1
|
Mahdavi P, Panahipoor Javaherdehi A, Khanjanpoor P, Aminian H, Zakeri M, Zafarani A, Razizadeh MH. The role of c-Myc in Epstein-Barr virus-associated cancers: Mechanistic insights and therapeutic implications. Microb Pathog 2024; 197:107025. [PMID: 39426639 DOI: 10.1016/j.micpath.2024.107025] [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: 08/05/2024] [Revised: 09/26/2024] [Accepted: 10/16/2024] [Indexed: 10/21/2024]
Abstract
c-Myc is an important proto-oncogene belonging to the MYC family. In normal conditions, c-Myc regulates different aspects of cellular function. However, its dysregulation can result in the development of cancer due to various mechanisms. Epstein-Barr virus is a ubiquitous viral pathogen that infects a huge proportion of the global population. This virus is linked to various cancers, such as different types of lymphoma, nasopharyngeal, and gastric cancers. It can manipulate host cells, and many cellular and viral genes are important in the Epstein-Barr virus carcinogenesis. This review explores the complex relationship between c-Myc and Epstein-Barr virus in the context of cancer development. Also, potential therapeutic strategies targeting c-Myc to treat EBV-related cancers are discussed.
Collapse
Affiliation(s)
- Pooya Mahdavi
- College of Public Health, University of South Florida, Tampa, FL, 33612, USA
| | | | - Parinaz Khanjanpoor
- Department of Health and Science, University of Piedmont Orientale (UPO), Novara, Italy
| | - Hesam Aminian
- Department of Health and Science, University of Piedmont Orientale (UPO), Novara, Italy
| | - Mehrasa Zakeri
- Department of Immunology, Pasteur Institute of Iran, Tehran, Iran
| | - Alireza Zafarani
- Cellular and Molecular Research Center, Iran University of Medical Sciences, Tehran, Iran; Department of Hematology & Blood Banking, School of Allied Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Mohammad Hossein Razizadeh
- Department of Virology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran; Antimicrobial Resistance Research Center, Institute of Immunology and Infectious Diseases, Iran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
2
|
Zhang D, Yang J, Huang Q, Zhao D, Wang T, Yu D, Qin L, Zhang K. Molecular functions of HAX1 during disease progress. Virus Genes 2024; 60:435-445. [PMID: 38992331 DOI: 10.1007/s11262-024-02081-8] [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: 01/11/2024] [Accepted: 05/29/2024] [Indexed: 07/13/2024]
Abstract
HCLS1-associated protein X-1 (HAX1) is a newly discovered multifunctional cell regulatory protein that is widely expressed in cells and has a close relationship with multiple cellular proteins. HAX1 plays important roles in various processes, including the regulation of apoptosis, maintenance of mitochondrial membrane potential stability and calcium homeostasis, occurrence and development of diseases, post-transcriptional regulation of gene expression, and host immune response after viral infection. In this article, we have reviewed the research progress on the biological functions of HAX1, thereby laying a theoretical foundation for further exploration of its underlying mechanisms and targeted application.
Collapse
Affiliation(s)
- Dajun Zhang
- College of Life Sciences and Engineering, Foshan University, Foshan, 528225, Guangdong Province, People's Republic of China
| | - Jinke Yang
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, 730000, China
| | - Qi Huang
- College of Life Sciences and Engineering, Foshan University, Foshan, 528225, Guangdong Province, People's Republic of China
| | - Dengshuai Zhao
- College of Life Sciences and Engineering, Foshan University, Foshan, 528225, Guangdong Province, People's Republic of China
| | - Tianyu Wang
- College of Life Sciences and Engineering, Foshan University, Foshan, 528225, Guangdong Province, People's Republic of China
| | - Dixi Yu
- College of Life Sciences and Engineering, Foshan University, Foshan, 528225, Guangdong Province, People's Republic of China
| | - Limei Qin
- College of Life Sciences and Engineering, Foshan University, Foshan, 528225, Guangdong Province, People's Republic of China.
| | - Keshan Zhang
- College of Life Sciences and Engineering, Foshan University, Foshan, 528225, Guangdong Province, People's Republic of China.
| |
Collapse
|
3
|
Otálora-Otálora BA, Payán-Gómez C, López-Rivera JJ, Pedroza-Aconcha NB, Aristizábal-Guzmán C, Isaza-Ruget MA, Álvarez-Moreno CA. Global transcriptomic network analysis of the crosstalk between microbiota and cancer-related cells in the oral-gut-lung axis. Front Cell Infect Microbiol 2024; 14:1425388. [PMID: 39228892 PMCID: PMC11368877 DOI: 10.3389/fcimb.2024.1425388] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Accepted: 07/15/2024] [Indexed: 09/05/2024] Open
Abstract
Background The diagnosis and treatment of lung, colon, and gastric cancer through the histologic characteristics and genomic biomarkers have not had a strong impact on the mortality rates of the top three global causes of death by cancer. Methods Twenty-five transcriptomic analyses (10 lung cancer, 10 gastric cancer, and 5 colon cancer datasets) followed our own bioinformatic pipeline based on the utilization of specialized libraries from the R language and DAVID´s gene enrichment analyses to identify a regulatory metafirm network of transcription factors and target genes common in every type of cancer, with experimental evidence that supports its relationship with the unlocking of cell phenotypic plasticity for the acquisition of the hallmarks of cancer during the tumoral process. The network's regulatory functional and signaling pathways might depend on the constant crosstalk with the microbiome network established in the oral-gut-lung axis. Results The global transcriptomic network analysis highlighted the impact of transcription factors (SOX4, TCF3, TEAD4, ETV4, and FOXM1) that might be related to stem cell programming and cancer progression through the regulation of the expression of genes, such as cancer-cell membrane receptors, that interact with several microorganisms, including human T-cell leukemia virus 1 (HTLV-1), the human papilloma virus (HPV), the Epstein-Barr virus (EBV), and SARS-CoV-2. These interactions can trigger the MAPK, non-canonical WNT, and IFN signaling pathways, which regulate key transcription factor overexpression during the establishment and progression of lung, colon, and gastric cancer, respectively, along with the formation of the microbiome network. Conclusion The global transcriptomic network analysis highlights the important interaction between key transcription factors in lung, colon, and gastric cancer, which regulates the expression of cancer-cell membrane receptors for the interaction with the microbiome network during the tumorigenic process.
