1
|
Kim YS. Gastric Carcinoma. Curr Top Microbiol Immunol 2025. [PMID: 40423781 DOI: 10.1007/82_2025_303] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/28/2025]
Abstract
Epstein-Barr virus (EBV)-associated gastric cancers (EBVaGCs) account for about 10% of gastric cancers globally, with higher prevalence in East Asia and Latin America. These cancers develop through a "gastritis-infection-cancer sequence" and are characterized by unique molecular signatures, including CpG island methylator phenotype and mutations in ARID1A and PIK3CA genes. EBVaGCs typically present in the proximal stomach with diffuse-type histology and dense lymphocytic infiltration. Key viral proteins EBNA1 and LMP2A drive oncogenesis by altering cellular processes and immune responses. The IFN-γ signature and extensive epigenetic modifications contribute to their distinct profile. Despite often presenting at advanced stages, EBVaGCs generally have a more favorable prognosis. EBV employs sophisticated strategies to evade immune detection, utilizing latent proteins and noncoding RNAs. Paradoxically, despite an immune-hot environment, EBVaGCs demonstrate effective immune evasion, partly due to the expression of immune checkpoint molecules like PD-L1 and LAG3. Treatment approaches vary based on disease stage, from endoscopic resection for early-stage cancers to systemic therapies for advanced cases. Immunotherapy, particularly PD-1/PD-L1 inhibitors, shows promising results. Emerging research suggests combining these with LAG3 inhibitors may enhance efficacy. Ongoing research and advanced genomic techniques continue to reveal new insights, paving the way for personalized therapies and novel diagnostic approaches.
Collapse
Affiliation(s)
- Young-Sik Kim
- Department of Pathology, Korea University Ansan Hospital, Korea University College of Medicine, Seoul, Republic of Korea.
| |
Collapse
|
2
|
Zhou YQ, Jiang JX, He S, Li YQ, Cheng XX, Liu SQ, Wei PP, Guan XY, Ong CK, Wang VYF, Luo CL, Bei JX. Epstein-Barr virus hijacks histone demethylase machinery to drive epithelial malignancy progression through KDM5B upregulation. Signal Transduct Target Ther 2025; 10:83. [PMID: 40059116 PMCID: PMC11891327 DOI: 10.1038/s41392-025-02163-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2024] [Revised: 01/19/2025] [Accepted: 02/01/2025] [Indexed: 05/13/2025] Open
Abstract
Epstein-Barr virus (EBV) is a significant epigenetic driver in the development of epithelial-origin nasopharyngeal carcinoma (NPC) and gastric cancer (GC), which together represent 80% of EBV-associated malignancies. Despite its known association, the specific mechanisms, particularly those involving EBV-induced histone modifications, remain poorly understood. Through integrative analyses of single-cell and bulk transcriptome data from epithelial tumor tissues and EBV-infected cells, we identified KDM5B as a critical histone-modifying factor consistently upregulated following EBV infection. We demonstrated that EBV stimulates KDM5B expression via interactions of its latent gene EBNA1 with transcription factor CEBPB and through direct binding of its lytic gene BZLF1 to Zta-response elements on the KDM5B promoter. Functional assays revealed that KDM5B acts as an oncogene, correlating with poor survival outcomes in EBV-associated epithelial cancers. Mechanistically, KDM5B inhibited the tumor suppressor gene PLK2 through histone demethylation, thereby activating the PI3K/AKT/mTOR signaling pathway and promoting malignant progression. Furthermore, treatment with the KDM5B inhibitor AS-8351 markedly attenuated this signaling activity and exhibited strong anti-tumor effect in both in vitro and in vivo patient-derived xenograft models from EBV-associated tumors. Together, these findings provide novel insights into how EBV hijacks KDM5B to mediate histone demethylation of PLK2, facilitating tumor progression through the PI3K/AKT/mTOR pathway in epithelial cancers, highlighting promising therapeutic strategies targeting epigenetic alterations in EBV-associated cancers.
Collapse
Affiliation(s)
- Ya-Qing Zhou
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, 510060, P.R. China
- Department of Clinical Laboratory, Sun Yat-sen University Cancer Center, Guangzhou, P.R. China
| | - Jia-Xin Jiang
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, 510060, P.R. China
- Department of Experimental Research, Sun Yat-sen University Cancer Center, Guangzhou, P.R. China
| | - Shuai He
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, 510060, P.R. China
- Department of Experimental Research, Sun Yat-sen University Cancer Center, Guangzhou, P.R. China
| | - Yi-Qi Li
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, 510060, P.R. China
- Department of Experimental Research, Sun Yat-sen University Cancer Center, Guangzhou, P.R. China
| | - Xi-Xi Cheng
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, 510060, P.R. China
- Department of Experimental Research, Sun Yat-sen University Cancer Center, Guangzhou, P.R. China
| | - Shu-Qiang Liu
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, 510060, P.R. China
- Department of Experimental Research, Sun Yat-sen University Cancer Center, Guangzhou, P.R. China
| | - Pan-Pan Wei
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, 510060, P.R. China
- Department of Experimental Research, Sun Yat-sen University Cancer Center, Guangzhou, P.R. China
| | - Xin-Yuan Guan
- Department of Clinical Oncology, The University of Hong Kong, Hong Kong, P.R. China
| | - Choon Kiat Ong
- Lymphoma Translational Research Laboratory, Division of Cellular and Molecular Research, National Cancer Centre Singapore, Singapore, Singapore
- Cancer and Stem Cell Biology Program, Duke-NUS Medical School, Singapore, Singapore
| | - Vivien Ya-Fan Wang
- Faculty of Health, University of Macau, Avenida da Universidade, Taipa, Macau SAR, P.R. China
| | - Chun-Ling Luo
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, 510060, P.R. China.
- Department of Experimental Research, Sun Yat-sen University Cancer Center, Guangzhou, P.R. China.
| | - Jin-Xin Bei
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, 510060, P.R. China.
- Department of Experimental Research, Sun Yat-sen University Cancer Center, Guangzhou, P.R. China.
- Department of Medical Oncology, National Cancer Centre of Singapore, Singapore, Singapore.
- Sun Yat-sen University Institute of Advanced Studies Hong Kong, Science Park, Hong Kong SAR, P.R. China.
- Department of Clinical Oncology, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, University of Hong Kong, Hong Kong SAR, P.R. China.
| |
Collapse
|
3
|
Wang H, Hu J, Zhou W, Qian A. Metabolic reprogramming in the pathogenesis and progression of nasopharyngeal carcinoma: molecular mechanisms and therapeutic implications. Am J Cancer Res 2024; 14:4049-4064. [PMID: 39267663 PMCID: PMC11387871 DOI: 10.62347/vyat9271] [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: 06/14/2024] [Accepted: 08/23/2024] [Indexed: 09/15/2024] Open
Abstract
Nasopharyngeal carcinoma (NPC) is a unique head and neck cancer with a complex etiology involving genetic predispositions, environmental factors, and Epstein-Barr virus (EBV) infection. Despite progress in radiotherapy and chemotherapy, the prognosis for advanced NPC is still unfavorable, prompting the need for innovative therapeutic approaches. Metabolic reprogramming plays a crucial role in the development and progression of NPC, marked by substantial changes in glycolysis, lipid, and amino acid metabolism. These alterations aid tumor cell proliferation, survival under stress, and immune evasion, with features such as enhanced aerobic glycolysis (Warburg effect) and shifts in lipid and amino acid pathways. Oncogenic drivers like MYC, RAS, EGFR, and the loss of tumor suppressors such as TP53 and PTEN, along with key signaling pathways including mTOR, AMPK, and HIF-1α, orchestrate these metabolic changes. This review discusses the molecular mechanisms of metabolic reprogramming in NPC and outlines potential therapeutic targets within these pathways. Advances in metabolic imaging and biomarker discovery are also enhancing the precision of diagnostics and treatment monitoring, fostering personalized medicine in NPC treatment. This manuscript aims to provide a detailed overview of the current research and its implications for improving NPC management and patient outcomes through targeted metabolic therapies.
Collapse
Affiliation(s)
- Hongli Wang
- Department of Otolaryngology, The Affiliated People's Hospital of Ningbo University Ningbo, Zhejiang, China
| | - Jiandao Hu
- Department of Otolaryngology, The Affiliated People's Hospital of Ningbo University Ningbo, Zhejiang, China
| | - Weibang Zhou
- Department of Otolaryngology, The Affiliated People's Hospital of Ningbo University Ningbo, Zhejiang, China
| | - Aijuan Qian
- Department of Otolaryngology, The Affiliated People's Hospital of Ningbo University Ningbo, Zhejiang, China
| |
Collapse
|
4
|
Maroui MA, Odongo GA, Mundo L, Manara F, Mure F, Fusil F, Jay A, Gheit T, Michailidis TM, Ferrara D, Leoncini L, Murray P, Manet E, Ohlmann T, De Boevre M, De Saeger S, Cosset FL, Lazzi S, Accardi R, Herceg Z, Gruffat H, Khoueiry R. Aflatoxin B1 and Epstein-Barr virus-induced CCL22 expression stimulates B cell infection. Proc Natl Acad Sci U S A 2024; 121:e2314426121. [PMID: 38574017 PMCID: PMC11032484 DOI: 10.1073/pnas.2314426121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2023] [Accepted: 02/20/2024] [Indexed: 04/06/2024] Open
Abstract
Epstein-Barr Virus (EBV) infects more than 90% of the adult population worldwide. EBV infection is associated with Burkitt lymphoma (BL) though alone is not sufficient to induce carcinogenesis implying the involvement of co-factors. BL is endemic in African regions faced with mycotoxins exposure. Exposure to mycotoxins and oncogenic viruses has been shown to increase cancer risks partly through the deregulation of the immune response. A recent transcriptome profiling of B cells exposed to aflatoxin B1 (AFB1) revealed an upregulation of the Chemokine ligand 22 (CCL22) expression although the underlying mechanisms were not investigated. Here, we tested whether mycotoxins and EBV exposure may together contribute to endemic BL (eBL) carcinogenesis via immunomodulatory mechanisms involving CCL22. Our results revealed that B cells exposure to AFB1 and EBV synergistically stimulated CCL22 secretion via the activation of Nuclear Factor-kappa B pathway. By expressing EBV latent genes in B cells, we revealed that elevated levels of CCL22 result not only from the expression of the latent membrane protein LMP1 as previously reported but also from the expression of other viral latent genes. Importantly, CCL22 overexpression resulting from AFB1-exposure in vitro increased EBV infection through the activation of phosphoinositide-3-kinase pathway. Moreover, inhibiting CCL22 in vitro and in humanized mice in vivo limited EBV infection and decreased viral genes expression, supporting the notion that CCL22 overexpression plays an important role in B cell infection. These findings unravel new mechanisms that may underpin eBL development and identify novel pathways that can be targeted in drug development.
Collapse
Affiliation(s)
- Mohamed Ali Maroui
- Centre International de Recherche en Infectiologie, University Claude Bernard Lyon I, INSERM U1111, CNRS UMR5308, Ecole Normale Supérieure, Lyon69366 Cedex 07, France
| | - Grace Akinyi Odongo
- Epigenomics and Mechanisms Branch, International Agency for Research on Cancer, World Health Organization, Lyon69366 Cedex 07, France
| | - Lucia Mundo
- Limerick Digital Cancer Research Centre, Health Research Institute, Bernal Institute and School of Medicine, University of Limerick, LimerickV94 T9PX, Ireland
- Department of Medical Biotechnology, Section of Pathology, University of Siena, Siena53100, Italy
| | - Francesca Manara
- Epigenomics and Mechanisms Branch, International Agency for Research on Cancer, World Health Organization, Lyon69366 Cedex 07, France
| | - Fabrice Mure
- Centre International de Recherche en Infectiologie, University Claude Bernard Lyon I, INSERM U1111, CNRS UMR5308, Ecole Normale Supérieure, Lyon69366 Cedex 07, France
| | - Floriane Fusil
- Centre International de Recherche en Infectiologie, University Claude Bernard Lyon I, INSERM U1111, CNRS UMR5308, Ecole Normale Supérieure, Lyon69366 Cedex 07, France
| | - Antonin Jay
- Epigenomics and Mechanisms Branch, International Agency for Research on Cancer, World Health Organization, Lyon69366 Cedex 07, France
| | - Tarik Gheit
- Epigenomics and Mechanisms Branch, International Agency for Research on Cancer, World Health Organization, Lyon69366 Cedex 07, France
| | - Thanos M. Michailidis
- Centre of Excellence in Mycotoxicology and Public Health, Department of Bioanalysis, Faculty of Pharmaceutical Sciences, Ghent University, Ghent9000, Belgium
| | - Domenico Ferrara
- Department of Medical Biotechnology, Section of Pathology, University of Siena, Siena53100, Italy
| | - Lorenzo Leoncini
- Department of Medical Biotechnology, Section of Pathology, University of Siena, Siena53100, Italy
| | - Paul Murray
- Limerick Digital Cancer Research Centre, Health Research Institute, Bernal Institute and School of Medicine, University of Limerick, LimerickV94 T9PX, Ireland
| | - Evelyne Manet
- Centre International de Recherche en Infectiologie, University Claude Bernard Lyon I, INSERM U1111, CNRS UMR5308, Ecole Normale Supérieure, Lyon69366 Cedex 07, France
| | - Théophile Ohlmann
- Centre International de Recherche en Infectiologie, University Claude Bernard Lyon I, INSERM U1111, CNRS UMR5308, Ecole Normale Supérieure, Lyon69366 Cedex 07, France
| | - Marthe De Boevre
- Centre of Excellence in Mycotoxicology and Public Health, Department of Bioanalysis, Faculty of Pharmaceutical Sciences, Ghent University, Ghent9000, Belgium
| | - Sarah De Saeger
- Centre of Excellence in Mycotoxicology and Public Health, Department of Bioanalysis, Faculty of Pharmaceutical Sciences, Ghent University, Ghent9000, Belgium
- Department of Biotechnology and Food Technology, Faculty of Science, University of Johannesburg, Gauteng2028, South Africa
| | - François-Loïc Cosset
- Centre International de Recherche en Infectiologie, University Claude Bernard Lyon I, INSERM U1111, CNRS UMR5308, Ecole Normale Supérieure, Lyon69366 Cedex 07, France
| | - Stefano Lazzi
- Department of Medical Biotechnology, Section of Pathology, University of Siena, Siena53100, Italy
| | - Rosita Accardi
- Epigenomics and Mechanisms Branch, International Agency for Research on Cancer, World Health Organization, Lyon69366 Cedex 07, France
| | - Zdenko Herceg
- Epigenomics and Mechanisms Branch, International Agency for Research on Cancer, World Health Organization, Lyon69366 Cedex 07, France
| | - Henri Gruffat
- Centre International de Recherche en Infectiologie, University Claude Bernard Lyon I, INSERM U1111, CNRS UMR5308, Ecole Normale Supérieure, Lyon69366 Cedex 07, France
| | - Rita Khoueiry
- Epigenomics and Mechanisms Branch, International Agency for Research on Cancer, World Health Organization, Lyon69366 Cedex 07, France
| |
Collapse
|
5
|
Yuan C, Chang S, Zhang C, Dong D, Ding J, Mahdavian AR, Hu Z, Sun L, Tan S. Post cross-linked ROS-responsive poly(β-amino ester)-plasmid polyplex NPs for gene therapy of EBV-associated nasopharyngeal carcinoma. J Mater Chem B 2024; 12:3129-3143. [PMID: 38451208 DOI: 10.1039/d3tb02926c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/08/2024]
Abstract
Nasopharyngeal carcinoma (NPC) is one of the most common tumors in South China and Southeast Asia and is thought to be associated with Epstein-Barr virus (EBV) infection. Downregulation of latent membrane protein 1 (LMP1) encoded by EBV can reduce the expression of NF-κB and PI3K, induce apoptosis, and inhibit the growth of EBV-related NPC. For targeted cleavage of the Lmp1 oncogene via the CRISPR/Cas9 gene editing system, a post cross-linked ROS-responsive poly(β-amino ester) (PBAE) polymeric vector was developed for the delivery of CRISPR/Cas9 plasmids both in vitro and in vivo. After composition optimization, the resultant polymer-plasmid polyplex nanoparticles (NPs) showed a diameter of ∼230 nm and a zeta potential of 22.3 mV with good stability. Compared with the non-cross-linked system, the cross-linked NPs exhibited efficient and quick cell uptake, higher transfection efficiency in EBV-positive C666-1 cells (53.5% vs. 40.6%), more efficient gene editing ability against the Mucin2 model gene (Muc2) (17.9% vs. 15.4%) and Lmp1 (8.5% vs. 5.6%), and lower intracellular reactive oxygen species (ROS) levels. The NPs achieved good tumor penetration and tumor growth inhibition in the C666-1 xenograft tumor model via Lmp1 cleavage, indicating their potential for gene therapy of EBV-related NPC.
Collapse
Affiliation(s)
- Caiyan Yuan
- School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China.
