguang Tao
- Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, Hunan, 410008 China; Cancer Research Institute, Key Laboratory of Carcinogenesis, Ministry of Health, Central South University, 110 Xiangya Road, Changsha, Hunan, 410078 China; Department of Thoracic Surgery, Second Xiangya Hospital, Central South University, Changsha, China.
| |
Collapse
|
18
|
Yan B, Liu S, Shi Y, Liu N, Chen L, Wang X, Xiao D, Liu X, Mao C, Jiang Y, Lai W, Xin X, Tang CE, Luo D, Tan T, Jia J, Liu Y, Yang R, Huang J, Zhou H, Cheng Y, Cao Y, Yu W, Muegge K, Tao Y. Activation of AhR with nuclear IKKα regulates cancer stem-like properties in the occurrence of radioresistance. Cell Death Dis 2018; 9:490. [PMID: 29706625 PMCID: PMC5924755 DOI: 10.1038/s41419-018-0542-9] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2018] [Revised: 03/04/2018] [Accepted: 03/27/2018] [Indexed: 12/19/2022]
Abstract
Most cancer patients receive radiotherapy in the course of their disease and the occurrence of radioresistance is associated with poor prognosis. The molecular pathways that drive enhanced tumorigenic potential during the development of radioresistance are poorly understood. Here, we demonstrate that aryl hydrocarbon receptor (AhR) plays a vital role in the maintenance of cancer stem-like properties. AhR promotes the cancer stem-like phenotype and drives metastasis by directly targeting the promoters of 'stemness' genes, such as the ATP-binding cassette sub-family G member 2 (ABCG2) gene. Moreover, the radioresistant sublines display high levels of oncometabolites including α-ketoglutarate, and treatment of cancer cells with α-ketoglutarate enhances their stem-like properties in an AhR activation-dependent manner. IKKα directly activates stemness-related genes through an interaction with AhR as a bone fide chromatin modifier. Thus, AhR is functionally linked with cancer stem-like properties, and it drives tumorigenesis in the occurrence of radioresistance.
Collapse
Affiliation(s)
- Bin Yan
- Institute of Medical Sciences, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, 410008, Hunan, China
- Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, 410008, Hunan, China
- Cancer Research Institute, Central South University, 110 Xiangya Road, Changsha, 410078, Hunan, China
| | - Shuang Liu
- Institute of Medical Sciences, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, 410008, Hunan, China.
- Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, 410008, Hunan, China.
| | - Ying Shi
- Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, 410008, Hunan, China
- Cancer Research Institute, Central South University, 110 Xiangya Road, Changsha, 410078, Hunan, China
| | - Na Liu
- Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, 410008, Hunan, China
- Cancer Research Institute, Central South University, 110 Xiangya Road, Changsha, 410078, Hunan, China
| | - Ling Chen
- Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, 410008, Hunan, China
- Cancer Research Institute, Central South University, 110 Xiangya Road, Changsha, 410078, Hunan, China
| | - Xiang Wang
- Department of Thoracic Surgery, Second Xiangya Hospital, Central South University, Changsha, China
| | - Desheng Xiao
- Department of Pathology, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
| | - Xiaoli Liu
- Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, 410008, Hunan, China
- Cancer Research Institute, Central South University, 110 Xiangya Road, Changsha, 410078, Hunan, China
| | - Chao Mao
- Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, 410008, Hunan, China
- Cancer Research Institute, Central South University, 110 Xiangya Road, Changsha, 410078, Hunan, China
| | - Yiqun Jiang
- Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, 410008, Hunan, China
- Cancer Research Institute, Central South University, 110 Xiangya Road, Changsha, 410078, Hunan, China
| | - Weiwei Lai
- Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, 410008, Hunan, China
- Cancer Research Institute, Central South University, 110 Xiangya Road, Changsha, 410078, Hunan, China
| | - Xing Xin
- Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, 410008, Hunan, China
- Cancer Research Institute, Central South University, 110 Xiangya Road, Changsha, 410078, Hunan, China
| | - Can-E Tang
- Cancer Research Institute, Central South University, 110 Xiangya Road, Changsha, 410078, Hunan, China
| | - Dixian Luo
- National and Local Joint Engineering Laboratory of High-throughput Molecular Diagnosis Technology, Translational Medicine Institute, the First People's Hospital of Chenzhou, University of South China, 102 Luojiajing Road, Chenzhou, 423000, Hunan, China
| | - Tan Tan
- National and Local Joint Engineering Laboratory of High-throughput Molecular Diagnosis Technology, Translational Medicine Institute, the First People's Hospital of Chenzhou, University of South China, 102 Luojiajing Road, Chenzhou, 423000, Hunan, China
| | - Jiantao Jia
- Department of Pathophysiology, Changzhi Medical College, Changzhi, Shanxi, China
| | - Yating Liu
- Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, 410008, Hunan, China
- Cancer Research Institute, Central South University, 110 Xiangya Road, Changsha, 410078, Hunan, China
| | - Rui Yang
- Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, 410008, Hunan, China
- Cancer Research Institute, Central South University, 110 Xiangya Road, Changsha, 410078, Hunan, China
| | - Jun Huang
- Department of Neurosugery, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, 410078, Hunan, China
| | - Hu Zhou
- Shanghai Institute of Material Medica, Chinese Academy of Sciences (CAS), 555 Zu Chongzhi Road, Zhangjiang Hi-Tech Park, 201203, Shanghai, China
| | - Yan Cheng
- Department of Pharmacology, School of Pharmaceutical Sciences, Central South University, Changsha, 410078, Hunan, China
| | - Ya Cao
- Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, 410008, Hunan, China
- Cancer Research Institute, Central South University, 110 Xiangya Road, Changsha, 410078, Hunan, China
| | - Weishi Yu
- Cipher Gene (Beijing) Co. Ltd., 100089, Beijing, China
| | - Kathrin Muegge
- Mouse Cancer Genetics Program, National Cancer Institute, Basic Science Program, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, MD, 21702, USA
| | - Yongguang Tao
- Institute of Medical Sciences, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, 410008, Hunan, China.
- Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, 410008, Hunan, China.
- Cancer Research Institute, Central South University, 110 Xiangya Road, Changsha, 410078, Hunan, China.
- Department of Thoracic Surgery, Second Xiangya Hospital, Central South University, Changsha, China.
| |
Collapse
|
19
|
The Microenvironment in Epstein-Barr Virus-Associated Malignancies. Pathogens 2018; 7:pathogens7020040. [PMID: 29652813 PMCID: PMC6027429 DOI: 10.3390/pathogens7020040] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2018] [Revised: 04/08/2018] [Accepted: 04/11/2018] [Indexed: 12/27/2022] Open
Abstract
The Epstein–Barr virus (EBV) can cause a wide variety of cancers upon infection of different cell types and induces a highly variable composition of the tumor microenvironment (TME). This TME consists of both innate and adaptive immune cells and is not merely an aspecific reaction to the tumor cells. In fact, latent EBV-infected tumor cells utilize several specific mechanisms to form and shape the TME to their own benefit. These mechanisms have been studied largely in the context of EBV+ Hodgkin lymphoma, undifferentiated nasopharyngeal carcinoma, and EBV+ gastric cancer. This review describes the composition, immune escape mechanisms, and tumor cell promoting properties of the TME in these three malignancies. Mechanisms of susceptibility which regularly involve genes related to immune system function are also discussed, as only a small proportion of EBV-infected individuals develops an EBV-associated malignancy.
Collapse
|
20
|
Zhao J, Saunders J, Schussler SD, Rios S, Insaidoo FK, Fridman AL, Li H, Liu YH. Characterization of a novel modification of a CHO-produced mAb: Evidence for the presence of tyrosine sulfation. MAbs 2017; 9:985-995. [PMID: 28590151 DOI: 10.1080/19420862.2017.1332552] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022] Open
Abstract
Herein we describe the investigation of a Chinese hamster ovary (CHO)-expressed human mAb molecule found partially modified by a +80 Da adduct. This mass difference, suggestive of a single sulfation or phosphorylation addition, was observed by mass analysis of the intact and reduced molecule by mass spectrometry (MS). The modification was located on tyrosine 31 (Y31) of the light chain in the complementarity-determining region 1 by liquid chromatography (LC)-MS peptide mapping and electron transfer dissociation fragmentation. The complete loss of the 80 Da modification moiety during collision induced dissociation fragmentation suggested this modification could not be a tyrosine phosphorylation. Treatment of the mAb with alkaline phosphatase confirmed our hypothesis. Western blot experiment using anti-tyrosine sulfation antibody and LC retention time correlation with corresponding synthetic sulfated peptides further confirmed the identification of tyrosine sulfation on the light chain. The unique sequence motif with neighboring acidic amino acids and local secondary structure might play a role to make Y31 a substrate residue for sulfation. This type of modification, to our knowledge, has not been previously reported for CHO-produced human IgG antibodies.