- The First Hospital of Nanchang, Nanchang 330008, China
| | - Shuangyan Chang
- Department of Obstetrics and Gynecology, Zhongnan Hospital of Wuhan University, Donghu 169th Road, Wuchang District, Wuhan 430062, Hubei, China.
| | - Chong Zhang
- School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China.
| | - Dirong Dong
- Department of Obstetrics and Gynecology, Zhongnan Hospital of Wuhan University, Donghu 169th Road, Wuchang District, Wuhan 430062, Hubei, China.
| | - Jiahui Ding
- School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China.
| | - Ali Reza Mahdavian
- Polymer Science Department, Iran Polymer and Petrochemical Institute, Tehran 14967, Iran
| | - Zheng Hu
- Department of Obstetrics and Gynecology, Zhongnan Hospital of Wuhan University, Donghu 169th Road, Wuchang District, Wuhan 430062, Hubei, China.
| | - Lili Sun
- Department of Obstetrics and Gynecology, Zhongnan Hospital of Wuhan University, Donghu 169th Road, Wuchang District, Wuhan 430062, Hubei, China.
| | - Songwei Tan
- School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China.
| |
Collapse
|
6
|
Cao Y, Wen H, Leng C, Feng S. MiR-29a mediates the apoptotic effects of TNF-α on endothelial cells through inhibiting PI3K/AKT/BCL-2 axis. J Biochem Mol Toxicol 2024; 38:e23598. [PMID: 38047396 DOI: 10.1002/jbt.23598] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Revised: 09/04/2023] [Accepted: 11/20/2023] [Indexed: 12/05/2023]
Abstract
Endothelial cell apoptosis driven by inflammation (TNF-α) plays a critical role in the pathogenesis of atherosclerosis, but the exact molecular mechanisms are not clearly elucidated. MicroRNA (miR)-29 families (a/b/c) take important roles in pathophysiological processes of atherosclerosis, also the underlying mechanisms have not been fully clarified. The aims are to explore whether or not miR-29 families mediate the apoptotic effects of TNF-α on endothelial cells and uncover the underlying molecular mechanisms. In this study, MTT assay and flow cytometer analysis were employed respectively to determine the proliferation and apoptosis of human umbilical vascular endothelial cells (HUVECs) under TNF-α exposure. Real-time quantitative PCR and western blot were performed to detect the levels of target RNAs and proteins/their phosphorylation in HUVECs. TNF-α could inhibit HUVEC proliferation and induce HUVEC apoptosis in a positive dose- and time-dependent manner, with a similar way of miR-29a upregulation, but no effects on miR-29b/c. Upregulation of miR-29a with its mimics enhanced the apoptotic effect of TNF-α on HUVECs, but downregulation of miR-29a using anti-miR-29a blocked up its apoptotic effect. MiR-29a inhibited the expression of PI3Kp85α and Bcl-2 and blocked up the signal transduction of PI3K/AKT/Bcl-2 axis to mediate the apoptotic effect of TNF-α on HUVECs. Mediating the inflammation-driven endothelial cell apoptosis is an important biology mechanism by which miR-29a promotes atherosclerosis and its complications. MiR-29a will be a potential diagnostic and therapeutic target for atherosclerotic cardiovascular diseases; it is worthwhile to further study.
Collapse
Affiliation(s)
- Yunchang Cao
- Department of Molecular Biology, School of Intelligent Medicine and Biotechnology, Guilin Medical University, Guilin, China
| | - Hongbo Wen
- Department of Biochemistry and Molecular Biology, Hengyang Medicine School, University of South China, Hengyang, China
| | - Chaoqun Leng
- Department of Biochemistry and Molecular Biology, Hengyang Medicine School, University of South China, Hengyang, China
| | - Shaolong Feng
- The Guangxi Key Laboratory of Environmental Exposomics and Entire Lifecycle Heath, School of Public Health, Guilin Medical University, Guilin, China
| |
Collapse
|
7
|
Lv M, Ding Y, Zhang Y, Liu S. Targeting EBV-encoded products: Implications for drug development in EBV-associated diseases. Rev Med Virol 2024; 34:e2487. [PMID: 37905912 DOI: 10.1002/rmv.2487] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2023] [Revised: 10/02/2023] [Accepted: 10/22/2023] [Indexed: 11/02/2023]
Abstract
Epstein-Barr virus, a human gamma-herpesvirus, has a close connection to the pathogenesis of cancers and other diseases, which are a burden for public health worldwide. So far, several drugs or biomolecules have been discovered that can target EBV-encoded products for treatment, such as Silvestrol, affinity toxin, roscovitine, H20, H31, curcumin, thymoquinone, and ribosomal protein L22. These drugs activate or inhibit the function of some biomolecules, affecting subsequent signalling pathways by acting on the products of EBV. These drugs usually target LMP1, LMP2; EBNA1, EBNA2, EBNA3; EBER1, EBER2; Bam-HI A rightward transcript and BHRF1. Additionally, some promising findings in the fields of vaccines, immunological, and cellular therapies have been established. In this review, we mainly summarise the function of drugs mentioned above and unique mechanisms, hoping that we can help giving insight to the design of drugs for the treatment of EBV-associated diseases.
Collapse
Affiliation(s)
- Mengwen Lv
- Department of Blood Transfusion, The Affiliated Hospital of Qingdao University, Qingdao, China
- Department of Pathogenic Biology, Qingdao University Medical College, Qingdao, China
| | - Yuan Ding
- Department of Special Examination, Qingdao Women & Children Hospital, Qingdao, China
| | - Yan Zhang
- Department of Pathogenic Biology, Qingdao University Medical College, Qingdao, China
- Department of Clinical Laboratory, Zibo Central Hospital, Zibo, China
| | - Shuzhen Liu
- Department of Blood Transfusion, The Affiliated Hospital of Qingdao University, Qingdao, China
| |
Collapse
|
8
|
Khenchouche A, Salem-Bekhit MM, Mansour AA, Alomary MN, Wang X, Alzahrani HA, Hosiny IMA, Taha EI, Shazly GA, Benguerba Y, Houali K. Suppression of Nasopharyngeal and Gastric Tumor Growth in a Mouse Model by Antibodies to Epstein-Barr Virus LMP1 Protein. Microorganisms 2023; 11:1712. [PMID: 37512884 PMCID: PMC10383785 DOI: 10.3390/microorganisms11071712] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Accepted: 06/21/2023] [Indexed: 07/30/2023] Open
Abstract
The study aimed to investigate the antitumor efficacy of anti-LMP1 antibodies in EBV-positive nasopharyngeal and stomach cell lines and xenograft models. The study also examined the NF-κB expression and cell cycle activation of NPC-serum-exosome-associated LMP1. Anti-LMP1 antibody treatment before or during cell implantation prevented tumor growth in nude mice. A small dose of antibodies resulted in complete tumor regression for at least three months after the tumors had grown in size. The consumption of antigen-antibody complexes by tumor cells limited tumor growth. In vitro experiments showed that anti-LMP1 antibodies killed EBV-positive NPC- or GC-derived epithelial cell lines and EBV-positive human B-cell lines but not EBV-negative cell lines. Treatment with anti-LMP1 reduced NF-κB expression in cells. The animal model experiments showed that anti-LMP1 inhibited and prevented NPC- or GC-derived tumor growth. The results suggest that LMP1 antibody immunotherapy could cure nasopharyngeal cancer, EBV-positive gastric carcinoma, and EBV-associated lymphomas. However, further validation of these findings is required through human clinical trials.
Collapse
Affiliation(s)
- Abdelhalim Khenchouche
- Département de Microbiologie, Faculté des Sciences de la Nature et de la Vie, Université Ferhat Abbas Sétif 1, Sétif 19000, Algeria
- Laboratoire de Virologie Moléculaire, FRE3011, CNRS, Faculté de Médecine Laennec, Université Claude Bernard Lyon-1, 69008 Lyon, France
| | - Mounir M Salem-Bekhit
- Department of Pharmaceutics, College of Pharmacy, King Saud University, P.O. Box 2457, Riyadh 11451, Saudi Arabia
| | - Ahd A Mansour
- Medical Laboratory Science Department, Fakeeh College for Medical Sciences, P.O. Box 2537, Jeddah 21461, Saudi Arabia
| | - Mohammad N Alomary
- Advanced Diagnostic and Therapeutic Institute, King Abdulaziz City for Science and Technology, P.O. Box 6086, Riyadh 11442, Saudi Arabia
| | - Xiaohui Wang
- Laboratoire de Virologie Moléculaire, FRE3011, CNRS, Faculté de Médecine Laennec, Université Claude Bernard Lyon-1, 69008 Lyon, France
| | - Hayat Ali Alzahrani
- Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, Northern Border University, Arar 73211, Saudi Arabia
| | - Ibrahim M Al Hosiny
- Microbiology and Immunology Department, Faculty of Medicine, Al-Azhar University, Cairo 11651, Egypt
| | - Ehab I Taha
- Department of Pharmaceutics, College of Pharmacy, King Saud University, P.O. Box 2457, Riyadh 11451, Saudi Arabia
| | - Gamal A Shazly
- Department of Pharmaceutics, College of Pharmacy, King Saud University, P.O. Box 2457, Riyadh 11451, Saudi Arabia
| | - Yacine Benguerba
- Laboratoire de Biopharmacie Et Pharmacotechnie (LPBT), Ferhat Abbas Setif 1 University, Setif 19000, Algeria
| | - Karim Houali
- Laboratoire de Virologie Moléculaire, FRE3011, CNRS, Faculté de Médecine Laennec, Université Claude Bernard Lyon-1, 69008 Lyon, France
- Laboratoire de Biochimie Analytique et Biotechnologie (LABAB), Faculté des Sciences Biologiques et des Sciences Agronomiques, Université Mouloud Mammeri, Tizi-Ouzou 15000, Algeria
| |
Collapse
|
9
|
Kashyap D, Rele S, Bagde PH, Saini V, Chatterjee D, Jain AK, Pandey RK, Jha HC. Comprehensive insight into altered host cell-signaling cascades upon Helicobacter pylori and Epstein-Barr virus infections in cancer. Arch Microbiol 2023; 205:262. [PMID: 37310490 DOI: 10.1007/s00203-023-03598-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Revised: 05/22/2023] [Accepted: 05/23/2023] [Indexed: 06/14/2023]
Abstract
Cancer is characterized by mutagenic events that lead to disrupted cell signaling and cellular functions. It is one of the leading causes of death worldwide. Literature suggests that pathogens, mainly Helicobacter pylori and Epstein-Barr virus (EBV), have been associated with the etiology of human cancer. Notably, their co-infection may lead to gastric cancer. Pathogen-mediated DNA damage could be the first and crucial step in the carcinogenesis process that modulates numerous cellular signaling pathways. Altogether, it dysregulates the metabolic pathways linked with cell growth, apoptosis, and DNA repair. Modulation in these pathways leads to abnormal growth and proliferation. Several signaling pathways such RTK, RAS/MAPK, PI3K/Akt, NFκB, JAK/STAT, HIF1α, and Wnt/β-catenin are known to be altered in cancer. Therefore, this review focuses on the oncogenic roles of H. pylori, EBV, and its associated signaling cascades in various cancers. Scrutinizing these signaling pathways is crucial and may provide new insights and targets for preventing and treating H. pylori and EBV-associated cancers.
Collapse
Affiliation(s)
- Dharmendra Kashyap
- Lab No. POD 1B 602, Infection Bio-Engineering Group, Department of Biosciences and Biomedical Engineering, Indian Institute of Technology Indore, Indore, Madhya Pradesh, 453552, India
| | - Samiksha Rele
- Lab No. POD 1B 602, Infection Bio-Engineering Group, Department of Biosciences and Biomedical Engineering, Indian Institute of Technology Indore, Indore, Madhya Pradesh, 453552, India
| | - Pranit Hemant Bagde
- Lab No. POD 1B 602, Infection Bio-Engineering Group, Department of Biosciences and Biomedical Engineering, Indian Institute of Technology Indore, Indore, Madhya Pradesh, 453552, India
| | - Vaishali Saini
- Lab No. POD 1B 602, Infection Bio-Engineering Group, Department of Biosciences and Biomedical Engineering, Indian Institute of Technology Indore, Indore, Madhya Pradesh, 453552, India
| | | | | | - Rajan Kumar Pandey
- Department of Medical Biochemistry and Biophysics, Karolinska Institute, 17177, Solna, Sweden
| | - Hem Chandra Jha
- Lab No. POD 1B 602, Infection Bio-Engineering Group, Department of Biosciences and Biomedical Engineering, Indian Institute of Technology Indore, Indore, Madhya Pradesh, 453552, India.
- Centre for Rural Development and Technology, Indian Institute of Technology Indore, Madhya Pradesh, 453552, Indore, India.
| |
Collapse
|
10
|
Wang C, Huang C, Wang J, Ye J, Xue Z, Zhang J, Ren Y. Ginsenoside Rg5 attenuates hypoxia-induced cardiomyocyte apoptosis via regulating the Akt pathway. Chem Biol Drug Des 2023; 101:1348-1355. [PMID: 36762503 DOI: 10.1111/cbdd.14217] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Revised: 12/16/2022] [Accepted: 02/02/2023] [Indexed: 02/11/2023]
Abstract
Ginsenoside Rg5 has been implicated in a variety of diseases. However, it is unknown whether Ginsenoside Rg5 can protect against hypoxia-induced neonatal rat cardiomyocytes (NRMs). The purpose of this study was to look into the effect of Ginsenoside Rg5 on hypoxia-induced NRMs apoptosis as well as the underlying molecular mechanism. In this study, following isolation and culture of ventricular myocardial cells from neonatal rats, the appropriate concentration of Rg5 was determined using the MTT assay, the effect of Rg5 on apoptosis was assessed employing TUNEL staining and flow cytometry assays. Levels of apoptosis-related proteins and phosphorylated level of Akt (ser 473 and ser 308) were analyzed using the western blot analysis. Finally, the experimental results shown that Ginsenoside Rg5 significantly inhibited hypoxia-induced NRMs apoptosis, decreased the expression pro-apoptotic protein Bax, increased the expression of anti-apoptotic protein Bcl-2 ratio and the level of cleaved caspase 3. Akt signaling activation was found to be the mechanism of Ginsenoside Rg5s protective effect on hypoxia-induced NRMs apoptosis, as an Akt inhibitor eliminated the anti-apoptotic effects of Ginsenoside Rg5. Various analyses were performed and verified, ginsenoside Rg5 suppressed hypoxia-induced apoptosis in NRMs via activation of the Akt signaling.
Collapse
Affiliation(s)
- Chenxi Wang
- Cardiovascular medicine department, The Second People's Hospital of Kunshan, Suzhou, China
| | - Chenyang Huang
- Endocrine department, The Second People's Hospital of Kunshan, Suzhou, China
| | - Jiali Wang
- Cardiovascular medicine department, The Second People's Hospital of Kunshan, Suzhou, China
| | - Jianfeng Ye
- Cardiovascular medicine department, The Second People's Hospital of Kunshan, Suzhou, China
| | - Zhiqiang Xue
- Cardiovascular medicine department, The Second People's Hospital of Kunshan, Suzhou, China
| | - Jian Zhang
- Cardiovascular medicine department, The Second People's Hospital of Kunshan, Suzhou, China
| | - Yuke Ren
- Cardiovascular medicine department, Suzhou Hospital of traditional Chinese Medicine Affiliated to Nanjing University of traditional Chinese Medicine, Suzhou, China
| |
Collapse
|
11
|
Protein Kinase CK2 and Epstein-Barr Virus. Biomedicines 2023; 11:biomedicines11020358. [PMID: 36830895 PMCID: PMC9953236 DOI: 10.3390/biomedicines11020358] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Revised: 01/20/2023] [Accepted: 01/24/2023] [Indexed: 01/28/2023] Open
Abstract
Protein kinase CK2 is a pleiotropic protein kinase, which phosphorylates a number of cellular and viral proteins. Thereby, this kinase is implicated in the regulation of cellular signaling, controlling of cell proliferation, apoptosis, angiogenesis, immune response, migration and invasion. In general, viruses use host signaling mechanisms for the replication of their genome as well as for cell transformation leading to cancer. Therefore, it is not surprising that CK2 also plays a role in controlling viral infection and the generation of cancer cells. Epstein-Barr virus (EBV) lytically infects epithelial cells of the oropharynx and B cells. These latently infected B cells subsequently become resting memory B cells when passing the germinal center. Importantly, EBV is responsible for the generation of tumors such as Burkitt's lymphoma. EBV was one of the first human viruses, which was connected to CK2 in the early nineties of the last century. The present review shows that protein kinase CK2 phosphorylates EBV encoded proteins as well as cellular proteins, which are implicated in the lytic and persistent infection and in EBV-induced neoplastic transformation. EBV-encoded and CK2-phosphorylated proteins together with CK2-phosphorylated cellular signaling proteins have the potential to provide efficient virus replication and cell transformation. Since there are powerful inhibitors known for CK2 kinase activity, CK2 might become an attractive target for the inhibition of EBV replication and cell transformation.