Collapse
Affiliation(s)
- Jia Zhao
- a Protein Mass Spectrometry, Sterile Product and Analytical Development, Bioprocess Development , Merck Research Laboratories , Kenilworth , NJ , USA
| | - Jason Saunders
- b Preclinical Development, New Modalities, Bioprocess Development , Merck Research Laboratories , Kenilworth , NJ , USA
| | - Svetlana Dukleska Schussler
- c Purification Process Development & In-Process Analytical, Bioprocess Development , Merck Research Laboratories , Kenilworth , NJ , USA
| | - Sandra Rios
- c Purification Process Development & In-Process Analytical, Bioprocess Development , Merck Research Laboratories , Kenilworth , NJ , USA
| | - Francis Kobina Insaidoo
- c Purification Process Development & In-Process Analytical, Bioprocess Development , Merck Research Laboratories , Kenilworth , NJ , USA
| | - Aleksandr L Fridman
- a Protein Mass Spectrometry, Sterile Product and Analytical Development, Bioprocess Development , Merck Research Laboratories , Kenilworth , NJ , USA
| | - Huijuan Li
- a Protein Mass Spectrometry, Sterile Product and Analytical Development, Bioprocess Development , Merck Research Laboratories , Kenilworth , NJ , USA
| | - Yan-Hui Liu
- a Protein Mass Spectrometry, Sterile Product and Analytical Development, Bioprocess Development , Merck Research Laboratories , Kenilworth , NJ , USA
| |
Collapse
|
21
|
EBV based cancer prevention and therapy in nasopharyngeal carcinoma. NPJ Precis Oncol 2017; 1:10. [PMID: 29872698 PMCID: PMC5871899 DOI: 10.1038/s41698-017-0018-x] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2016] [Revised: 03/01/2017] [Accepted: 03/03/2017] [Indexed: 12/13/2022] Open
Abstract
Epstein-Barr virus is an important cancer causing virus. Nasopharyngeal carcinoma is an infection-related cancer strongly driven by Epstein-Barr virus. In this cancer model, we identified the major host targets of latent membrane protein 1 which is a driving oncogene encoded by Epstein-Barr virus in latency infection. latent membrane protein 1 activates several oncogenic signaling axes causing multiple malignant phenotypes and therapeutic resistance. Also, Epstein-Barr virus up-regulates DNA methyltransferase 1 and mediates onco-epigenetic effects in the carcinogenesis. The collaborating pathways activated by latent membrane protein 1 constructs an oncogenic signaling network, which makes latent membrane protein 1 an important potential target for effective treatment or preventive intervention. In Epstein-Barr virus lytic phase, the plasma level of Epstein-Barr virus DNA is considered as a distinguishing marker for nasopharyngeal carcinoma in subjects from healthy high-risk populations and is also a novel prognostic marker in Epstein-Barr virus-positive nasopharyngeal carcinoma. Now the early detection and screening of the lytic proteins and Epstein-Barr virus DNA have been applied to clinical and high-risk population. The knowledge generated regarding Epstein-Barr virus can be used in Epstein-Barr virus based precision cancer prevention and therapy in the near future.
Collapse
|
22
|
Arimont M, Sun SL, Leurs R, Smit M, de Esch IJP, de Graaf C. Structural Analysis of Chemokine Receptor-Ligand Interactions. J Med Chem 2017; 60:4735-4779. [PMID: 28165741 PMCID: PMC5483895 DOI: 10.1021/acs.jmedchem.6b01309] [Citation(s) in RCA: 73] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
![]()
This
review focuses on the construction and application of structural chemokine
receptor models for the elucidation of molecular determinants of chemokine
receptor modulation and the structure-based discovery and design of
chemokine receptor ligands. A comparative analysis of ligand binding
pockets in chemokine receptors is presented, including a detailed
description of the CXCR4, CCR2, CCR5, CCR9, and US28 X-ray structures,
and their implication for modeling molecular interactions of chemokine
receptors with small-molecule ligands, peptide ligands, and large
antibodies and chemokines. These studies demonstrate how the integration
of new structural information on chemokine receptors with extensive
structure–activity relationship and site-directed mutagenesis
data facilitates the prediction of the structure of chemokine receptor–ligand
complexes that have not been crystallized. Finally, a review of structure-based
ligand discovery and design studies based on chemokine receptor crystal
structures and homology models illustrates the possibilities and challenges
to find novel ligands for chemokine receptors.