Collapse
|
12
|
Awasthi P, Dwivedi M, Kumar D, Hasan S. Insights into intricacies of the Latent Membrane Protein-1 (LMP-1) in EBV-associated cancers. Life Sci 2023; 313:121261. [PMID: 36493876 DOI: 10.1016/j.lfs.2022.121261] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Revised: 11/29/2022] [Accepted: 11/29/2022] [Indexed: 12/12/2022]
Abstract
Numerous lymphomas, carcinomas, and other disorders have been associated with Epstein-Barr Virus (EBV) infection. EBV's carcinogenic potential can be correlated to latent membrane protein 1 (LMP1), which is essential for fibroblast and primary lymphocyte transformation. LMP1, a transmembrane protein with constitutive activity, belongs to the tumour necrosis factor receptor (TNFR) superfamily. LMP1 performs number of role in the life cycle of EBV and the pathogenesis by interfering with, reprogramming, and influencing a vast range of host cellular activities and functions that are getting well-known but still poorly understood. LMP1, pleiotropically perturbs, reprograms and balances a wide range of various processes of cell such as extracellular vesicles, epigenetics, ubiquitin machinery, metabolism, cell proliferation and survival, and also promotes oncogenic transformation, angiogenesis, anchorage-independent cell growth, metastasis and invasion, tumour microenvironment. By the help of various experiments, it is proven that EBV-encoded LMP1 activates multiple cell signalling pathways which affect antigen presentation, cell-cell interactions, chemokine and cytokine production. Therefore, it is assumed that LMP1 may perform majorly in EBV associated malignancies. For the development of novel techniques toward targeted therapeutic applications, it is essential to have a complete understanding of the LMP1 signalling landscape in order to identify potential targets. The focus of this review is on LMP1-interacting proteins and related signalling processes. We further discuss tactics for using the LMP1 protein as a potential therapeutic for cancers caused by the EBV.
Collapse
Affiliation(s)
- Prankur Awasthi
- Amity Institute of Biotechnology, Amity University Uttar Pradesh, Lucknow 226028, India
| | - Manish Dwivedi
- Amity Institute of Biotechnology, Amity University Uttar Pradesh, Lucknow 226028, India
| | - Dhruv Kumar
- School of Health Sciences and Technology, UPES University Dehradun, Uttarakhand, India
| | - Saba Hasan
- Amity Institute of Biotechnology, Amity University Uttar Pradesh, Lucknow 226028, India.
| |
Collapse
|
13
|
Kim B, Kim KM. Role of Exosomes and Their Potential as Biomarkers in Epstein-Barr Virus-Associated Gastric Cancer. Cancers (Basel) 2023; 15:cancers15020469. [PMID: 36672418 PMCID: PMC9856651 DOI: 10.3390/cancers15020469] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Revised: 01/05/2023] [Accepted: 01/10/2023] [Indexed: 01/15/2023] Open
Abstract
Exosomes are a subtype of extracellular vesicles ranging from 30 to 150 nm and comprising many cellular components, including DNA, RNA, proteins, and metabolites, encapsulated in a lipid bilayer. Exosomes are secreted by many cell types and play important roles in intercellular communication in cancer. Viruses can hijack the exosomal pathway to regulate viral propagation, cellular immunity, and the microenvironment. Cells infected with Epstein-Barr virus (EBV), one of the most common oncogenic viruses, have also been found to actively secrete exosomes, and studies on their roles in EBV-related malignancies are ongoing. In this review, we focus on the role of exosomes in EBV-associated gastric cancer and their clinical applicability in diagnosis and treatment.
Collapse
Affiliation(s)
- Binnari Kim
- Department of Pathology, Ulsan University Hospital, University of Ulsan College of Medicine, Ulsan 44610, Republic of Korea
| | - Kyoung-Mee Kim
- Department of Pathology and Translational Genomics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul 06351, Republic of Korea
- Center of Companion Diagnostics, Samsung Medical Center, Seoul 06351, Republic of Korea
- Correspondence: ; Tel.: +82-2-3410-2807; Fax: +82-2-3410-6396
| |
Collapse
|
14
|
Patrad E, Khalighfard S, Amiriani T, Khori V, Alizadeh AM. Molecular mechanisms underlying the action of carcinogens in gastric cancer with a glimpse into targeted therapy. Cell Oncol 2022; 45:1073-1117. [PMID: 36149600 DOI: 10.1007/s13402-022-00715-3] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/03/2022] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Gastric cancer imposes a substantial global health burden despite its overall incidence decrease. A broad spectrum of inherited, environmental and infectious factors contributes to the development of gastric cancer. A profound understanding of the molecular underpinnings of gastric cancer has lagged compared to several other tumors with similar incidence and morbidity rates, owing to our limited knowledge of the role of carcinogens in this malignancy. The International Agency for Research on Cancer (IARC) has classified gastric carcinogenic agents into four groups based on scientific evidence from human and experimental animal studies. This review aims to explore the potential comprehensive molecular and biological impacts of carcinogens on gastric cancer development and their interactions and interferences with various cellular signaling pathways. CONCLUSIONS In this review, we highlight recent clinical trial data reported in the literature dealing with different ways to target various carcinogens in gastric cancer. Moreover, we touch upon other multidisciplinary therapeutic approaches such as surgery, adjuvant and neoadjuvant chemotherapy. Rational clinical trials focusing on identifying suitable patient populations are imperative to the success of single-agent therapeutics. Novel insights regarding signaling pathways that regulate gastric cancer can potentially improve treatment responses to targeted therapy alone or in combination with other/conventional treatments. Preventive strategies such as control of H. pylori infection through eradication or immunization as well as dietary habit and lifestyle changes may reduce the incidence of this multifactorial disease, especially in high prevalence areas. Further in-depth understanding of the molecular mechanisms involved in the role of carcinogenic agents in gastric cancer development may offer valuable information and update state-of-the-art resources for physicians and researchers to explore novel ways to combat this disease, from bench to bedside. A schematic outlining of the interaction between gastric carcinogenic agents and intracellular pathways in gastric cancer H. pylori stimulates multiple intracellular pathways, including PI3K/AKT, NF-κB, Wnt, Shh, Ras/Raf, c-MET, and JAK/STAT, leading to epithelial cell proliferation and differentiation, apoptosis, survival, motility, and inflammatory cytokine release. EBV can stimulate intracellular pathways such as the PI3K/Akt, RAS/RAF, JAK/STAT, Notch, TGF-β, and NF-κB, leading to cell survival and motility, proliferation, invasion, metastasis, and the transcription of anti-apoptotic genes and pro-inflammatory cytokines. Nicotine and alcohol can lead to angiogenesis, metastasis, survival, proliferation, pro-inflammatory, migration, and chemotactic by stimulating various intracellular signaling pathways such as PI3K/AKT, NF-κB, Ras/Raf, ROS, and JAK/STAT. Processed meat contains numerous carcinogenic compounds that affect multiple intracellular pathways such as sGC/cGMP, p38 MAPK, ERK, and PI3K/AKT, leading to anti-apoptosis, angiogenesis, metastasis, inflammatory responses, proliferation, and invasion. Lead compounds may interact with multiple signaling pathways such as PI3K/AKT, NF-κB, Ras/Raf, DNA methylation-dependent, and epigenetic-dependent, leading to tumorigenesis, carcinogenesis, malignancy, angiogenesis, DNA hypermethylation, cell survival, and cell proliferation. Stimulating signaling pathways such as PI3K/Akt, RAS/RAF, JAK/STAT, WNT, TGF-β, EGF, FGFR2, and E-cadherin through UV ionizing radiation leads to cell survival, proliferation, and immortalization in gastric cancer. The consequence of PI3K/AKT, NF-κB, Ras/Raf, ROS, JAK/STAT, and WNT signaling stimulation by the carcinogenic component of Pickled vegetables and salted fish is the Warburg effect, tumorigenesis, angiogenesis, proliferation, inflammatory response, and migration.
Collapse
Affiliation(s)
- Elham Patrad
- Cancer Research Center, Cancer Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Solmaz Khalighfard
- Cancer Research Center, Cancer Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Taghi Amiriani
- Ischemic Disorders Research Center, Golestan University of Medical Sciences, Gorgan, Iran
| | - Vahid Khori
- Ischemic Disorders Research Center, Golestan University of Medical Sciences, Gorgan, Iran
| | - Ali Mohammad Alizadeh
- Cancer Research Center, Cancer Institute, Tehran University of Medical Sciences, Tehran, Iran.
- Breast Disease Research Center, Cancer Institute, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
15
|
Shechter O, Sausen DG, Gallo ES, Dahari H, Borenstein R. Epstein-Barr Virus (EBV) Epithelial Associated Malignancies: Exploring Pathologies and Current Treatments. Int J Mol Sci 2022; 23:14389. [PMID: 36430864 PMCID: PMC9699474 DOI: 10.3390/ijms232214389] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2022] [Revised: 11/06/2022] [Accepted: 11/14/2022] [Indexed: 11/22/2022] Open
Abstract
Epstein-Barr virus (EBV) is one of eight known herpesviruses with the potential to infect humans. Globally, it is estimated that between 90-95% of the population has been infected with EBV. EBV is an oncogenic virus that has been strongly linked to various epithelial malignancies such as nasopharyngeal and gastric cancer. Recent evidence suggests a link between EBV and breast cancer. Additionally, there are other, rarer cancers with weaker evidence linking them to EBV. In this review, we discuss the currently known epithelial malignancies associated with EBV. Additionally, we discuss and establish which treatments and therapies are most recommended for each cancer associated with EBV.
Collapse
Affiliation(s)
- Oren Shechter
- Department of Microbiology and Molecular Cell Biology, Eastern Virginia Medical School, Norfolk, VA 23501, USA
| | - Daniel G. Sausen
- Department of Microbiology and Molecular Cell Biology, Eastern Virginia Medical School, Norfolk, VA 23501, USA
| | - Elisa S. Gallo
- Tel-Aviv Sourasky Medical Center, Division of Dermatology, Tel-Aviv 6423906, Israel
| | - Harel Dahari
- The Program for Experimental and Theoretical Modeling, Division of Hepatology, Department of Medicine, Stritch School of Medicine, Loyola University Chicago, Maywood, IL 60153, USA
| | - Ronen Borenstein
- The Program for Experimental and Theoretical Modeling, Division of Hepatology, Department of Medicine, Stritch School of Medicine, Loyola University Chicago, Maywood, IL 60153, USA
| |
Collapse
|
16
|
Abstract
The severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is the pathogen responsible for the coronavirus disease 2019 (COVID-19) pandemic. Of particular interest for this topic are the signaling cascades that regulate cell survival and death, two opposite cell programs whose control is hijacked by viral infections. The AKT and the Unfolded Protein Response (UPR) pathways, which maintain cell homeostasis by regulating these two programs, have been shown to be deregulated during SARS-CoVs infection as well as in the development of cancer, one of the most important comorbidities in relation to COVID-19. Recent evidence revealed two way crosstalk mechanisms between the AKT and the UPR pathways, suggesting that they might constitute a unified homeostatic control system. Here, we review the role of the AKT and UPR pathways and their interaction in relation to SARS-CoV-2 infection as well as in tumor onset and progression. Feedback regulation between AKT and UPR pathways emerges as a master control mechanism of cell decision making in terms of survival or death and therefore represents a key potential target for developing treatments for both viral infection and cancer. In particular, drug repositioning, the investigation of existing drugs for new therapeutic purposes, could significantly reduce time and costs compared to de novo drug discovery.
Collapse
|
17
|
Sang W, Tu D, Zhang M, Qin Y, Yin W, Song X, Sun C, Yan D, Wang X, Zeng L, Li Z, Xu K, Xu L. l-Asparaginase synergizes with etoposide via the PI3K/Akt/mTOR pathway in Epstein-Barr virus-positive Burkitt lymphoma. J Biochem Mol Toxicol 2022; 36:e23117. [PMID: 35757978 DOI: 10.1002/jbt.23117] [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: 09/24/2021] [Revised: 04/24/2022] [Accepted: 05/18/2022] [Indexed: 11/09/2022]
Abstract
Burkitt lymphoma (BL) is an aggressive Epstein-Barr virus (EBV)-driven B-cell lymphoma characterized by the translocation and rearrangement of the c-Myc proto-oncogene. High-intensity multidrug chemotherapy regimens have a limited effect on the survival of refractory or relapsed BL patients, mainly owing to the high EBV load and drug resistance. l-asparaginase ( l-Asp) and etoposide (VP-16) play a beneficial role in EBV-related lymphoproliferative diseases; however, their roles and mechanisms in BL remain unclear. In this study, we found that VP-16 inhibited BL cell proliferation and arrested the cell cycle at the G2 /M phase. It also induced autophagy and activated the extrinsic and intrinsic apoptotic signaling pathways in BL cells. Mechanistically, VP-16 inhibited c-Myc expression and regulated the PI3K/Akt/mTOR signaling pathway. Notably, VP-16 also showed a specific synergistic effect with l-Asp to induce apoptosis in EBV-positive BL cells but not in EBV-negative BL cells. VP-16 combined with l-Asp further inhibited c-Myc expression and downregulated the PI3K/Akt/mTOR signaling pathway. Additionally, we found that VP-16 inhibited the expression of latent membrane protein 1 (LMP1), and in combination with l-Asp further decreased LMP1 expression in Raji cells. Our in vivo data also showed that the dual-drug combination significantly inhibited the growth of BL tumors and prolonged the survival of mice compared to VP-16 alone. In conclusion, this study provides new evidence that l-Asp may enhance the antitumor effect of VP-16 by inhibiting the PI3K/Akt/mTOR signaling pathway in EBV-positive BL cells.
Collapse
Affiliation(s)
- Wei Sang
- Blood Diseases Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China.,Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China.,Key Laboratory of Bone Marrow Stem Cell, Xuzhou, Jiangsu, China
| | - Dongyun Tu
- Blood Diseases Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China.,Key Laboratory of Bone Marrow Stem Cell, Xuzhou, Jiangsu, China.,Department of Cardiology, Yancheng TCM Hospital Affiliated to Nanjing University of Chinese Medicine, Yancheng, Jiangsu, China
| | - Meng Zhang
- Blood Diseases Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China.,Key Laboratory of Bone Marrow Stem Cell, Xuzhou, Jiangsu, China
| | - Yuanyuan Qin
- Blood Diseases Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China.,Key Laboratory of Bone Marrow Stem Cell, Xuzhou, Jiangsu, China
| | - Wenjing Yin
- Blood Diseases Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China.,Key Laboratory of Bone Marrow Stem Cell, Xuzhou, Jiangsu, China
| | - Xuguang Song
- Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Cai Sun
- Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Dongmei Yan
- Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Xiangmin Wang
- Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Lingyu Zeng
- Blood Diseases Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China.,Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China.,Key Laboratory of Bone Marrow Stem Cell, Xuzhou, Jiangsu, China
| | - Zhenyu Li
- Blood Diseases Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China.,Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China.,Key Laboratory of Bone Marrow Stem Cell, Xuzhou, Jiangsu, China
| | - Kailin Xu
- Blood Diseases Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China.,Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China.,Key Laboratory of Bone Marrow Stem Cell, Xuzhou, Jiangsu, China
| | - Linyan Xu
- Blood Diseases Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China.,Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China.,Key Laboratory of Bone Marrow Stem Cell, Xuzhou, Jiangsu, China
| |
Collapse
|
18
|
Din SRU, Nisar MA, Ramzan MN, Saleem MZ, Ghayas H, Ahmad B, Batool S, Kifayat K, Guo X, Huang M, Zhong M. Latcripin-7A from Lentinula edodes C 91-3 induces apoptosis, autophagy, and cell cycle arrest at G1 phase in human gastric cancer cells via inhibiting PI3K/Akt/mTOR signaling. Eur J Pharmacol 2021; 907:174305. [PMID: 34224698 DOI: 10.1016/j.ejphar.2021.174305] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Revised: 06/29/2021] [Accepted: 07/01/2021] [Indexed: 11/28/2022]
Abstract
Gastric cancer (G.C) is one of the most lethal cancer types worldwide. Current treatment requires surgery along with chemotherapy, which causes obstacles for speedy recovery. The discovery of novel drugs is needed for better treatment of G.C with minimum side effects. Latcripin-7A (LP-7A) is a newly discovered peptide extracted from Lentinula edodes. It is recently studied for its anti-cancer activity. In this study, LP-7A was modeled using a phyre2 server. Anti-proliferation effects of LP-7A on G.C cells were examined via CCK-8, colony formation, and morphology assay. Apoptosis of LP-7A treated G.C cells was evaluated via Hoechst Stain, western blot and flow cytometry. Autophagy was assessed via acridine orange staining and western blot. The cell cycle was assessed via flow cytometry assay and western blot. Pathway was studied via western blot and STRING database. Anti-migratory effects of LP-7A treated G.C cells were analyzed via wound healing, western blot, and migration and invasion assay. LP-7A effectively inhibited the growth of G.C cells by inhibiting the PI3K/Akt/mTOR pathway. G.C cells treated with LP-7A arrested the cell cycle at the G1 phase, contributing to the inhibition of migration and invasion. Furthermore, LP-7A induced apoptosis and autophagy in gastric cancer cells. These results indicated that LP-7A is a promising anti-cancer agent. It affected the proliferation and growth of G.C cells (SGC-7901 and BGC-823) by inducing apoptosis, autophagy, and inhibiting cell cycle at the G1 phase in G.C cells.
Collapse
Affiliation(s)
- Syed Riaz Ud Din
- Department of Microbiology, College of Basic Medical Sciences, Dalian Medical University, Dalian, PR China.
| | - Muhammad Azhar Nisar
- Department of Biochemistry and Molecular Biology, College of Basic Medical Sciences, Dalian Medical University, Dalian, PR China.
| | - Muhammad Noman Ramzan
- Department of Biochemistry and Molecular Biology, College of Basic Medical Sciences, Dalian Medical University, Dalian, PR China.
| | - Muhammad Zubair Saleem
- Department of Pathology and Pathophysiology, College of Basic Medical Sciences, Dalian Medical University, Dalian, PR China; School of Pharmacy, Fujian Provincial Key Laboratory of Natural Medicine Pharmacology, Fujian Medical University, Fuzhou, 350122, PR China.
| | - Hassan Ghayas
- Institute of Cancer Stem Cell, Dalian Medical University, Dalian, PR China.
| | - Bashir Ahmad
- Department of Pathology and Pathophysiology, College of Basic Medical Sciences, Dalian Medical University, Dalian, PR China; Department of Biology, The University of Haripur, Pakistan.
| | - Samana Batool
- Department of Microbiology, College of Basic Medical Sciences, Dalian Medical University, Dalian, PR China; Department of Microbiology and Molecular Genetics, University of Okara, 56300, Pakistan.
| | - Kashif Kifayat
- Department of Biochemistry and Molecular Biology, College of Basic Medical Sciences, Dalian Medical University, Dalian, PR China.
| | - Xiaorong Guo
- Department of Microbiology, College of Basic Medical Sciences, Dalian Medical University, Dalian, PR China.
| | - Min Huang
- Department of Microbiology, College of Basic Medical Sciences, Dalian Medical University, Dalian, PR China.
| | - Mintao Zhong
- Department of Microbiology, College of Basic Medical Sciences, Dalian Medical University, Dalian, PR China.
| |
Collapse
|
19
|
EBNA1 inhibitors have potent and selective antitumor activity in xenograft models of Epstein-Barr virus-associated gastric cancer. Gastric Cancer 2021; 24:1076-1088. [PMID: 33929613 PMCID: PMC8338878 DOI: 10.1007/s10120-021-01193-6] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Accepted: 04/20/2021] [Indexed: 02/07/2023]
Abstract
BACKGROUND AND AIMS Epstein-Barr virus (EBV)-associated gastric carcinoma (EBVaGC) is the most common EBV-associated cancer and accounts for ~ 10% of all gastric cancers (GC). Epstein-Barr virus nuclear antigen 1 (EBNA1), which is critical for the replication and maintenance of the EBV latent genome, is consistently expressed in all EBVaGC tumors. We previously developed small molecule inhibitors of EBNA1. In this study, we investigated the efficacy and selectivity of an EBNA1 inhibitor in cell-based and animal xenograft models of EBV-positive and EBV-negative gastric carcinoma. METHODS We tested the potency of an EBNA1 inhibitor, VK-1727, in vitro and in xenograft studies, using EBV-positive (SNU719 and YCCEL1) and EBV-negative (AGS and MKN74) GC cell lines. After treatment, we analyzed cell viability, proliferation, and RNA expression of EBV genes by RT-qPCR. RESULTS Treatment with VK-1727 selectively inhibits cell cycle progression and proliferation in vitro. In animal studies, treatment with an EBNA1 inhibitor resulted in a significant dose-dependent decrease in tumor growth in EBVaGC xenograft models, but not in EBV-negative GC xenograft studies. Gene expression analysis revealed that short term treatment in cell culture tended towards viral gene activation, while long-term treatment in animal xenografts showed a significant decrease in viral gene expression. CONCLUSIONS EBNA1 inhibitors are potent and selective inhibitors of cell growth in tissue culture and animal models of EBV-positive GC. Long-term treatment with EBNA1 inhibitors may lead to loss of EBV in mouse xenografts. These results suggest that pharmacological targeting of EBNA1 may be an effective strategy to treat patients with EBVaGC.
Collapse
|
20
|
Jiang Y, Ding Y, Liu S, Luo B. The role of Epstein–Barr virus-encoded latent membrane proteins in host immune escape. Future Virol 2021. [DOI: 10.2217/fvl-2020-0320] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Epstein–Barr virus (EBV) is a type IV herpesvirus that widely infects the vast majority of adults, and establishes a latent infection pattern in host cells to escape the clearance of immune system. The virus is intimately associated with the occurrence and progression of lymphomas and epithelial cell cancers. EBV latent membrane proteins (LMPs) can assist its immune escape by downregulating host immune response. Besides EBV, LMPs have important effects on the functions of exosomes and autophagy, which also help EBV to escape immune surveillance. These escape mechanisms may provide conditions for further development of EBV-associated tumors. In this article, we discussed the potential functions of EBV-encoded LMPs in promoting immune escape.
Collapse
Affiliation(s)
- Yuanyuan Jiang
- Department of Medical Affairs of The Affiliated Hospital of Qingdao University, No. 1677 Wutaishan Road, Qingdao, 266000, China
- Department of Pathogenic Biology, Qingdao University Medical College, 308 Ningxia Road, Qingdao, 266021, China
| | - Yuan Ding
- Department of Special Examination, Qingdao Women & Children Hospital, Qingdao, 266035, China
| | - Shuzhen Liu
- Department of Medical Affairs of The Affiliated Hospital of Qingdao University, No. 1677 Wutaishan Road, Qingdao, 266000, China
| | - Bing Luo
- Department of Pathogenic Biology, Qingdao University Medical College, 308 Ningxia Road, Qingdao, 266021, China
| |
Collapse
|
21
|
Cirone M. Cancer cells dysregulate PI3K/AKT/mTOR pathway activation to ensure their survival and proliferation: mimicking them is a smart strategy of gammaherpesviruses. Crit Rev Biochem Mol Biol 2021; 56:500-509. [PMID: 34130564 DOI: 10.1080/10409238.2021.1934811] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
The serine/threonine kinase mammalian target of rapamycin (mTOR) is the catalytic subunit of two complexes, mTORC1 and mTORC2, which have common and distinct subunits that mediate separate and overlapping functions. mTORC1 is activated by plenty of nutrients, and the two complexes can be activated by PI3K signaling. mTORC2 acts as an upstream regulator of AKT, and mTORC1 acts as a downstream effector. mTOR signaling integrates both intracellular and extracellular signals, acting as a key regulator of cellular metabolism, growth, and survival. A dysregulated activation of mTOR, as result of PI3K pathway or mTOR regulatory protein mutations or even due to the presence of cellular or viral oncogenes, is a common finding in cancer and represents a central mechanism in cancerogenesis. In the final part of this review, we will focus on the PI3K/AKT/mTOR activation by the human gammaherpesviruses EBV and KSHV that hijack this pathway to promote their-mediated oncogenic transformation and pathologies.
Collapse
Affiliation(s)
- Mara Cirone
- Department of Experimental Medicine, "Sapienza" University of Rome, Rome, Italy.,Laboratory affiliated to Istituto Pasteur Italia-Fondazione Cenci Bolognetti, Rome, Italy
| |
Collapse
|
22
|
Suares A, Medina MV, Coso O. Autophagy in Viral Development and Progression of Cancer. Front Oncol 2021; 11:603224. [PMID: 33763351 PMCID: PMC7982729 DOI: 10.3389/fonc.2021.603224] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2020] [Accepted: 01/12/2021] [Indexed: 12/12/2022] Open
Abstract
Autophagy is a complex degradative process by which eukaryotic cells capture cytoplasmic components for subsequent degradation through lysosomal hydrolases. Although this catabolic process can be triggered by a great variety of stimuli, action in cells varies according to cellular context. Autophagy has been previously linked to disease development modulation, including cancer. Autophagy helps suppress cancer cell advancement in tumor transformation early stages, while promoting proliferation and metastasis in advanced settings. Oncoviruses are a particular type of virus that directly contribute to cell transformation and tumor development. Extensive molecular studies have revealed complex ways in which autophagy can suppress or improve oncovirus fitness while still regulating viral replication and determining host cell fate. This review includes recent advances in autophagic cellular function and emphasizes its antagonistic role in cancer cells.
Collapse
Affiliation(s)
- Alejandra Suares
- Departamento de Fisiología y Biología Molecular, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires, Argentina
- Instituto de Fisiología, Biología Molecular y Neurociencias (IFIBYNE), CONICET—Universidad de Buenos Aires, Buenos Aires, Argentina
| | - María Victoria Medina
- Departamento de Fisiología y Biología Molecular, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires, Argentina
- Instituto de Fisiología, Biología Molecular y Neurociencias (IFIBYNE), CONICET—Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Omar Coso
- Departamento de Fisiología y Biología Molecular, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires, Argentina
- Instituto de Fisiología, Biología Molecular y Neurociencias (IFIBYNE), CONICET—Universidad de Buenos Aires, Buenos Aires, Argentina
| |
Collapse
|
23
|
Suares A, Medina MV, Coso O. Autophagy in Viral Development and Progression of Cancer. Front Oncol 2021. [DOI: 10.3389/fonc.2021.603224
expr 816899697 + 824303767] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/16/2023] Open
Abstract
Autophagy is a complex degradative process by which eukaryotic cells capture cytoplasmic components for subsequent degradation through lysosomal hydrolases. Although this catabolic process can be triggered by a great variety of stimuli, action in cells varies according to cellular context. Autophagy has been previously linked to disease development modulation, including cancer. Autophagy helps suppress cancer cell advancement in tumor transformation early stages, while promoting proliferation and metastasis in advanced settings. Oncoviruses are a particular type of virus that directly contribute to cell transformation and tumor development. Extensive molecular studies have revealed complex ways in which autophagy can suppress or improve oncovirus fitness while still regulating viral replication and determining host cell fate. This review includes recent advances in autophagic cellular function and emphasizes its antagonistic role in cancer cells.
Collapse
|
24
|
Rivalta B, Amodio D, Milito C, Chiriaco M, Di Cesare S, Giancotta C, Conti F, Santilli V, Pacillo L, Cifaldi C, Desimio MG, Doria M, Quinti I, De Vito R, Di Matteo G, Finocchi A, Palma P, Trizzino A, Tommasini A, Cancrini C. Case Report: EBV Chronic Infection and Lymphoproliferation in Four APDS Patients: The Challenge of Proper Characterization, Therapy, and Follow-Up. Front Pediatr 2021; 9:703853. [PMID: 34540765 PMCID: PMC8448282 DOI: 10.3389/fped.2021.703853] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Accepted: 08/03/2021] [Indexed: 11/13/2022] Open
Abstract
Activated PI3K-kinase Delta Syndrome (APDS) is an autosomal-dominant primary immunodeficiency (PID) caused by the constitutive activation of the PI3Kδ kinase. The consequent hyperactivation of the PI3K-Akt-mTOR pathway leads to an impaired T- and B-cells differentiation and function, causing progressive lymphopenia, hypogammaglobulinemia and hyper IgM. Patients with APDS show recurrent sinopulmonary and chronic herpes virus infections, immune dysregulation manifestations, including cytopenia, arthritis, inflammatory enteropathy, and a predisposition to persistent non-neoplastic splenomegaly/lymphoproliferation and lymphoma. The recurrence of the lymphoproliferative disorder and the difficulties in the proper definition of malignancy on histological examination represents the main challenge in the clinical management of APDS patients, since a prompt and correct diagnosis is needed to avoid major complications. Targeted therapies with PI3Kδ-Akt-mTOR pathway pharmacologic inhibitors (i.e., Rapamycin, Theophylline, PI3K inhibitors) represent a good therapeutic strategy. They can also be used as bridge therapies when HSCT is required in order to control refractory symptoms. Indeed, treated patients showed a good tolerance, improved immunologic phenotype and reduced incidence/severity of immune dysregulation manifestations. Here, we describe our experience in the management of four patients, one male affected with APDS1 (P1) and the other three, a male and two females, with APDS2 (P2, P3, P4) presenting with chronic EBV replication, recurrent episodes of immune dysregulation manifestations and lymphomas. These cases highlighted the importance of a tailored and close follow-up, including serial endoscopic and lymph nodes biopsies control to detect a prompt and correct diagnosis and offer the best therapeutic strategy.
Collapse
Affiliation(s)
- Beatrice Rivalta
- Research Unit of Primary Immunodeficiencies, Immune and Infectious Diseases Division, Academic Department of Pediatrics (DPUO), Bambino Gesù Children's Hospital, IRCCS, Rome, Italy.,Chair of Pediatrics, Department of Systems Medicine, University of Rome "Tor Vergata", Rome, Italy
| | - Donato Amodio
- Research Unit of Clinical Immunology and Vaccinology, Academic Department of Pediatrics (DPUO), Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Cinzia Milito
- Department of Molecular Medicine, Sapienza University of Rome, Rome, Italy
| | - Maria Chiriaco
- Chair of Pediatrics, Department of Systems Medicine, University of Rome "Tor Vergata", Rome, Italy
| | - Silvia Di Cesare
- Research Unit of Primary Immunodeficiencies, Immune and Infectious Diseases Division, Academic Department of Pediatrics (DPUO), Bambino Gesù Children's Hospital, IRCCS, Rome, Italy.,Chair of Pediatrics, Department of Systems Medicine, University of Rome "Tor Vergata", Rome, Italy
| | - Carmela Giancotta
- Research Unit of Clinical Immunology and Vaccinology, Academic Department of Pediatrics (DPUO), Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Francesca Conti
- Pediatric Unit, IRCCS Azienda Ospedaliero-Universitaria di Bologna, University of Bologna, Bologna, Italy
| | - Veronica Santilli
- Research Unit of Clinical Immunology and Vaccinology, Academic Department of Pediatrics (DPUO), Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Lucia Pacillo
- Research Unit of Primary Immunodeficiencies, Immune and Infectious Diseases Division, Academic Department of Pediatrics (DPUO), Bambino Gesù Children's Hospital, IRCCS, Rome, Italy.,Chair of Pediatrics, Department of Systems Medicine, University of Rome "Tor Vergata", Rome, Italy
| | - Cristina Cifaldi
- Research Unit of Primary Immunodeficiencies, Immune and Infectious Diseases Division, Academic Department of Pediatrics (DPUO), Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Maria Giovanna Desimio
- Research Unit of Primary Immunodeficiencies, Immune and Infectious Diseases Division, Academic Department of Pediatrics (DPUO), Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Margherita Doria
- Research Unit of Primary Immunodeficiencies, Immune and Infectious Diseases Division, Academic Department of Pediatrics (DPUO), Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Isabella Quinti
- Department of Molecular Medicine, Sapienza University of Rome, Rome, Italy
| | - Rita De Vito
- Pathology Unit, Department of Laboratories, Bambino Gesù Children's Hospital, Rome, Italy
| | - Gigliola Di Matteo
- Research Unit of Primary Immunodeficiencies, Immune and Infectious Diseases Division, Academic Department of Pediatrics (DPUO), Bambino Gesù Children's Hospital, IRCCS, Rome, Italy.,Chair of Pediatrics, Department of Systems Medicine, University of Rome "Tor Vergata", Rome, Italy
| | - Andrea Finocchi
- Research Unit of Primary Immunodeficiencies, Immune and Infectious Diseases Division, Academic Department of Pediatrics (DPUO), Bambino Gesù Children's Hospital, IRCCS, Rome, Italy.,Chair of Pediatrics, Department of Systems Medicine, University of Rome "Tor Vergata", Rome, Italy
| | - Paolo Palma
- Chair of Pediatrics, Department of Systems Medicine, University of Rome "Tor Vergata", Rome, Italy.,Research Unit of Clinical Immunology and Vaccinology, Academic Department of Pediatrics (DPUO), Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Antonino Trizzino
- Department of Pediatric Hematology and Oncology, ARNAS Civico Di Cristina and Benfratelli Hospital, Palermo, Italy
| | - Alberto Tommasini
- Institute for Maternal and Child Health, IRCCS Burlo Garofolo, Trieste, Italy.,Department of Medicine, Surgery and Health Sciences, University of Trieste, Trieste, Italy
| | - Caterina Cancrini
- Research Unit of Primary Immunodeficiencies, Immune and Infectious Diseases Division, Academic Department of Pediatrics (DPUO), Bambino Gesù Children's Hospital, IRCCS, Rome, Italy.,Chair of Pediatrics, Department of Systems Medicine, University of Rome "Tor Vergata", Rome, Italy
| |
Collapse
|
25
|
Re VD, Brisotto G, Repetto O, De Zorzi M, Caggiari L, Zanussi S, Alessandrini L, Canzonieri V, Miolo G, Puglisi F, Belluco C, Steffan A, Cannizzaro R. Overview of Epstein-Barr-Virus-Associated Gastric Cancer Correlated with Prognostic Classification and Development of Therapeutic Options. Int J Mol Sci 2020; 21:E9400. [PMID: 33321820 PMCID: PMC7764600 DOI: 10.3390/ijms21249400] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2020] [Revised: 12/07/2020] [Accepted: 12/08/2020] [Indexed: 02/08/2023] Open
Abstract
Gastric cancer (GC) is a deadly disease with poor prognosis that is characterized by heterogeneity. New classifications based on histologic features, genotypes, and molecular phenotypes, for example, the Cancer Genome Atlas subtypes and those by the Asian Cancer Research Group, help understand the carcinogenic differences in GC and have led to the identification of an Epstein-Barr virus (EBV)-related GC subtype (EBVaGC), providing new indications for tailored treatment and prognostic factors. This article provides a review of the features of EBVaGC and an update on the latest insights from EBV-related research with a particular focus on the strict interaction between EBV infection and the gastric tumor environment, including the host immune response. This information may help increase our knowledge of EBVaGC pathogenesis and the mechanisms that sustain the immune response of patients since this mechanism has been demonstrated to offer a survival advantage in a proportion of patients with GC.