Collapse
Affiliation(s)
- Marta Arimont
- Division of Medicinal Chemistry, Faculty of Sciences, Amsterdam Institute of Molecules, Medicines and Systems (AIMMS), Vrije Universiteit Amsterdam , De Boelelaan 1108, 1081 HZ Amsterdam, The Netherlands
| | - Shan-Liang Sun
- Division of Medicinal Chemistry, Faculty of Sciences, Amsterdam Institute of Molecules, Medicines and Systems (AIMMS), Vrije Universiteit Amsterdam , De Boelelaan 1108, 1081 HZ Amsterdam, The Netherlands
| | - Rob Leurs
- Division of Medicinal Chemistry, Faculty of Sciences, Amsterdam Institute of Molecules, Medicines and Systems (AIMMS), Vrije Universiteit Amsterdam , De Boelelaan 1108, 1081 HZ Amsterdam, The Netherlands
| | - Martine Smit
- Division of Medicinal Chemistry, Faculty of Sciences, Amsterdam Institute of Molecules, Medicines and Systems (AIMMS), Vrije Universiteit Amsterdam , De Boelelaan 1108, 1081 HZ Amsterdam, The Netherlands
| | - Iwan J P de Esch
- Division of Medicinal Chemistry, Faculty of Sciences, Amsterdam Institute of Molecules, Medicines and Systems (AIMMS), Vrije Universiteit Amsterdam , De Boelelaan 1108, 1081 HZ Amsterdam, The Netherlands
| | - Chris de Graaf
- Division of Medicinal Chemistry, Faculty of Sciences, Amsterdam Institute of Molecules, Medicines and Systems (AIMMS), Vrije Universiteit Amsterdam , De Boelelaan 1108, 1081 HZ Amsterdam, The Netherlands
| |
Collapse
|
23
|
Zhou W, Wang Y, Xie J, Geraghty RJ. A fluorescence-based high-throughput assay to identify inhibitors of tyrosylprotein sulfotransferase activity. Biochem Biophys Res Commun 2017; 482:1207-1212. [DOI: 10.1016/j.bbrc.2016.12.013] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2016] [Accepted: 12/02/2016] [Indexed: 12/14/2022]
|
24
|
Banko AV, Lazarevic IB, Folic MM, Djukic VB, Cirkovic AM, Karalic DZ, Cupic MD, Jovanovic TP. Characterization of the Variability of Epstein-Barr Virus Genes in Nasopharyngeal Biopsies: Potential Predictors for Carcinoma Progression. PLoS One 2016; 11:e0153498. [PMID: 27071030 PMCID: PMC4829223 DOI: 10.1371/journal.pone.0153498] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2015] [Accepted: 03/17/2016] [Indexed: 12/15/2022] Open
Abstract
Epstein-Barr virus (EBV) infection is a significant factor in the pathogenesis of nasopharyngeal carcinoma, especially in the undifferentiated carcinoma of nasopharyngeal type (UCNT, World Health Organization type III), which is the dominant histopathological type in high-risk areas. The major EBV oncogene is latent membrane protein 1 (LMP1). LMP1 gene shows variability with different tumorigenic and immunogenic potentials. EBV nuclear antigen 1 (EBNA1) regulates progression of EBV-related tumors; however, the influence of EBNA1 sequence variability on tumor pathogenesis is controversial. The aims of this study were to characterize polymorphisms of EBV genes in non-endemic nasopharyngeal carcinoma biopsies and to investigate potential sequence patterns that correlate with the clinical presentation of nasopharyngeal carcinoma. In total, 116 tumor biopsies of undifferentiated carcinoma of nasopharyngeal type (UCNT), collected from 2008 to 2014, were evaluated in this study. The genes EBNA2, LMP1, and EBNA1 were amplified using nested-PCR. EBNA2 genotyping was performed by visualization of PCR products using gel electrophoresis. Investigation of LMP1 and EBNA1 included sequence, phylogenetic, and statistical analyses. The presence of EBV DNA was significantly distributed between TNM stages. LMP1 variability showed six variants, with the detection of the first China1 and North Carolina variants in European nasopharyngeal carcinoma biopsies. Newly discovered variants Srb1 and Srb2 were UCNT-specific LMP1 polymorphisms. The B95-8 and North Carolina variants are possible predictors for favorable TNM stages. In contrast, deletions in LMP1 are possible risk factors for the most disfavorable TNM stage, independent of EBNA2 or EBNA1 variability. A newly discovered EBNA1 subvariant, P-thr-sv-5, could be a potential diagnostic marker, as it represented a UCNT-specific EBNA1 subvariant. A particular combination of EBNA2, LMP1, and EBNA1 polymorphisms, type 1/Med/P-thr was identified as a possible risk factor for TNM stage IVB or progression to the N3 stage.
Collapse
Affiliation(s)
- Ana V. Banko
- Institute of Microbiology and Immunology, Faculty of Medicine, University of Belgrade, Belgrade, Serbia
- * E-mail:
| | - Ivana B. Lazarevic
- Institute of Microbiology and Immunology, Faculty of Medicine, University of Belgrade, Belgrade, Serbia
| | - Miljan M. Folic
- Clinic of Otorhinolaryngology and Maxillofacial Surgery, Clinical Center of Serbia, Faculty of Medicine, University of Belgrade, Belgrade, Serbia
| | - Vojko B. Djukic
- Clinic of Otorhinolaryngology and Maxillofacial Surgery, Clinical Center of Serbia, Faculty of Medicine, University of Belgrade, Belgrade, Serbia
| | - Andja M. Cirkovic
- Institute for Medical Statistics and Informatics, Faculty of Medicine, University of Belgrade, Belgrade, Serbia
| | - Danijela Z. Karalic
- Institute of Microbiology and Immunology, Faculty of Medicine, University of Belgrade, Belgrade, Serbia
| | - Maja D. Cupic
- Institute of Microbiology and Immunology, Faculty of Medicine, University of Belgrade, Belgrade, Serbia
| | - Tanja P. Jovanovic
- Institute of Microbiology and Immunology, Faculty of Medicine, University of Belgrade, Belgrade, Serbia
| |
Collapse
|
25
|
Elgui de Oliveira D, Müller-Coan BG, Pagano JS. Viral Carcinogenesis Beyond Malignant Transformation: EBV in the Progression of Human Cancers. Trends Microbiol 2016; 24:649-664. [PMID: 27068530 DOI: 10.1016/j.tim.2016.03.008] [Citation(s) in RCA: 76] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2015] [Revised: 03/02/2016] [Accepted: 03/16/2016] [Indexed: 01/31/2023]
Abstract
Cancer progression begins when malignant cells colonize adjacent sites, and it is characterized by increasing tumor heterogeneity, invasion and dissemination of cancer cells. Clinically, progression is the most relevant stage in the natural history of cancers. A given virus is usually regarded as oncogenic because of its ability to induce malignant transformation of cells. Nonetheless, oncogenic viruses may also be important for the progression of infection-associated cancers. Recently this hypothesis has been addressed because of studies on the contribution of the Epstein-Barr virus (EBV) to the aggressiveness of nasopharyngeal carcinoma (NPC). Several EBV products modulate cancer progression phenomena, such as the epithelial-mesenchymal transition, cell motility, invasiveness, angiogenesis, and metastasis. In this regard, there are compelling data about the effects of EBV latent membrane proteins (LMPs) and EBV nuclear antigens (EBNAs), as well as nontranslated viral RNAs, such as the EBV-encoded small nonpolyadenylated RNAs (EBERs) and viral microRNAs, notably EBV miR-BARTs. The available data on the mechanisms and players involved in the contribution of EBV infection to the aggressiveness of NPC are discussed in this review. Overall, this conceptual framework may be valuable for the understanding of the contribution of some infectious agents in the progression of cancers.
Collapse
Affiliation(s)
- Deilson Elgui de Oliveira
- Viral Carcinogenesis and Cancer Biology Research Group (ViriCan) at Biotechnology Institute (IBTEC), São Paulo State University (UNESP), Brazil; Pathology Department at Botucatu Medical School, São Paulo State University (UNESP), Brazil.
| | - Bárbara G Müller-Coan
- Viral Carcinogenesis and Cancer Biology Research Group (ViriCan) at Biotechnology Institute (IBTEC), São Paulo State University (UNESP), Brazil
| | - Joseph S Pagano
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, NC, USA
| |
Collapse
|
26
|
Qiao N, Wang L, Wang T, Li H. Inflammatory CXCL12-CXCR4/CXCR7 axis mediates G-protein signaling pathway to influence the invasion and migration of nasopharyngeal carcinoma cells. Tumour Biol 2015; 37:8169-79. [PMID: 26715277 DOI: 10.1007/s13277-015-4686-2] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2015] [Accepted: 12/16/2015] [Indexed: 12/28/2022] Open
Abstract
This study explored whether the migration, invasion, and apoptosis of nasopharyngeal carcinoma (NPC) cells were affected by the CXCR4/CXCR7-CXCL12 axis and if this mechanism was related to G-protein signaling pathway. A total of 72 NPC patients admitted in our hospital between April 2013 and February 2015 were incorporated in this study. Immunohistochemistry was performed to compare the expression levels of CXCR4, CXCR7, and CXCL12 between NPC tissues and adjacent normal tissues. Then, the correlation analysis was implemented to assess the association among CXCR4, CXCR7, and CXCL12 expressions. Jellyfish glow protein experiment was carried out after the cultivation of CNE-2Z cell lines in order to observe the intracellular calcium mobilization resulted from G-protein activation contributed by CXCR4/CXCR7-CXCL12 axis. The impact of CXCR4/CXCR7-CXCL12 axis on the migration and invasion of NPC cells was explored using transwell experiments. Finally, the anti-apoptosis effects of CXCR4/CXCR7-CXCL12 axis on NPC cells were investigated by the splicing of poly ADP-ribose polymerase (PARP). Compared to NPC patients with low-grade (stage I-II) tumor node metastasis (TNM) and those without lymph node metastasis, the expression of CXCR4, CXCR7, and CXCL12 were significantly higher in NPC patients with high-grade (stage III-IV) TNM and those with lymph node metastasis (P < 0.05). Moreover, there was significant positive correlation between the expression level of CXCL12 and CXCR7 (r s = 0.484, P < 0.001) as well as the expression level of CXCL12 and CXCR4 (r s = 0.414, P < 0.001). As suggested by cellular experiments using CNE-2Z, the calcium mobilization degree induced by CXCR4-CXCL12 axis in activating G proteins seemed to be slightly more effective than that induced by CXCR4/CXCR7-CXCL12 axis, while the CXCR7-CXCL12 axis could hardly activate calcium mobilization. Furthermore, the transwell experiment showed that CXCR4/CXCR7-CXCL12 axis could exacerbate the migration and invasion of NPC cells (P < 0.05). The transwell experiment also suggested that the CXCR4/CXCR7-CXCL12 axis was associated with the expression of matrix metallo proteinase 9 (MMP9) which is a substance in the downstream of G-protein pathways (P < 0.05). Results from PARP shear zone also indicated that the CXCR4/CXCR7-CXCL12 axis could suppress NPC cell apoptosis (P < 0.05). The expressional levels of CXCR4, CXCR7, and CXCL12 significantly varied with clinical stages and status of lymph node metastasis of NPC patients. This revealed potential indicators which can be used for NPC prognosis. Additionally, the CXCR4/CXCR7-CXCL12 axis may regulate the expression of downstream proteins (e.g., MMP-9) through the activation of G-protein signaling pathways. These conclusions may provide key evidence for NPC aetiology which can be further investigated to develop novel molecular targets for NPC treatments.