Collapse
Affiliation(s)
- Valli De Re
- Immunopathology and Cancer Biomarkers, Department of Translational Research, Bioproteomic Facility, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, 33077 Aviano, Italy; (G.B.); (O.R.); (M.D.Z.); (L.C.); (S.Z.); (A.S.)
| | - Giulia Brisotto
- Immunopathology and Cancer Biomarkers, Department of Translational Research, Bioproteomic Facility, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, 33077 Aviano, Italy; (G.B.); (O.R.); (M.D.Z.); (L.C.); (S.Z.); (A.S.)
| | - Ombretta Repetto
- Immunopathology and Cancer Biomarkers, Department of Translational Research, Bioproteomic Facility, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, 33077 Aviano, Italy; (G.B.); (O.R.); (M.D.Z.); (L.C.); (S.Z.); (A.S.)
| | - Mariangela De Zorzi
- Immunopathology and Cancer Biomarkers, Department of Translational Research, Bioproteomic Facility, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, 33077 Aviano, Italy; (G.B.); (O.R.); (M.D.Z.); (L.C.); (S.Z.); (A.S.)
| | - Laura Caggiari
- Immunopathology and Cancer Biomarkers, Department of Translational Research, Bioproteomic Facility, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, 33077 Aviano, Italy; (G.B.); (O.R.); (M.D.Z.); (L.C.); (S.Z.); (A.S.)
| | - Stefania Zanussi
- Immunopathology and Cancer Biomarkers, Department of Translational Research, Bioproteomic Facility, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, 33077 Aviano, Italy; (G.B.); (O.R.); (M.D.Z.); (L.C.); (S.Z.); (A.S.)
| | - Lara Alessandrini
- Pathology, Department of Medicine DIMED, University of Padova, 61-35121 Padova, Italy;
| | - Vincenzo Canzonieri
- Surgical and Health Sciences, Department of Medical, University of Trieste Medical School, 34100 Trieste, Italy;
- Pathology, Department of Translational Research, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, 33081 Aviano, Italy
| | - Gianmaria Miolo
- Medical Oncology and Cancer Prevention, Department of Medical Oncology, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, 33081 Aviano, Italy; (G.M.); (F.P.)
| | - Fabio Puglisi
- Medical Oncology and Cancer Prevention, Department of Medical Oncology, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, 33081 Aviano, Italy; (G.M.); (F.P.)
- Department of Medicine, University of Udine, 33100 Udine, Italy
| | - Claudio Belluco
- Surgical Oncology, Department of Surgery, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, 33081 Aviano, Italy;
| | - Agostino Steffan
- Immunopathology and Cancer Biomarkers, Department of Translational Research, Bioproteomic Facility, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, 33077 Aviano, Italy; (G.B.); (O.R.); (M.D.Z.); (L.C.); (S.Z.); (A.S.)
| | - Renato Cannizzaro
- Gastroenterology, Department of Medical Oncology, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, 33081 Aviano, Italy;
| |
Collapse
|
26
|
Fattahi S, Amjadi-Moheb F, Tabaripour R, Ashrafi GH, Akhavan-Niaki H. PI3K/AKT/mTOR signaling in gastric cancer: Epigenetics and beyond. Life Sci 2020; 262:118513. [PMID: 33011222 DOI: 10.1016/j.lfs.2020.118513] [Citation(s) in RCA: 239] [Impact Index Per Article: 47.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Revised: 09/22/2020] [Accepted: 09/22/2020] [Indexed: 02/07/2023]
Abstract
PI3K/AKT/mTOR pathway is one of the most important signaling pathways involved in normal cellular processes. Its aberrant activation modulates autophagy, epithelial-mesenchymal transition, apoptosis, chemoresistance, and metastasis in many human cancers. Emerging evidence demonstrates that some infections as well as epigenetic regulatory mechanisms can control PI3K/AKT/mTOR signaling pathway. In this review, we focused on the role of this pathway in gastric cancer development, prognosis, and metastasis, with an emphasis on epigenetic alterations including DNA methylation, histone modifications, and post-transcriptional modulations through non-coding RNAs fluctuations as well as H. pylori and Epstein-Barr virus infections. Finally, we reviewed different molecular targets and therapeutic agents in clinical trials as a potential strategy for gastric cancer treatment through the PI3K/AKT/mTOR pathway.
Collapse
Affiliation(s)
- Sadegh Fattahi
- Student Research Committee, Babol University of Medical Sciences, Babol, Iran; Cellular and Molecular Biology Research Center, Health Research Institute, Babol University of Medical Sciences, Babol, Iran; North Research Center, Pasteur Institute, Amol, Iran
| | - Fatemeh Amjadi-Moheb
- Department of Genetics, Faculty of Medicine, Babol University of Medical Sciences, Babol, Iran
| | - Reza Tabaripour
- Department of Cellular and Molecular Biology, Islamic Azad University Babol-Branch, Iran
| | - Gholam Hossein Ashrafi
- Kingston University London, Cancer theme, School of Life Science, Pharmacy and Chemistry, SEC Faculty, Kingston upon Thames, KT12EE, London, UK
| | - Haleh Akhavan-Niaki
- Department of Genetics, Faculty of Medicine, Babol University of Medical Sciences, Babol, Iran.
| |
Collapse
|
27
|
Stanland LJ, Luftig MA. The Role of EBV-Induced Hypermethylation in Gastric Cancer Tumorigenesis. Viruses 2020; 12:v12111222. [PMID: 33126718 PMCID: PMC7693998 DOI: 10.3390/v12111222] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Revised: 10/24/2020] [Accepted: 10/26/2020] [Indexed: 02/07/2023] Open
Abstract
Epstein–Barr-virus-associated Gastric Cancer (EBVaGC) comprises approximately 10% of global gastric cancers and is known to be the most hypermethylated of all tumor types. EBV infection has been shown to directly induce the hypermethylation of both the host and viral genome following initial infection of gastric epithelial cells. Many studies have been completed in an attempt to identify genes that frequently become hypermethylated and therefore significant pathways that become silenced to promote tumorigenesis. It is clear that EBV-induced hypermethylation silences key tumor suppressor genes, cell cycle genes and cellular differentiation factors to promote a highly proliferative and poorly differentiated cell population. EBV infection has been shown to induce methylation in additional malignancies including Nasopharyngeal Carcinoma and Burkitt’s Lymphoma though not to the same level as in EBVaGC. Lastly, some genes silenced in EBVaGC are common to other heavily methylated tumors such as colorectal and breast tumors; however, some genes are unique to EBVaGC and can provide insights into the major pathways involved in tumorigenesis.
Collapse
|
28
|
Shestakova A, Grove N, Said J, Song S, Quintero-Rivera F. Trisomy 3, a sole recurrent cytogenetic abnormality in pediatric polymorphic post-transplant lymphoproliferative disorder (PTLD). Cancer Genet 2020; 248-249:39-48. [PMID: 33065430 DOI: 10.1016/j.cancergen.2020.09.006] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2020] [Revised: 08/06/2020] [Accepted: 09/28/2020] [Indexed: 11/18/2022]
Abstract
Trisomy 3 has been previously reported in association with T-cell lymphomas and less commonly in different types of non-Hodgkin B-cell lymphomas. Trisomy 3 has also been reported in two cases of pediatric post-transplant lymphoproliferative disorder (PTLD). We present comprehensive clinicopathologic review of two pediatric patients with cardiac and liver/intestinal allografts that developed polymorphic PTLD characterized by trisomy 3. Both patients had Epstein-Barr virus (EBV) viremia and EBV was positive in tissue by EBER in situ hybridization. Using karyotype analysis and fluorescence in situ hybridization, we identified trisomy 3 in both patients. Both patients responded to treatment and are now free of the PTLD. Trisomy 3, an uncommon cytogenetic finding in pediatric polymorphic PTLD, may be a recurrent cytogenetic aberration if confirmed in a larger study of pediatric PTLDs. Further clinical follow up might help stratify significance of trisomy 3 as a prognostic factor.
Collapse
Affiliation(s)
- Anna Shestakova
- Department of Pathology and Laboratory Medicine, University of California, Irvine, UCIMC, Bldg. 1, Rm. 3426, Mail Code: 4805, Orange, CA 92868, USA.
| | - Narina Grove
- Advanced Dermatology of Colorado, 1100 Poudre River Dr ste a, Fort Collins, CO 80524, USA
| | - Jonathan Said
- Department of Pathology and Laboratory Medicine, University of California, Los Angeles, Los Angeles, CA , USA
| | - Sophie Song
- Department of Pathology and Laboratory Medicine, University of California, Los Angeles, Los Angeles, CA , USA
| | - Fabiola Quintero-Rivera
- Department of Pathology and Laboratory Medicine, University of California, Irvine, UCIMC, Bldg. 1, Rm. 3426, Mail Code: 4805, Orange, CA 92868, USA.
| |
Collapse
|
29
|
Richardo T, Prattapong P, Ngernsombat C, Wisetyaningsih N, Iizasa H, Yoshiyama H, Janvilisri T. Epstein-Barr Virus Mediated Signaling in Nasopharyngeal Carcinoma Carcinogenesis. Cancers (Basel) 2020; 12:2441. [PMID: 32872147 PMCID: PMC7565514 DOI: 10.3390/cancers12092441] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2020] [Revised: 08/21/2020] [Accepted: 08/24/2020] [Indexed: 12/14/2022] Open
Abstract
Nasopharyngeal carcinoma (NPC) is one of the most common tumors occurring in China and Southeast Asia. Etiology of NPC seems to be complex and involves many determinants, one of which is Epstein-Barr virus (EBV) infection. Although evidence demonstrates that EBV infection plays a key role in NPC carcinogenesis, the exact relationship between EBV and dysregulation of signaling pathways in NPC needs to be clarified. This review focuses on the interplay between EBV and NPC cells and the corresponding signaling pathways, which are modulated by EBV oncoproteins and non-coding RNAs. These altered signaling pathways could be critical for the initiation and progression of NPC.
Collapse
Affiliation(s)
- Timmy Richardo
- Institute of Virology, Hannover Medical School, 30625 Hannover, Germany;
- Department of Biomedicine, Indonesia International Institute for Life Science (i3L), Jakarta 13210, Indonesia;
- Department of Microbiology, Shimane University, Izumo 693-8501, Japan; (H.I.); (H.Y.)
| | - Pongphol Prattapong
- Graduate Program in Molecular Medicine, Faculty of Science, Mahidol University, Bangkok 10400, Thailand; (P.P.); (C.N.)
| | - Chawalit Ngernsombat
- Graduate Program in Molecular Medicine, Faculty of Science, Mahidol University, Bangkok 10400, Thailand; (P.P.); (C.N.)
| | - Nurulfitri Wisetyaningsih
- Department of Biomedicine, Indonesia International Institute for Life Science (i3L), Jakarta 13210, Indonesia;
| | - Hisashi Iizasa
- Department of Microbiology, Shimane University, Izumo 693-8501, Japan; (H.I.); (H.Y.)
| | - Hironori Yoshiyama
- Department of Microbiology, Shimane University, Izumo 693-8501, Japan; (H.I.); (H.Y.)
| | - Tavan Janvilisri
- Department of Biochemistry, Faculty of Science, Mahidol University, Bangkok 10400, Thailand
| |
Collapse
|
30
|
Velu P, Vijayalakshmi A, Vinothkumar V. Inhibiting the PI3K/Akt, NF-κB signalling pathways with syringic acid for attenuating the development of oral squamous cell carcinoma cells SCC131. J Pharm Pharmacol 2020; 72:1595-1606. [PMID: 32790092 DOI: 10.1111/jphp.13350] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2020] [Accepted: 07/15/2020] [Indexed: 12/25/2022]
Abstract
OBJECTIVES To evaluate the anti-inflammatory and antiproliferative effect of syringic acid (SRA) on oral squamous cell carcinoma (OSCC) SCC131 cells via suppression of NF-κB-induced PI3K/Akt signalling pathway. METHODS The present study assesses the anticancer effects of SRA alongside human oral cancer (HOC) SCC131 cells through the fabrication of reactive oxygen species (ROS) and activated apoptosis. DAPI and Rh-123 staining were used to assess the apoptotic nuclear characteristic, mitochondrial membrane potential, cell adhesion and migration by fluorescence microscope with SRA treatment. KEY FINDINGS Syringic acid inhibits cell viability (IC50 values of 25 µm), adhesion, migration and induced apoptosis. MTT assay demonstrated SRA-induced apoptotic events, inhibition of invasion and angiogenic signalling in SCC131 cell line. Furthermore, SRA treated with SCC131 cells suppresses the protein expression of inflammatory, angiogenesis and PI3K/Akt signalling pathways. It is suggested that SRA prevents the translocation of NF-κB and PI3K/Akt activated products to the nucleus, thereby suppressing angiogenesis via downregulation of vascular endothelial growth factor. CONCLUSIONS Therefore, addition of SRA to SCC131 cells may provide a promising natural therapeutic strategy against squamous cell carcinomas with potential application in clinical analysis.
Collapse
Affiliation(s)
- Periyannan Velu
- Department of Biochemistry and Biotechnology, Faculty of Science, Annamalai University, Chidambaram, Tamil Nadu, India
| | - Annamalai Vijayalakshmi
- Department of Biochemistry and Biotechnology, Faculty of Science, Annamalai University, Chidambaram, Tamil Nadu, India
| | - Veerasamy Vinothkumar
- Department of Biochemistry and Biotechnology, Faculty of Science, Annamalai University, Chidambaram, Tamil Nadu, India
| |
Collapse
|
31
|
Zhang X, Li S, Zheng M, Zhang L, Bai R, Li R, Hao S, Bai B, Kang H. Effects of the PI3K/Akt signaling pathway on the apoptosis of early host cells infected with Eimeria tenella. Parasitol Res 2020; 119:2549-2561. [PMID: 32562065 DOI: 10.1007/s00436-020-06738-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2019] [Accepted: 05/31/2020] [Indexed: 12/14/2022]
Abstract
This study investigated the role of PI3K/Akt signaling pathway on host cell apoptosis in the early infection of Eimeria tenella. Chicken cecal epithelial cells were treated with apoptosis-inducer Actinomycin D (Act D) or PI3K/Akt signaling pathway inhibitor LY294002 and then infected with E. tenella. Results demonstrated that the E. tenella-infected group had less apoptosis 4-8 h after the infection and more apoptosis 12-20 h after the infection than the control group. At 4-20 h after the infection, the apoptotic/necrotic rate and the Caspase-3 activity in the Act D + E. tenella group were lower (P < 0.01) than those in the Act D-treated group. The p-Akt and NF-κB contents in the E. tenella-infected group were higher (P < 0.01) than those in the control group 4-12 h after the infection. However, the bad content and the Caspase-9/3 activity were lower (P < 0.05) in the E. tenella-infected group than in the control group. Compared with the E. tenella-infected group, the LY294002 + E. tenella group showed decreased p-Akt content and increased apoptotic/necrotic rate, bad content, NF-κB expression, membrane permeability transition pore (MPTP) openness, and Caspase-9/3 activity. Thus, the early development of E. tenella could inhibit host cell apoptosis by downregulating the Caspase-3 activity. Upregulating this activity promoted apoptosis. In addition, activating the PI3K/Akt signaling pathway inhibited the apoptosis of E. tenella host cells in the early infection by reducing the expression of the bad content, limiting the MPTP opening, and decreasing the Caspase-9 and Caspase-3 activities.