Collapse
Affiliation(s)
- Naian Qiao
- Department of Radiation Oncology, Qilu Hospital, Shandong University, Jinan, 250012, Shandong Province, China
| | - Lin Wang
- Department of Radiation Oncology, Qilu Hospital, Shandong University, Jinan, 250012, Shandong Province, China
| | - Tao Wang
- Department of Radiation Oncology, Qilu Hospital, Shandong University, Jinan, 250012, Shandong Province, China
| | - Haiying Li
- Department of Ultrasonography, Qilu Hospital, Shandong University, No. 107 West Wenhua Road, Lixia District, Jinan, 250012, Shandong Province, China.
| |
Collapse
|
27
|
JIANG ZHIBIN, ZHU JIALIANG, MA YUCHAO, HONG CAO, XIAO SHENG, JIN LONGYU. Tyrosylprotein sulfotransferase 1 expression is negatively correlated with c-Met and lymph node metastasis in human lung cancer. Mol Med Rep 2015; 12:5217-22. [DOI: 10.3892/mmr.2015.4096] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2014] [Accepted: 06/23/2015] [Indexed: 11/06/2022] Open
|
28
|
Yang YS, Wang CC, Chen BH, Hou YH, Hung KS, Mao YC. Tyrosine sulfation as a protein post-translational modification. Molecules 2015; 20:2138-64. [PMID: 25635379 PMCID: PMC6272617 DOI: 10.3390/molecules20022138] [Citation(s) in RCA: 92] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2014] [Revised: 01/06/2015] [Accepted: 01/14/2015] [Indexed: 12/17/2022] Open
Abstract
Integration of inorganic sulfate into biological molecules plays an important role in biological systems and is directly involved in the instigation of diseases. Protein tyrosine sulfation (PTS) is a common post-translational modification that was first reported in the literature fifty years ago. However, the significance of PTS under physiological conditions and its link to diseases have just begun to be appreciated in recent years. PTS is catalyzed by tyrosylprotein sulfotransferase (TPST) through transfer of an activated sulfate from 3'-phosphoadenosine-5'-phosphosulfate to tyrosine in a variety of proteins and peptides. Currently, only a small fraction of sulfated proteins is known and the understanding of the biological sulfation mechanisms is still in progress. In this review, we give an introductory and selective brief review of PTS and then summarize the basic biochemical information including the activity and the preparation of TPST, methods for the determination of PTS, and kinetics and reaction mechanism of TPST. This information is fundamental for the further exploration of the function of PTS that induces protein-protein interactions and the subsequent biochemical and physiological reactions.
Collapse
Affiliation(s)
- Yuh-Shyong Yang
- Department of Biological Science and Technology, National Chiao Tung University, 75 Po-Ai Street, Hsinchu 30068, Taiwan.
| | - Chen-Chu Wang
- Department of Biological Science and Technology, National Chiao Tung University, 75 Po-Ai Street, Hsinchu 30068, Taiwan.
| | - Bo-Han Chen
- Department of Biological Science and Technology, National Chiao Tung University, 75 Po-Ai Street, Hsinchu 30068, Taiwan.
| | - You-Hua Hou
- Department of Biological Science and Technology, National Chiao Tung University, 75 Po-Ai Street, Hsinchu 30068, Taiwan.
| | - Kuo-Sheng Hung
- Department of Neurosurgery, Center of Excellence for Clinical Trial and Research, Taipei Medical University-Wan Fang Medical Center, Taipei 11696, Taiwan.
| | - Yi-Chih Mao
- Department of Biological Science and Technology, National Chiao Tung University, 75 Po-Ai Street, Hsinchu 30068, Taiwan.