Collapse
Affiliation(s)
- Xuesong Zhang
- College of Animal Science and Veterinary Medicine, Shanxi Agricultural University, Taigu, Jinzhong, 030801, China
| | - Shan Li
- College of Animal Science and Veterinary Medicine, Shanxi Agricultural University, Taigu, Jinzhong, 030801, China
| | - Mingxue Zheng
- College of Animal Science and Veterinary Medicine, Shanxi Agricultural University, Taigu, Jinzhong, 030801, China.
| | - Li Zhang
- College of Animal Science and Veterinary Medicine, Shanxi Agricultural University, Taigu, Jinzhong, 030801, China
| | - Rui Bai
- College of Animal Science and Veterinary Medicine, Shanxi Agricultural University, Taigu, Jinzhong, 030801, China
| | - Ruiqi Li
- College of Animal Science and Veterinary Medicine, Shanxi Agricultural University, Taigu, Jinzhong, 030801, China
| | - Siyuan Hao
- College of Animal Science and Veterinary Medicine, Shanxi Agricultural University, Taigu, Jinzhong, 030801, China
| | - Bing Bai
- College of Animal Science and Veterinary Medicine, Shanxi Agricultural University, Taigu, Jinzhong, 030801, China
| | - Huixin Kang
- College of Animal Science and Veterinary Medicine, Shanxi Agricultural University, Taigu, Jinzhong, 030801, China
| |
Collapse
|
32
|
Zhu J, Kamara S, Cen D, Tang W, Gu M, Ci X, Chen J, Wang L, Zhu S, Jiang P, Chen S, Xue X, Zhang L. Generation of novel affibody molecules targeting the EBV LMP2A N-terminal domain with inhibiting effects on the proliferation of nasopharyngeal carcinoma cells. Cell Death Dis 2020; 11:213. [PMID: 32238802 PMCID: PMC7113277 DOI: 10.1038/s41419-020-2410-7] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2019] [Revised: 03/12/2020] [Accepted: 03/12/2020] [Indexed: 12/17/2022]
Abstract
Nasopharyngeal carcinoma (NPC) induced by latent infection with Epstein-Barr virus (EBV) remains the most common head and neck cancer in Southeast Asia, especially in the southern part of China. It is well known that persistent expression of two EBV latent membrane proteins (LMP1/LMP2A) plays a key role in nasopharyngeal carcinogenesis. Therefore, the therapeutic approach of targeting the LMP1/LMP2A protein and subsequently blocking the LMP1/LMP2A-mediated signalling pathway has been considered for treating patients with NPC. Recently, affibody molecules, a new class of small (~6.5 kDa) affinity proteins, have been confirmed to be powerful generalisable tools for developing imaging or therapeutic agents by targeting specific molecules. In this study, three EBV LMP2A N-terminal domain-binding affibody molecules (ZLMP2A-N85, ZLMP2A-N110 and ZLMP2A-N252) were identified by screening a phage-displayed peptide library, and their high affinity and specificity for the EBV LMP2A N-terminal domain were confirmed by surface plasmon resonance (SPR), indirect immunofluorescence, co-immunoprecipitation and near-infrared small animal fluorescence imaging in vitro and in vivo. Moreover, affibody molecules targeting the EBV LMP2A N-terminal domain significantly reduced the viability of the EBV-positive cell lines C666-1, CNE-2Z and B95-8. Further investigations showed that affibody ZLMP2A-N110 could inhibit the phosphorylation of AKT, GSK-3β and β-catenin signalling proteins, leading to suppression of β-catenin nuclear translocation and subsequent inhibition of c-Myc oncogene expression, which may be responsible for the reduced viability of NPC-derived cell lines. In conclusion, our findings provide a strong evidence that three novel EBV LMP2A N-terminal domain-binding affibody molecules have great potential for utilisation and development as agents for both molecular imaging and targeted therapy of EBV-related NPC.
Collapse
Affiliation(s)
- Jinshun Zhu
- Institute of Molecular Virology and Immunology, Department of Microbiology and Immunology, School of Basic Medical Sciences, Wenzhou Medical University, 325035, Zhejiang, Wenzhou, China
| | - Saidu Kamara
- Institute of Molecular Virology and Immunology, Department of Microbiology and Immunology, School of Basic Medical Sciences, Wenzhou Medical University, 325035, Zhejiang, Wenzhou, China
| | - Danwei Cen
- Institute of Molecular Virology and Immunology, Department of Microbiology and Immunology, School of Basic Medical Sciences, Wenzhou Medical University, 325035, Zhejiang, Wenzhou, China
| | - Wanlin Tang
- Institute of Molecular Virology and Immunology, Department of Microbiology and Immunology, School of Basic Medical Sciences, Wenzhou Medical University, 325035, Zhejiang, Wenzhou, China
| | - Meiping Gu
- Institute of Molecular Virology and Immunology, Department of Microbiology and Immunology, School of Basic Medical Sciences, Wenzhou Medical University, 325035, Zhejiang, Wenzhou, China
| | - Xingyuan Ci
- Institute of Molecular Virology and Immunology, Department of Microbiology and Immunology, School of Basic Medical Sciences, Wenzhou Medical University, 325035, Zhejiang, Wenzhou, China
| | - Jun Chen
- Institute of Molecular Virology and Immunology, Department of Microbiology and Immunology, School of Basic Medical Sciences, Wenzhou Medical University, 325035, Zhejiang, Wenzhou, China
| | - Lude Wang
- Institute of Molecular Virology and Immunology, Department of Microbiology and Immunology, School of Basic Medical Sciences, Wenzhou Medical University, 325035, Zhejiang, Wenzhou, China
| | - Shanli Zhu
- Institute of Molecular Virology and Immunology, Department of Microbiology and Immunology, School of Basic Medical Sciences, Wenzhou Medical University, 325035, Zhejiang, Wenzhou, China
| | - Pengfei Jiang
- Institute of Molecular Virology and Immunology, Department of Microbiology and Immunology, School of Basic Medical Sciences, Wenzhou Medical University, 325035, Zhejiang, Wenzhou, China
| | - Shao Chen
- Institute of Molecular Virology and Immunology, Department of Microbiology and Immunology, School of Basic Medical Sciences, Wenzhou Medical University, 325035, Zhejiang, Wenzhou, China
| | - Xiangyang Xue
- Institute of Molecular Virology and Immunology, Department of Microbiology and Immunology, School of Basic Medical Sciences, Wenzhou Medical University, 325035, Zhejiang, Wenzhou, China
| | - Lifang Zhang
- Institute of Molecular Virology and Immunology, Department of Microbiology and Immunology, School of Basic Medical Sciences, Wenzhou Medical University, 325035, Zhejiang, Wenzhou, China.
| |
Collapse
|
33
|
Liu W, Song YY, Wang JY, Xiao H, Zhang Y, Luo B. Dysregulation of FOXO transcription factors in Epstein-Barr virus-associated gastric carcinoma. Virus Res 2019; 276:197808. [PMID: 31712122 DOI: 10.1016/j.virusres.2019.197808] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2019] [Revised: 11/07/2019] [Accepted: 11/07/2019] [Indexed: 12/24/2022]
Abstract
Epstein-Barr virus (EBV) infection is associated with the development of gastric cancer (GC). Forkhead box class O (FOXO) transcription factors play important roles in tumor suppression. This study aims to investigate the interplay between EBV and FOXOs in EBV-associated GC (EBVaGC). The results showed that EBV infection of GC cells led to the downregulation of FOXO1 by the inhibition of its mRNA and protein expression. FOXO3 protein is repressed by EBV infection. FOXO4 mRNA is upregulated in EBV-positive cell lines, while its protein expression is downregulated. FOXO1, FOXO3 and FOXO4 proteins are upregulated following PI3K inhibition in GT39 cells, confirming that they are partially suppressed by the PI3K/AKT pathway. However, the upregulation of FOXO1 and FOXO3 by single transfection with LMP1 or LMP2A implies that the dysregulation of FOXOs in EBVaGC is affected by various EBV latent genes and that PI3K/AKT signaling is not the only mechanism of FOXO regulation.
Collapse
Affiliation(s)
- Wen Liu
- Department of Pathogenic Biology, Faculty of Medicine, Qingdao University, Qingdao, China
| | - Ying-Ying Song
- Department of Pathogenic Biology, Faculty of Medicine, Qingdao University, Qingdao, China; Key Laboratory of Medical Molecular Virology (MOH/MOE), Shanghai Medical College, Fudan University, Shanghai, China
| | - Jia-Yi Wang
- Department of Pathogenic Biology, Faculty of Medicine, Qingdao University, Qingdao, China
| | - Hua Xiao
- Department of Pathogenic Biology, Faculty of Medicine, Qingdao University, Qingdao, China
| | - Yan Zhang
- Department of Pathogenic Biology, Faculty of Medicine, Qingdao University, Qingdao, China; Department of Clinical Laboratory, Central Hospital of Zibo, 54 Gongqingtuan Road, ZiBo, 255036, China
| | - Bing Luo
- Department of Pathogenic Biology, Faculty of Medicine, Qingdao University, Qingdao, China.
| |
Collapse
|
34
|
TEEG Induced A549 Cell Autophagy by Regulating the PI3K/AKT/mTOR Signaling Pathway. Anal Cell Pathol (Amst) 2019; 2019:7697610. [PMID: 31183317 PMCID: PMC6515120 DOI: 10.1155/2019/7697610] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2019] [Revised: 03/20/2019] [Accepted: 04/07/2019] [Indexed: 12/14/2022] Open
Abstract
TEEG (3β,16β,23-trihydroxy-13,28-epoxyurs-11-ene-3-O-β-D-glucopyranoside) is derived from the chloroform extract of the Chinese medicine formula Shenqi San (CE-SS). In the present study, we aimed to elucidate the anticancer effect and possible molecular mechanism underlying the action of TEEG against the human non-small cell lung cancer (NSCLC) cell line A549 in vitro. A549 cells were incubated with different concentrations of TEEG. Cell proliferation was assessed by MTT assay. Autophagy was evaluated by immunofluorescence staining. Autophagy-associated proteins were examined by Western blot analysis. TEEG markedly inhibited A549 cell proliferation in a concentration-dependent manner. Immunofluorescence staining showed that TEEG induced autophagy in A549 cells. The LC3-II : LC3-I conversion ratio and the expression of Beclin-1, Atg5, Atg7, and Atg12 increased with the concentration of TEEG. In addition, increased TEEG concentration enhanced the expression of Class III p-PI3K and reduced the expression of Class I p-PI3K, p-AKT, p-mTOR, and p-P70S6K. These results indicate that TEEG induces autophagy of A549 cells through regulation of the PI3K/AKT/mTOR signaling pathway.
Collapse
|
35
|
Visalli RJ, Schwartz AM, Patel S, Visalli MA. Identification of the Epstein Barr Virus portal. Virology 2019; 529:152-159. [PMID: 30710799 DOI: 10.1016/j.virol.2019.01.003] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2018] [Revised: 12/21/2018] [Accepted: 01/03/2019] [Indexed: 12/12/2022]
Abstract
Little is known about Epstein Barr Virus (EBV) proteins that participate in viral DNA cleavage and packaging. Genes encoding potential terminase subunit and portal protein homologs include BGRF1/BDRF1, BALF3, BFRF1A and BBRF1 respectively. EBV mutants with deletions in one or more of these genes were impaired for DNA packaging (Pavlova et al., 2013). In the current study, BBRF1 oligomers were purified from recombinant baculovirus infected insect cell extracts. Transmission electron microscopy revealed that purified EBV portals retained features typically found in other portals including a central channel with clip, stem and wing/crown domains. Although compounds have been identified that target DNA encapsidation in human cytomegalovirus, herpes simplex viruses and varicella-zoster virus, the identification of new EBV targets has lagged significantly. Characterization of the EBV portal will direct studies aimed at developing potential small molecular inhibitors of the EBV encapsidation process.
Collapse
Affiliation(s)
- Robert J Visalli
- Department of Biomedical Sciences, Mercer University School of Medicine, Savannah, GA 31404, USA.
| | - Adam M Schwartz
- Department of Biomedical Sciences, Mercer University School of Medicine, Savannah, GA 31404, USA
| | - Shivam Patel
- Department of Biomedical Sciences, Mercer University School of Medicine, Savannah, GA 31404, USA
| | - Melissa A Visalli
- Department of Biomedical Sciences, Mercer University School of Medicine, Savannah, GA 31404, USA
| |
Collapse
|
36
|
Lang F, Pei Y, Lamplugh ZL, Robertson ES. Molecular Biology of EBV in Relationship to HIV/AIDS-Associated Oncogenesis. Cancer Treat Res 2019; 177:81-103. [PMID: 30523622 DOI: 10.1007/978-3-030-03502-0_4] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
Herpesvirus-induced disease is one of the most lethal factors which leads to high mortality in HIV/AIDS patients. EBV, also known as human herpesvirus 4, can transform naive B cells into immortalized cells in vitro through the regulation of cell cycle, cell proliferation, and apoptosis. EBV infection is associated with several lymphoma and epithelial cancers in humans, which occurs at a much higher rate in immune deficient individuals than in healthy people, demonstrating that the immune system plays a vital role in inhibiting EBV activities. EBV latency infection proteins can mimic suppression cytokines or upregulate PD-1 on B cells to repress the cytotoxic T cells response. Many malignancies, including Hodgkin Lymphoma and non-Hodgkin's lymphomas occur at a much higher frequency in EBV positive individuals than in EBV negative people during the development of HIV infection. Importantly, understanding EBV pathogenesis at the molecular level will aid the development of novel therapies for EBV-induced diseases in HIV/AIDS patients.
Collapse
Affiliation(s)
- Fengchao Lang
- Department of Otorhinolaryngology-Head and Neck Surgery and Tumor Virology and Global Cancer Programs, Abramson Cancer Center, Philadelphia, USA.,Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Yonggang Pei
- Department of Otorhinolaryngology-Head and Neck Surgery and Tumor Virology and Global Cancer Programs, Abramson Cancer Center, Philadelphia, USA.,Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Zachary L Lamplugh
- Department of Otorhinolaryngology-Head and Neck Surgery and Tumor Virology and Global Cancer Programs, Abramson Cancer Center, Philadelphia, USA.,Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Erle S Robertson
- Department of Otorhinolaryngology-Head and Neck Surgery and Tumor Virology and Global Cancer Programs, Abramson Cancer Center, Philadelphia, USA. .,Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA. .,, 3610 Hamilton Walk, 201E Johnson Pavilion, Philadelphia, PA, 19104, USA.
| |
Collapse
|
37
|
Cheerathodi MR, Meckes DG. The Epstein-Barr virus LMP1 interactome: biological implications and therapeutic targets. Future Virol 2018; 13:863-887. [PMID: 34079586 DOI: 10.2217/fvl-2018-0120] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The oncogenic potential of Epstein-Barr virus (EBV) is mostly attributed to latent membrane protein 1 (LMP1), which is essential and sufficient for transformation of fibroblast and primary lymphocytes. LMP1 expression results in the activation of multiple signaling cascades like NF-ΚB and MAP kinases that trigger cell survival and proliferative pathways. LMP1 specific signaling events are mediated through the recruitment of a number of interacting proteins to various signaling domains. Based on these properties, LMP1 is an attractive target to develop effective therapeutics to treat EBV-related malignancies. In this review, we focus on LMP1 interacting proteins, associated signaling events, and potential targets that could be exploited for therapeutic strategies.
Collapse
Affiliation(s)
- Mujeeb R Cheerathodi
- Department of Biomedical Sciences, Florida State University College of Medicine, Tallahassee, FL, 32306
| | - David G Meckes
- Department of Biomedical Sciences, Florida State University College of Medicine, Tallahassee, FL, 32306
| |
Collapse
|
38
|
Phosphatidylinositol 3-Kinase/Akt and MEK/ERK Signaling Pathways Facilitate Sapovirus Trafficking and Late Endosomal Acidification for Viral Uncoating in LLC-PK Cells. J Virol 2018; 92:JVI.01674-18. [PMID: 30282712 PMCID: PMC6258943 DOI: 10.1128/jvi.01674-18] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2018] [Accepted: 09/22/2018] [Indexed: 12/24/2022] Open
Abstract
Sapovirus, an important cause of acute gastroenteritis in humans and animals, travels from the early to the late endosomes and requires late endosomal acidification for viral uncoating. However, the signaling pathways responsible for these viral entry processes remain unknown. Here we demonstrate the receptor-mediated early activation of phosphatidylinositol 3-kinase (PI3K)/Akt and mitogen-activated protein extracellular signal-regulated kinase/extracellular signal-regulated kinase (MEK/ERK) signaling pathways involved in sapovirus entry processes. Both signaling pathways were activated during the early stage of porcine sapovirus (PSaV) infection. However, depletion of the cell surface carbohydrate receptors by pretreatment with sodium periodate or neuraminidase reduced the PSaV-induced early activation of these signaling pathways, indicating that PSaV binding to the cell surface carbohydrate receptors triggered these cascades. Addition of bile acid, known to be essential for PSaV escape from late endosomes, was also found to exert a stiffening effect to stimulate both pathways. Inhibition of these signaling pathways by use of inhibitors specific for PI3K or MEK or small interfering RNAs (siRNAs) against PI3K or MEK resulted in entrapment of PSaV particles in early endosomes and prevented their trafficking to late endosomes. Moreover, phosphorylated PI3K and ERK coimmunoprecipitated subunit E of the V-ATPase proton pump that is important for endosomal acidification. Based on our data, we conclude that receptor binding of PSaV activates both PI3K/Akt and MEK/ERK signaling pathways, which in turn promote PSaV trafficking from early to late endosomes and acidification of late endosomes for PSaV uncoating. These signaling cascades may provide a target for potent therapeutics against infections by PSaV and other caliciviruses.IMPORTANCE Sapoviruses cause acute gastroenteritis in both humans and animals. However, the host signaling pathway(s) that facilitates host cell entry by sapoviruses remains largely unknown. Here we demonstrate that porcine sapovirus (PSaV) activates both PI3K/Akt and MEK/ERK cascades at an early stage of infection. Removal of cell surface receptors decreased PSaV-induced early activation of both cascades. Moreover, blocking of PI3K/Akt and MEK/ERK cascades entrapped PSaV particles in early endosomes and prevented their trafficking to the late endosomes. PSaV-induced early activation of PI3K and ERK molecules further mediated V-ATPase-dependent late endosomal acidification for PSaV uncoating. This work unravels a new mechanism by which receptor-mediated early activation of both cascades may facilitate PSaV trafficking from early to late endosomes and late endosomal acidification for PSaV uncoating, which in turn can be a new target for treatment of sapovirus infection.