| |
Collapse
|
29
|
Tan G, Tang X, Huang D, Li Y, Liu N, Peng Z, Zhang Z, Duan C, Lu J, Yan G, Tang F. Dinitrosopiperazine-mediated phosphorylated-proteins are involved in nasopharyngeal carcinoma metastasis. Int J Mol Sci 2014; 15:20054-71. [PMID: 25375189 PMCID: PMC4264155 DOI: 10.3390/ijms151120054] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2014] [Revised: 09/12/2014] [Accepted: 10/21/2014] [Indexed: 12/11/2022] Open
Abstract
N,N'-dinitrosopiperazine (DNP) with organ specificity for nasopharyngeal epithelium, is involved in nasopharyngeal carcinoma (NPC) metastasis, though its mechanism is unclear. To reveal the pathogenesis of DNP-induced metastasis, immunoprecipitation was used to identify DNP-mediated phosphoproteins. DNP-mediated NPC cell line (6-10B) motility and invasion was confirmed. Twenty-six phosphoproteins were increased at least 1.5-fold following DNP exposure. Changes in the expression levels of selected phosphoproteins were verified by Western-blotting analysis. DNP treatment altered the phosphorylation of ezrin (threonine 567), vimentin (serine 55), stathmin (serine 25) and STAT3 (serine 727). Furthermore, it was shown that DNP-dependent metastasis is mediated in part through ezrin at threonine 567, as DNP-mediated metastasis was decreased when threonine 567 of ezrin was mutated. Strikingly, NPC metastatic tumors exhibited a higher expression of phosphorylated-ezrin at threonine 567 than the primary tumors. These findings provide novel insight into DNP-induced NPC metastasis and may contribute to a better understanding of the metastatic mechanisms of NPC tumors.
Collapse
Affiliation(s)
- Gongjun Tan
- Medical Research Center and Clinical Laboratory, Zhuhai Hospital of Jinan University, 79 Kangning Road, Zhuhai 519000, China.
| | - Xiaowei Tang
- Metallurgical Science and Engineering, Central South University, 21 Lushan South Road, Changsha 410083, China.
| | - Damao Huang
- Clinical Laboratory and Medical Research Center, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha 410008, China.
| | - Yuejin Li
- Medical Research Center and Clinical Laboratory, Zhuhai Hospital of Jinan University, 79 Kangning Road, Zhuhai 519000, China.
| | - Na Liu
- Clinical Laboratory and Medical Research Center, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha 410008, China.
| | - Zhengke Peng
- Medical Research Center and Clinical Laboratory, Zhuhai Hospital of Jinan University, 79 Kangning Road, Zhuhai 519000, China.
| | - Zhenlin Zhang
- Medical Research Center and Clinical Laboratory, Zhuhai Hospital of Jinan University, 79 Kangning Road, Zhuhai 519000, China.
| | - Chaojun Duan
- Clinical Laboratory and Medical Research Center, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha 410008, China.
| | - Jinping Lu
- Medical Research Center and Clinical Laboratory, Zhuhai Hospital of Jinan University, 79 Kangning Road, Zhuhai 519000, China.
| | - Guangrong Yan
- Institute of Life and Health Engineering, National Engineering and Research Center for Genetic Medicine, Jinan University, 601 Huangpu Road West, Guangzhou 510632, China.
| | - Faqing Tang
- Medical Research Center and Clinical Laboratory, Zhuhai Hospital of Jinan University, 79 Kangning Road, Zhuhai 519000, China.
| |
Collapse
|
30
|
Ou H, Li Y, Kang M. Activation of miR-21 by STAT3 induces proliferation and suppresses apoptosis in nasopharyngeal carcinoma by targeting PTEN gene. PLoS One 2014; 9:e109929. [PMID: 25365510 PMCID: PMC4217720 DOI: 10.1371/journal.pone.0109929] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2014] [Accepted: 09/12/2014] [Indexed: 01/08/2023] Open
Abstract
The present study is to investigate the role of microRNA-21 (miR-21) in nasopharyngeal carcinoma (NPC) and the mechanisms of regulation of PTEN by miR-21. Fifty-four tissue samples were collected from 42 patients with NPC and 12 healthy controls. Human NPC cell lines CNE-1, CNE-2, TWO3 and C666-1 were used for cell assays. To investigate the expression of miR-21, RT-PCR was employed. RT-PCR, Western blotting, and immunohistochemistry were used to measure the expression of STAT3 mRNA and STAT3 protein. To test the effect of miR-21 on the cell growth and apoptosis of NPC cells in vitro, transfection of CNE1 and CNE2 cell lines and flow cytometry were performed. TUNEL assay was used to detect DNA fragmentation. To validate whether miR-21 directly recognizes the 3'-UTRs of PTEN mRNA, luciferase reporter assay was employed. miR-21 expression was increased in NPC tissues compared with control and the same result was found in NPC cell lines. Notably, increased expression of miR-21 was directly related to advanced clinical stage and lymph node metastasis. STAT3, a transcription factor activated by IL-6, directly activated miR-21 in transformed NPC cell lines. Furthermore, miR-21 markedly inhibited PTEN tumor suppressor, leading to increased AKT activity. Both in vitro and in vivo assays revealed that miR-21 enhanced NPC cell proliferation and suppressed apoptosis. miR-21, activated by STAT3, induced proliferation and suppressed apoptosis in NPC by targeting PTEN-AKT pathway.