Collapse
|
39
|
Li H, Zhu J, He M, Luo Q, Liu F, Chen R. Marek's Disease Virus Activates the PI3K/Akt Pathway Through Interaction of Its Protein Meq With the P85 Subunit of PI3K to Promote Viral Replication. Front Microbiol 2018; 9:2547. [PMID: 30405592 PMCID: PMC6206265 DOI: 10.3389/fmicb.2018.02547] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2018] [Accepted: 10/05/2018] [Indexed: 11/25/2022] Open
Abstract
It is known that viruses can active the phosphatidylinositol 3-kinase (PI3K)/Akt signaling pathway in host cells to support cell survival and viral replication; however, the role of PI3K/Akt signaling in the pathogenic mechanisms induced by Marek’s disease virus (MDV) which causes a neoplastic Marek’s disease in poultry, remains unknown. In this study, we showed that MDV activated the PI3K/Akt pathway in chicken embryo fibroblasts (CEFs) at the early phase of infection, whereas treatment with a PI3K inhibitor LY294002 prior to MDV infection decreased viral replication and DNA synthesis. Flow cytometry analysis showed that inhibition of the PI3K/Akt pathway could significantly increase apoptosis in MDV-infected host cells, indicating that activation of PI3K/Akt signaling could facilitate viral replication through support of cell survival during infection. Evaluation of the underlying molecular mechanism by co-immunoprecipitation and laser confocal microscopy revealed that a viral protein Meq interacted with both p85α and p85β regulatory subunits of PI3K and could induce PI3K/Akt signaling in Meq-overexpressing chicken fibroblasts. Our results showed, for the first time, that MDV activated PI3K/Akt signaling in host cells through interaction of its Meq protein with the regulatory p85 subunit of PI3K to delay cell apoptosis and promote viral replication. This study provides clues for further studies of the molecular mechanisms underlying MDV infection and pathogenicity for the host.
Collapse
Affiliation(s)
- Huimin Li
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| | - Jiaojiao Zhu
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| | - Minyi He
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| | - Qiong Luo
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| | - Fan Liu
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| | - Ruiai Chen
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| |
Collapse
|
40
|
Batool S, Joseph TP, Hussain M, Vuai MS, Khinsar KH, Din SRU, Padhiar AA, Zhong M, Ning A, Zhang W, Cao J, Huang M. LP1 from Lentinula edodes C 91-3 Induces Autophagy, Apoptosis and Reduces Metastasis in Human Gastric Cancer Cell Line SGC-7901. Int J Mol Sci 2018; 19:E2986. [PMID: 30274346 PMCID: PMC6213425 DOI: 10.3390/ijms19102986] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2018] [Revised: 09/24/2018] [Accepted: 09/27/2018] [Indexed: 12/30/2022] Open
Abstract
Present study aimed to elucidate the anticancer effect and the possible molecular mechanism underlying the action of Latcripin 1 (LP1), from the mushroom Lentinula edodes strain C91-3 against gastric cancer cell lines SGC-7901 and BGC-823. Cell viability was measured by Cell Counting Kit-8 (CCK-8); morphological changes were observed by phase contrast microscope; autophagy was determined by transmission electron microscope and fluorescence microscope. Apoptosis and cell cycle were assessed by flow cytometer; wound-healing, transwell migration and invasion assays were performed to investigate the effect of LP1 on gastric cancer cell's migration and invasion. Herein, we found that LP1 resulted in the induction of autophagy by the formation of autophagosomes and conversion of light chain 3 (LC3I into LC3II. LP1 up-regulated the expression level of autophagy-related gene (Atg7, Atg5, Atg12, Atg14) and Beclin1; increased and decreased the expression level of pro-apoptotic (Bax) and anti-apoptotic (Bcl-2) proteins respectively, along with the activation of Caspase-3. At lower-doses, LP1 have shown to arrest cells in the S phase of the cell cycle and decreased the expression level of matrix metalloproteinase MMP-2 and MMP-9. In addition, it has also been shown to regulate the phosphorylation of one of the most hampered gastric cancer pathway, that is, protein kinase B/mammalian target of rapamycin (Akt/mTOR) channel and resulted in cell death. These findings suggested LP1 as a potential natural anti-cancer agent, for exploring the gastric cancer therapies and as a contender for further in vitro and in vivo investigations.
Collapse
Affiliation(s)
- Samana Batool
- Department of Microbiology, College of Basic Medical Sciences, Dalian Medical University, Dalian 116044, China.
| | - Thomson Patrick Joseph
- Department of Microbiology, College of Basic Medical Sciences, Dalian Medical University, Dalian 116044, China.
| | - Mushraf Hussain
- State Key Laboratory of Fine Chemicals, School of Chemical Engineering, Dalian University of Technology, E-208 West Campus, Dalian 116024, China.
| | - Miza S Vuai
- Department of Microbiology, College of Basic Medical Sciences, Dalian Medical University, Dalian 116044, China.
| | - Kavish H Khinsar
- Department of Microbiology, College of Basic Medical Sciences, Dalian Medical University, Dalian 116044, China.
| | - Syed Riaz Ud Din
- Department of Microbiology, College of Basic Medical Sciences, Dalian Medical University, Dalian 116044, China.
| | - Arshad Ahmed Padhiar
- Department of Microbiology, College of Basic Medical Sciences, Dalian Medical University, Dalian 116044, China.
| | - Mintao Zhong
- Department of Microbiology, College of Basic Medical Sciences, Dalian Medical University, Dalian 116044, China.
| | - Anhong Ning
- Department of Microbiology, College of Basic Medical Sciences, Dalian Medical University, Dalian 116044, China.
| | - Wei Zhang
- Department of Microbiology, College of Basic Medical Sciences, Dalian Medical University, Dalian 116044, China.
| | - Jing Cao
- Department of Microbiology, College of Basic Medical Sciences, Dalian Medical University, Dalian 116044, China.
| | - Min Huang
- Department of Microbiology, College of Basic Medical Sciences, Dalian Medical University, Dalian 116044, China.
| |
Collapse
|
41
|
Turunen A, Rautava J, Grénman R, Syrjänen K, Syrjänen S. Epstein-Barr virus (EBV)-encoded small RNAs (EBERs) associated with poor prognosis of head and neck carcinomas. Oncotarget 2018; 8:27328-27338. [PMID: 28423694 PMCID: PMC5432338 DOI: 10.18632/oncotarget.16033] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2016] [Accepted: 02/12/2017] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND Epstein-Barr virus (EBV) is the main cause of nasopharyngeal carcinoma (NPC), also found in other head and neck carcinomas (HNSCCs) where its role remains controversial. RESULTS EBV was found in 80% and 21% of the samples with PCR and ISH (in cancer cells), respectively. Eight of ISH-positive samples were not NPCs. EBER-RNA detection in carcinoma cells was associated with worse prognosis, whether or not NPCs were included. HPV/EBV and HSV/HPV coinfections associated with a shorter survival. LMP-1 expression, positive in 51% of samples did not correlate with the disease outcome. MATERIALS AND METHODS We analyzed EBV in 73 HNSCC samples with a known HPV and HSV-1 status, using in situ hybridization (ISH) and immunohistochemistry (IHC) for EBV-early transcripts (EBER) and LMP-1 protein, respectively. EBV-DNA was detected with a Luminex-based method. The results were correlated with HPV-status and disease outcome. CONCLUSIONS EBV is transcriptionally active in NPC cells but also in a subgroup of other HNSCCs.
Collapse
Affiliation(s)
- Aaro Turunen
- Department of Oral Pathology, Institute of Dentistry, University of Turku, Turku, Finland
| | - Jaana Rautava
- Department of Oral Pathology, Institute of Dentistry, University of Turku, Turku, Finland.,Department of Pathology, Turku University Central Hospital, Turku, Finland
| | - Reidar Grénman
- Department of Otorhinolaryngology - Head and Neck Surgery, Turku University Hospital, Turku, Finland
| | - Kari Syrjänen
- Department of Clinical Research, Biohit HealthCare Oyj, Helsinki, Finland
| | - Stina Syrjänen
- Department of Oral Pathology, Institute of Dentistry, University of Turku, Turku, Finland.,Department of Pathology, Turku University Central Hospital, Turku, Finland
| |
Collapse
|
42
|
Pizzi M, Margolskee E, Inghirami G. Pathogenesis of Peripheral T Cell Lymphoma. ANNUAL REVIEW OF PATHOLOGY-MECHANISMS OF DISEASE 2018; 13:293-320. [DOI: 10.1146/annurev-pathol-020117-043821] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Affiliation(s)
- Marco Pizzi
- Department of Pathology and Laboratory Medicine, Weill Cornell Medical College, Cornell University, New York, NY 10021, USA
- Surgical Pathology and Cytopathology Unit, Department of Medicine-DIMED, University of Padova, 35121 Padova, Italy
| | - Elizabeth Margolskee
- Department of Pathology and Laboratory Medicine, Weill Cornell Medical College, Cornell University, New York, NY 10021, USA
| | - Giorgio Inghirami
- Department of Pathology and Laboratory Medicine, Weill Cornell Medical College, Cornell University, New York, NY 10021, USA
- Department of Molecular Biotechnology and Health Science and Center for Experimental Research and Medical Studies (CeRMS), University of Torino, 10126 Torino, Italy
- Department of Pathology and NYU Cancer Center, NYU School of Medicine, New York, NY 10016, USA
| |
Collapse
|
43
|
Mui UN, Haley CT, Tyring SK. Viral Oncology: Molecular Biology and Pathogenesis. J Clin Med 2017; 6:E111. [PMID: 29186062 PMCID: PMC5742800 DOI: 10.3390/jcm6120111] [Citation(s) in RCA: 120] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2017] [Revised: 11/17/2017] [Accepted: 11/22/2017] [Indexed: 02/06/2023] Open
Abstract
Oncoviruses are implicated in approximately 12% of all human cancers. A large number of the world's population harbors at least one of these oncoviruses, but only a small proportion of these individuals go on to develop cancer. The interplay between host and viral factors is a complex process that works together to create a microenvironment conducive to oncogenesis. In this review, the molecular biology and oncogenic pathways of established human oncoviruses will be discussed. Currently, there are seven recognized human oncoviruses, which include Epstein-Barr Virus (EBV), Human Papillomavirus (HPV), Hepatitis B and C viruses (HBV and HCV), Human T-cell lymphotropic virus-1 (HTLV-1), Human Herpesvirus-8 (HHV-8), and Merkel Cell Polyomavirus (MCPyV). Available and emerging therapies for these oncoviruses will be mentioned.
Collapse
Affiliation(s)
- Uyen Ngoc Mui
- Center for Clinical Studies, Houston, TX 77004, USA.
| | | | - Stephen K Tyring
- Center for Clinical Studies, Houston, TX 77004, USA.
- Department of Dermatology, University of Texas Health Science Center at Houston, Houston, TX 77004, USA.
| |
Collapse
|
44
|
Ni Z, Yi J. Oxymatrine induces nasopharyngeal cancer cell death through inhibition of PI3K/AKT and NF‑κB pathways. Mol Med Rep 2017; 16:9701-9706. [PMID: 29152662 DOI: 10.3892/mmr.2017.7822] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2016] [Accepted: 07/26/2017] [Indexed: 11/06/2022] Open
Abstract
Oxymatrine may inhibit tumor cell proliferation, induce cell cycle arrest, promote apoptosis, induce tumor cell differentiation and fight against tumor angiogenesis, as well as inhibit tumor invasion and metastasis. The present study aimed to investigate the anticancer effects of oxymatrine on nasopharyngeal cancer (NPC) cell death, and the underlying molecular mechanisms of these effects. NPC HK‑1 cells were incubated overnight and treated with oxymatrine (0, 2, 4, 6 and 8 mg/ml) for 1, 2 or 3 days. The results demonstrated that oxymatrine significantly inhibited NPC cell proliferation in a time‑ and dose‑dependent manner. Oxymatrine treatment also induced apoptosis, induced the activities of caspase‑3 and caspase‑9, promoted p53 and Bax protein expression, and suppressed cyclin D protein expression in these cells. The protein expression levels of phosphoinositide 3 kinase (PI3K), phosphorylated (p)‑AKT, p‑mammalian target of rapamycin, p‑p70 ribosomal protein S6 kinase and nuclear factor (NF)‑κB were significantly downregulated by oxymatrine treatment. In conclusion, results from the present study suggested that oxymatrine may induce NPC cell death through the inhibition of PI3K/AKT and NF‑κB signaling pathways.
Collapse
Affiliation(s)
- Zhili Ni
- Department of Otorhinolaryngology Head and Neck Surgery, Xuanwu Hospital, Capital Medical University, Beijing 100053, P.R. China
| | - Jingmei Yi
- Department of Paediatrics, Chinese PLA General Hospital, Beijing 100853, P.R. China
| |
Collapse
|
45
|
Comparison of Clinicopathologic Parameters and Survivals Between Epstein-Barr Virus–positive and Her2-positive Gastric Cancers. Appl Immunohistochem Mol Morphol 2017; 25:609-614. [DOI: 10.1097/pai.0000000000000353] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
46
|
Carrasco-Avino G, Riquelme I, Padilla O, Villaseca M, Aguayo FR, Corvalan AH. The conundrum of the Epstein-Barr virus-associated gastric carcinoma in the Americas. Oncotarget 2017; 8:75687-75698. [PMID: 29088902 PMCID: PMC5650457 DOI: 10.18632/oncotarget.18497] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2016] [Accepted: 05/29/2017] [Indexed: 02/07/2023] Open
Abstract
Epstein-Barr virus-associated gastric carcinoma shows a higher prevalence in the Americas than Asia. We summarize all studies of Epstein Barr virus-associated gastric carcinoma in the Americas, focusing on host characteristics, environmental associations and phylogeographic diversity of Epstein-Barr virus strains. In the Americas, the prevalence of Epstein Barr virus-associated gastric carcinoma is 11.4%, more frequent in males and portray predominantly diffuse-type histology. EBERs, EBNAs, BARTs and LMP are the highest expressed genes; their variations in healthy individuals may explain the phylogeographic diversity of Epstein-Barr virus across the region. Gastric cancer cases harbor exclusively the western genotype (subtype D and kept Xho I site), suggesting a disrupted co-evolution between the pathogen and its host. Epstein-Barr virus-associated gastric carcinoma molecular subtype cases from The Cancer Genome Atlas display PIK3CA gene mutations, amplification of JAK2, PD-L1 and PD-L2 and CpG island methylator phenotype, leading to more extensive methylation of host and viral genomes than any other subtypes from the study. Environmental conditions include negative- and positive- associations with being firstborn child and smoking, respectively. A marginal association with H. pylori has also been reported. Lymphoepithelioma-like carcinoma is associated with Epstein Barr virus in 80%-86% of cases, most of which have been included as part of Epstein Barr virus-associated gastric carcinoma series (prevalence 1.1%-7.6%). Whether these cases represent a variant of Epstein-Barr virus-associated gastric carcinoma is discussed. We propose novel research strategies to solve the conundrum of the high prevalence of Epstein-Barr virus-associated gastric carcinoma in the Americas.
Collapse
Affiliation(s)
- Gonzalo Carrasco-Avino
- Advanced Center for Chronic Diseases (ACCDIS), Pontificia Universidad Catolica de Chile, Santiago, Chile
- Department of Pathology, Faculty of Medicine, Universidad de Chile, Santiago, Chile
| | - Ismael Riquelme
- Scientific and Technological Bioresource Nucleus (BIOREN), Universidad de la Frontera, Temuco, Chile
- Department of Pathology, Universidad de la Frontera, Temuco, Chile
| | - Oslando Padilla
- Department of Public Health, School of Medicine, Pontificia Universidad Catolica de Chile, Santiago, Chile
| | - Miguel Villaseca
- Department of Pathology, Universidad de la Frontera, Temuco, Chile
| | - Francisco R. Aguayo
- Advanced Center for Chronic Diseases (ACCDIS), Pontificia Universidad Catolica de Chile, Santiago, Chile
- Department of Basic and Clinical Oncology, Faculty of Medicine, Universidad de Chile, Santiago, Chile
| | - Alejandro H. Corvalan
- Advanced Center for Chronic Diseases (ACCDIS), Pontificia Universidad Catolica de Chile, Santiago, Chile
- UC-Center for Investigational Oncology (CITO), Pontificia Universidad Catolica de Chile, Santiago, Chile
- Department of Hematology and Oncology, School of Medicine, Pontificia Universidad Catolica de Chile, Santiago, Chile
| |
Collapse
|
47
|
Fujieda M, Tsuruga K, Sato T, Kikuchi H, Tamaki W, Ishihara M, Yamamoto M, Oishi T, Tanaka H, Daibata M. Monitoring of Epstein-Barr virus load and killer T cells in patients with juvenile idiopathic arthritis treated with methotrexate or tocilizumab. Mod Rheumatol 2017; 27:66-71. [PMID: 27166661 DOI: 10.1080/14397595.2016.1177247] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2016] [Accepted: 03/29/2016] [Indexed: 10/21/2022]
Abstract
OBJECTIVES Methotrexate (MTX) is used for the treatment of polyarticular juvenile idiopathic arthritis (JIA), and an anti-interleukin-6 receptor monoclonal antibody (tocilizumab: TCZ) is also used and added for the treatment of intractable JIA. It has been reported that MTX might induce Epstein-Barr virus (EBV)-associated lymphoma, but the discussion about the effect of MTX and/or TCZ against reactivation of EBV in pediatric patients has been incomplete. METHODS The EBV loads in four polyarticular JIA and three systemic arthritis JIA patients treated with MTX and/or TCZ, and the percentage of EBV-specific killer T cells (EBV-CTLs) in some patients were prospectively monitored. RESULTS No patients had EBV-associated symptoms during the observation period. EBV loads in all patients were not significantly increased, and the levels of EBV loads were the same as EBV-seropositive healthy children following the administration of MTX and/or TCZ. EBV-CTLs were detectable during the observation period, but some patients had slightly low levels of EBV-CTLs. CONCLUSION Treatment with MTX and/or TCZ did not severely affect EBV load and prevent induction of EBV-CTLs in JIA patients.
Collapse
MESH Headings
- Adolescent
- Antibodies, Monoclonal, Humanized/adverse effects
- Antibodies, Monoclonal, Humanized/pharmacology
- Antibodies, Monoclonal, Humanized/therapeutic use
- Arthritis, Juvenile/drug therapy
- Arthritis, Juvenile/virology
- Child
- Child, Preschool
- Female
- Herpesvirus 4, Human/isolation & purification
- Humans
- Lymphoma/diagnosis
- Lymphoma/etiology
- Lymphoma/virology
- Male
- Methotrexate/adverse effects
- Methotrexate/pharmacology
- Methotrexate/therapeutic use
- T-Lymphocytes/drug effects
- Viral Load
- Young Adult
Collapse
Affiliation(s)
- Mikiya Fujieda
- a Department of Pediatrics , Kochi Medical School, Kochi University , Nankoku, Kochi , Japan
| | - Kazushi Tsuruga
- b Department of Pediatrics , Graduate School of Medicine, Hirosaki University , Aomori , Japan , and
| | - Tetsuya Sato
- a Department of Pediatrics , Kochi Medical School, Kochi University , Nankoku, Kochi , Japan
| | - Hiroaki Kikuchi
- a Department of Pediatrics , Kochi Medical School, Kochi University , Nankoku, Kochi , Japan
| | - Wataru Tamaki
- a Department of Pediatrics , Kochi Medical School, Kochi University , Nankoku, Kochi , Japan
| | - Masayuki Ishihara
- a Department of Pediatrics , Kochi Medical School, Kochi University , Nankoku, Kochi , Japan
| | - Masaki Yamamoto
- a Department of Pediatrics , Kochi Medical School, Kochi University , Nankoku, Kochi , Japan
| | - Taku Oishi
- a Department of Pediatrics , Kochi Medical School, Kochi University , Nankoku, Kochi , Japan
| | - Hiroshi Tanaka
- b Department of Pediatrics , Graduate School of Medicine, Hirosaki University , Aomori , Japan , and
| | - Masanori Daibata
- c Department of Microbiology , Kochi Medical School, Kochi University , Nankoku, Kochi , Japan
| |
Collapse
|
48
|
Guo Q, Lu T, Chen Y, Su Y, Zheng Y, Chen Z, Chen C, Lin S, Pan J, Yuan X. Genetic variations in the PI3K-PTEN-AKT-mTOR pathway are associated with distant metastasis in nasopharyngeal carcinoma patients treated with intensity-modulated radiation therapy. Sci Rep 2016; 6:37576. [PMID: 27876891 PMCID: PMC5120316 DOI: 10.1038/srep37576] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2016] [Accepted: 11/01/2016] [Indexed: 12/26/2022] Open
Abstract
Distant metastasis is the primary failure pattern of nasopharyngeal carcinoma(NPC) in intensity-modulated radiation therapy(IMRT) era. This study was conducted to find the impact of genetic variations in the phosphatidylinositol 3-kinase(PI3K)/phosphatase and tensin homologue(PTEN)/v-akt murine thymoma viral oncogene homologue(AKT)/mammalian target of rapamycin(mTOR) pathway on the risk of distant metastasis in NPC. We genotyped 16 single-nucleotide polymorphisms(SNPs) in five core genes in this pathway from 496 patients treated by IMRT with or without chemotherapy. The relationships between genetic polymorphisms and distant progression were evaluated. We observed that two loci in the AKT1 gene(rs3803300 and rs2494738 alone or combined) were associated with prognosis, with patients carrying at least one variant allele had significantly reduced risk of distant failure, especially in N2-3 group. In addition, we found that genetic variation may had some joint effect with N classification in recursive-partitioning analysis(RPA) analysis, with which patients were stratified into four different risk subgroups (RPA model): RPA1(low risk), RPA2(moderate risk), RPA3(high risk) and RPA4(highest risk). Our findings suggested that genetic variations within the PI3K signaling pathway modulate the development and invasion of NPC patients. Further research is needed to replicate the study in other centers and races, and to unravel the functional significance of these polymorphisms.
Collapse
Affiliation(s)
- Qiaojuan Guo
- Shengli Clinical Medicine College, Fujian Medical University, Fuzhou, Fujian Province, 350014, China
| | - Tianzhu Lu
- Shengli Clinical Medicine College, Fujian Medical University, Fuzhou, Fujian Province, 350014, China
| | - Yan Chen
- Shengli Clinical Medicine College, Fujian Medical University, Fuzhou, Fujian Province, 350014, China.,Department of Clinical Laboratory, Fujian Provincial Cancer Hospital, Fuzhou, Fujian Province, 350014, China
| | - Ying Su
- Shengli Clinical Medicine College, Fujian Medical University, Fuzhou, Fujian Province, 350014, China.,Department of Radiation Biology Laboratory, Fujian Provincial Cancer Hospital, Fuzhou, Fujian Province, 350014, China
| | - Yuhong Zheng
- Shengli Clinical Medicine College, Fujian Medical University, Fuzhou, Fujian Province, 350014, China.,Department of Clinical Laboratory, Fujian Provincial Cancer Hospital, Fuzhou, Fujian Province, 350014, China
| | - Zeng Chen
- Shengli Clinical Medicine College, Fujian Medical University, Fuzhou, Fujian Province, 350014, China.,Department of Radiation Biology Laboratory, Fujian Provincial Cancer Hospital, Fuzhou, Fujian Province, 350014, China
| | - Chao Chen
- Shengli Clinical Medicine College, Fujian Medical University, Fuzhou, Fujian Province, 350014, China.,Department of Radiation Biology Laboratory, Fujian Provincial Cancer Hospital, Fuzhou, Fujian Province, 350014, China
| | - Shaojun Lin
- Shengli Clinical Medicine College, Fujian Medical University, Fuzhou, Fujian Province, 350014, China.,Department of Radiation Oncology, Fujian Provincial Cancer Hospital, Fuzhou, Fujian Province, 350014, China.,Fujian Provincial Key Laboratory of Translational Cancer Medicine, Fuzhou, Fujian Province, 350014, China
| | - Jianji Pan
- Shengli Clinical Medicine College, Fujian Medical University, Fuzhou, Fujian Province, 350014, China.,Department of Radiation Oncology, Fujian Provincial Cancer Hospital, Fuzhou, Fujian Province, 350014, China.,Fujian Provincial Key Laboratory of Translational Cancer Medicine, Fuzhou, Fujian Province, 350014, China
| | - Xianglin Yuan
- Department of Oncology, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, Hubei Province, 430030, China
| |
Collapse
|
49
|
Jha HC, Pei Y, Robertson ES. Epstein-Barr Virus: Diseases Linked to Infection and Transformation. Front Microbiol 2016; 7:1602. [PMID: 27826287 PMCID: PMC5078142 DOI: 10.3389/fmicb.2016.01602] [Citation(s) in RCA: 81] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2016] [Accepted: 09/26/2016] [Indexed: 12/16/2022] Open
Abstract
Epstein–Barr virus (EBV) was first discovered in 1964, and was the first known human tumor virus now shown to be associated with a vast number of human diseases. Numerous studies have been conducted to understand infection, propagation, and transformation in various cell types linked to human diseases. However, a comprehensive lens through which virus infection, reactivation and transformation of infected host cells can be visualized is yet to be formally established and will need much further investigation. Several human cell types infected by EBV have been linked to associated diseases. However, whether these are a direct result of EBV infection or indirectly due to contributions by additional infectious agents will need to be fully investigated. Therefore, a thorough examination of infection, reactivation, and cell transformation induced by EBV will provide a more detailed view of its contributions that drive pathogenesis. This undoubtedly expand our knowledge of the biology of EBV infection and the signaling activities of targeted cellular factors dysregulated on infection. Furthermore, these insights may lead to identification of therapeutic targets and agents for clinical interventions. Here, we review the spectrum of EBV-associated diseases, the role of the encoded latent antigens, and the switch to latency or lytic replication which occurs in EBV infected cells. Furthermore, we describe the cellular processes and critical factors which contribute to cell transformation. We also describe the fate of B-cells and epithelial cells after EBV infection and the expected consequences which contribute to establishment of viral-associated pathologies.
Collapse
Affiliation(s)
- Hem C Jha
- Department of Otorhinolaryngology-Head and Neck Surgery and Tumor Virology Program, Abramson Cancer Center, Perelman School of Medicine at the University of Pennsylvania, Philadelphia PA, USA
| | - Yonggang Pei
- Department of Otorhinolaryngology-Head and Neck Surgery and Tumor Virology Program, Abramson Cancer Center, Perelman School of Medicine at the University of Pennsylvania, Philadelphia PA, USA
| | - Erle S Robertson
- Department of Otorhinolaryngology-Head and Neck Surgery and Tumor Virology Program, Abramson Cancer Center, Perelman School of Medicine at the University of Pennsylvania, Philadelphia PA, USA
| |
Collapse
|
50
|
Huang-Doran I, Tomlinson P, Payne F, Gast A, Sleigh A, Bottomley W, Harris J, Daly A, Rocha N, Rudge S, Clark J, Kwok A, Romeo S, McCann E, Müksch B, Dattani M, Zucchini S, Wakelam M, Foukas LC, Savage DB, Murphy R, O'Rahilly S, Barroso I, Semple RK. Insulin resistance uncoupled from dyslipidemia due to C-terminal PIK3R1 mutations. JCI Insight 2016; 1:e88766. [PMID: 27766312 PMCID: PMC5070960 DOI: 10.1172/jci.insight.88766] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Obesity-related insulin resistance is associated with fatty liver, dyslipidemia, and low plasma adiponectin. Insulin resistance due to insulin receptor (INSR) dysfunction is associated with none of these, but when due to dysfunction of the downstream kinase AKT2 phenocopies obesity-related insulin resistance. We report 5 patients with SHORT syndrome and C-terminal mutations in PIK3R1, encoding the p85α/p55α/p50α subunits of PI3K, which act between INSR and AKT in insulin signaling. Four of 5 patients had extreme insulin resistance without dyslipidemia or hepatic steatosis. In 3 of these 4, plasma adiponectin was preserved, as in insulin receptor dysfunction. The fourth patient and her healthy mother had low plasma adiponectin associated with a potentially novel mutation, p.Asp231Ala, in adiponectin itself. Cells studied from one patient with the p.Tyr657X PIK3R1 mutation expressed abundant truncated PIK3R1 products and showed severely reduced insulin-stimulated association of mutant but not WT p85α with IRS1, but normal downstream signaling. In 3T3-L1 preadipocytes, mutant p85α overexpression attenuated insulin-induced AKT phosphorylation and adipocyte differentiation. Thus, PIK3R1 C-terminal mutations impair insulin signaling only in some cellular contexts and produce a subphenotype of insulin resistance resembling INSR dysfunction but unlike AKT2 dysfunction, implicating PI3K in the pathogenesis of key components of the metabolic syndrome. C-terminal mutations in human PIK3R1 are associated with severe insulin resistance in the absence of dyslipidemia or hepatic steatosis.
Collapse
Affiliation(s)
- Isabel Huang-Doran
- The University of Cambridge Metabolic Research Laboratories, Wellcome Trust-MRC Institute of Metabolic Science, Cambridge, United Kingdom.,The National Institute for Health Research Cambridge Biomedical Research Centre, Cambridge, United Kingdom
| | - Patsy Tomlinson
- The University of Cambridge Metabolic Research Laboratories, Wellcome Trust-MRC Institute of Metabolic Science, Cambridge, United Kingdom.,The National Institute for Health Research Cambridge Biomedical Research Centre, Cambridge, United Kingdom
| | - Felicity Payne
- Metabolic Disease Group, Wellcome Trust Sanger Institute, Cambridge, United Kingdom
| | - Alexandra Gast
- The University of Cambridge Metabolic Research Laboratories, Wellcome Trust-MRC Institute of Metabolic Science, Cambridge, United Kingdom.,The National Institute for Health Research Cambridge Biomedical Research Centre, Cambridge, United Kingdom
| | - Alison Sleigh
- Wolfson Brain Imaging Centre, University of Cambridge, Cambridge, United Kingdom.,National Institute for Health Research/Wellcome Trust Clinical Research Facility, Cambridge, United Kingdom
| | - William Bottomley
- Metabolic Disease Group, Wellcome Trust Sanger Institute, Cambridge, United Kingdom
| | - Julie Harris
- The University of Cambridge Metabolic Research Laboratories, Wellcome Trust-MRC Institute of Metabolic Science, Cambridge, United Kingdom.,The National Institute for Health Research Cambridge Biomedical Research Centre, Cambridge, United Kingdom
| | - Allan Daly
- Metabolic Disease Group, Wellcome Trust Sanger Institute, Cambridge, United Kingdom
| | - Nuno Rocha
- The University of Cambridge Metabolic Research Laboratories, Wellcome Trust-MRC Institute of Metabolic Science, Cambridge, United Kingdom.,The National Institute for Health Research Cambridge Biomedical Research Centre, Cambridge, United Kingdom
| | - Simon Rudge
- Inositide Laboratory, Babraham Institute, Cambridge, United Kingdom
| | - Jonathan Clark
- Inositide Laboratory, Babraham Institute, Cambridge, United Kingdom
| | - Albert Kwok
- The University of Cambridge Metabolic Research Laboratories, Wellcome Trust-MRC Institute of Metabolic Science, Cambridge, United Kingdom.,The National Institute for Health Research Cambridge Biomedical Research Centre, Cambridge, United Kingdom
| | - Stefano Romeo
- Department of Molecular and Clinical Medicine, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden.,Clinical Nutrition Unit, Department of Medical and Surgical Sciences, University Magna Graecia, Catanzaro, Italy
| | - Emma McCann
- Department of Clinical Genetics, Glan Clwyd Hospital, Rhyl, United Kingdom
| | - Barbara Müksch
- Department of Pediatrics, Children's Hospital, Cologne, Germany
| | - Mehul Dattani
- Section of Genetics and Epigenetics in Health and Disease, Genetics and Genomic Medicine Programme, UCL Institute of Child Health, London, United Kingdom
| | - Stefano Zucchini
- Pediatric Endocrine Unit, S.Orsola-Malpighi Hospital, Bologna, Italy
| | - Michael Wakelam
- Inositide Laboratory, Babraham Institute, Cambridge, United Kingdom
| | - Lazaros C Foukas
- Institute of Healthy Ageing and Department of Genetics, Evolution and Environment, University College London, London, United Kingdom
| | - David B Savage
- The University of Cambridge Metabolic Research Laboratories, Wellcome Trust-MRC Institute of Metabolic Science, Cambridge, United Kingdom.,The National Institute for Health Research Cambridge Biomedical Research Centre, Cambridge, United Kingdom
| | - Rinki Murphy
- Department of Medicine, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| | - Stephen O'Rahilly
- The University of Cambridge Metabolic Research Laboratories, Wellcome Trust-MRC Institute of Metabolic Science, Cambridge, United Kingdom.,The National Institute for Health Research Cambridge Biomedical Research Centre, Cambridge, United Kingdom
| | - Inês Barroso
- The University of Cambridge Metabolic Research Laboratories, Wellcome Trust-MRC Institute of Metabolic Science, Cambridge, United Kingdom.,The National Institute for Health Research Cambridge Biomedical Research Centre, Cambridge, United Kingdom.,Metabolic Disease Group, Wellcome Trust Sanger Institute, Cambridge, United Kingdom
| | - Robert K Semple
- The University of Cambridge Metabolic Research Laboratories, Wellcome Trust-MRC Institute of Metabolic Science, Cambridge, United Kingdom.,The National Institute for Health Research Cambridge Biomedical Research Centre, Cambridge, United Kingdom
| |
Collapse
|