Collapse
Affiliation(s)
- Hesheng Ou
- College of Pharmacy, Guangxi Medical University, Nanning City, Guangxi Province, P.R. China
| | - Yumei Li
- College of Pharmacy, Guangxi Medical University, Nanning City, Guangxi Province, P.R. China
| | - Min Kang
- The First Affiliated Hospital, Guangxi Medical University, Nanning City, Guangxi Province, P.R. China
- * E-mail:
| |
Collapse
|
31
|
Genetic variation in microRNA-binding site and prognosis of patients with colorectal cancer. J Cancer Res Clin Oncol 2014; 141:35-41. [PMID: 25079514 DOI: 10.1007/s00432-014-1780-6] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2014] [Accepted: 07/10/2014] [Indexed: 12/20/2022]
Abstract
BACKGROUND Single nucleotide polymorphisms (SNPs) located in the 3'-UTR of miRNA target genes could affect miRNA-mediated gene regulation, thereby contributing to the susceptibility or prognosis of cancer. Accordingly, the present study analyzed SNPs located at putative miRNA-binding sites of the 3'-UTR of various genes and investigated their impact on the prognosis for patients with colorectal cancer. MATERIALS AND METHODS In total, 831 consecutive patients (discovery cohort, n = 309; validation cohort, n = 522) with curatively resected colorectal adenocarcinoma were enrolled. Plus, 157 SNPs were selected from an in silico analysis based on several miRNA and HapMap databases. The SNP genotyping was performed using a Sequenom MassARRAY. A luciferase assay was used to investigate whether miR-571 modulated PAUF gene expression when rs12373 was included in the PAUF 3'UTR region. RESULTS In the discovery cohort, 18 SNPs were identified as possible prognostic biomarkers in a survival analysis. In the validation cohort, two SNPs (TPST1 rs3757417T>G and PAUF rs12373A>C) were significantly associated with prognosis in the same direction as the discovery cohort when adjusted for age, preoperative carcinoembryonic antigen level, and pathologic stage (discovery + validation cohort; TPST1 rs3757417T>G, disease-free survival (DFS), p value = 0.0004, overall survival (OS), p value = 0.01 in recessive model; PAUF rs12373A>C, DFS, p value = <0.0001, OS, p value = 0.0008 in dominant model). A significantly lower Renilla activity was observed in the rs12373 CC construct when compared with the rs12373 AA construct (p = 0.002). CONCLUSION The current study provides evidence that the TPST1 rs3757417T>G and PAUF rs12373A>C polymorphisms are possible prognostic biomarkers for patients with colorectal cancer.
Collapse
|
32
|
Arnolds KL, Spencer JV. CXCR4: a virus's best friend? INFECTION GENETICS AND EVOLUTION 2014; 25:146-56. [PMID: 24793563 DOI: 10.1016/j.meegid.2014.04.018] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/23/2013] [Revised: 04/21/2014] [Accepted: 04/22/2014] [Indexed: 10/25/2022]
Abstract
Viruses are dependent on their hosts for replication and dispersal in the environment; thus, the most successful viruses are those that co-evolve with their hosts. CXCR4 is a cellular chemokine receptor that plays central roles in development, hematopoiesis, and immune surveillance through signaling induced by its ligand, CXCL12. The CXCR4-CXCL12 axis has been besieged by many pathogens that employ a range of strategies to modify or exploit CXCR4 activity. While CXCR4 was identified as a critical co-factor for entry of HIV into CD4+ T cells early on, other viruses may utilize CXCR4 to gain cell entry as well. Moreover, several viruses have been found to modulate CXCR4 expression or alter its functional activity, with direct effects on cell trafficking, immune responses, cell proliferation, and cell survival. Because CXCR4 is targeted by a diverse group of viral pathogens, modification of host CXCR4 signaling activity is emerging as a common theme in virus persistence and is likely to be important for subversion of the host immune system. This review highlights major viral pathogens that use and abuse CXCR4 and explores the possible reasons why this chemokine receptor has become "a virus's best friend".
Collapse
Affiliation(s)
- Kathleen L Arnolds
- Department of Biology, University of San Francisco, 2130 Fulton Street, San Francisco, CA 94403, United States
| | - Juliet V Spencer
- Department of Biology, University of San Francisco, 2130 Fulton Street, San Francisco, CA 94403, United States.
| |
Collapse
|
33
|
Polymorphisms of Toll-like receptor 9 are associated with nasopharyngeal carcinoma susceptibility. Tumour Biol 2014; 35:3247-53. [DOI: 10.1007/s13277-013-1424-5] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2013] [Accepted: 11/13/2013] [Indexed: 12/12/2022] Open
|