Collapse
Affiliation(s)
| | - César Payán-Gómez
- Dirección Académica, Universidad Nacional de Colombia, Sede de La Paz, La Paz, Colombia
| | - Juan Javier López-Rivera
- Grupo de Investigación INPAC, Specialized Laboratory, Clinica Universitaria Colombia, Clínica Colsanitas S.A., Bogotá, Colombia
| | | | - Claudia Aristizábal-Guzmán
- Grupo de Investigación INPAC, Unidad de Investigación, Fundación Universitaria Sanitas, Bogotá, Colombia
| | - Mario Arturo Isaza-Ruget
- Keralty, Sanitas International Organization, Grupo de Investigación INPAC, Fundación Universitaria Sanitas, Bogotá, Colombia
| | | |
Collapse
|
4
|
Yang Q, Meng D, Zhang Q, Wang J. Advances in the role of resveratrol and its mechanism of action in common gynecological tumors. Front Pharmacol 2024; 15:1417532. [PMID: 39086397 PMCID: PMC11288957 DOI: 10.3389/fphar.2024.1417532] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Accepted: 06/26/2024] [Indexed: 08/02/2024] Open
Abstract
The incidence of common gynecological malignancies remains high, with current treatments facing multiple limitations and adverse effects. Thus, continuing the search for safe and effective oncologic treatment strategies continues. Resveratrol (RES), a natural non-flavonoid polyphenolic compound, is widely found in various plants and fruits, such as grapes, Reynoutria japonica Houtt., peanuts, and berries. RES possesses diverse biological properties, including neuroprotective, antitumor, anti-inflammatory, and osteoporosis inhibition effects. Notably, RES is broadly applicable in antitumor therapy, particularly for treating gynecological tumors (cervical, endometrial, and ovarian carcinomas). RES exerts antitumor effects by promoting tumor cell apoptosis, inhibiting cell proliferation, invasion, and metastasis, regulating tumor cell autophagy, and enhancing the efficacy of antitumor drugs while minimizing their toxic side effects. However, comprehensive reviews on the role of RES in combating gynecological tumors and its mechanisms of action are lacking. This review aims to fill this gap by examining the RES antitumor mechanisms of action in gynecological tumors, providing valuable insights for clinical treatment.
Collapse
Affiliation(s)
- Qian Yang
- College of Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Dandan Meng
- College of Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Qingchen Zhang
- First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Jin Wang
- College of Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| |
Collapse
|
5
|
Makinwa Y, Luo Y, Musich PR, Zou Y. Canonical and Noncanonical Functions of the BH3 Domain Protein Bid in Apoptosis, Oncogenesis, Cancer Therapeutics, and Aging. Cancers (Basel) 2024; 16:2199. [PMID: 38927905 PMCID: PMC11202167 DOI: 10.3390/cancers16122199] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Revised: 06/05/2024] [Accepted: 06/09/2024] [Indexed: 06/28/2024] Open
Abstract
Effective cancer therapy with limited adverse effects is a major challenge in the medical field. This is especially complicated by the development of acquired chemoresistance. Understanding the mechanisms that underlie these processes remains a major effort in cancer research. In this review, we focus on the dual role that Bid protein plays in apoptotic cell death via the mitochondrial pathway, in oncogenesis and in cancer therapeutics. The BH3 domain in Bid and the anti-apoptotic mitochondrial proteins (Bcl-2, Bcl-XL, mitochondrial ATR) it associates with at the outer mitochondrial membrane provides us with a viable target in cancer therapy. We will discuss the roles of Bid, mitochondrial ATR, and other anti-apoptotic proteins in intrinsic apoptosis, exploring how their interaction sustains cellular viability despite the initiation of upstream death signals. The unexpected upregulation of this Bid protein in cancer cells can also be instrumental in explaining the mechanisms behind acquired chemoresistance. The stable protein associations at the mitochondria between tBid and anti-apoptotic mitochondrial ATR play a crucial role in maintaining the viability of cancer cells, suggesting a novel mechanism to induce cancer cell apoptosis by freeing tBid from the ATR associations at mitochondria.
Collapse
Affiliation(s)
- Yetunde Makinwa
- Department of Cell and Cancer Biology, University of Toledo College of Medicine and Life Sciences, Toledo, OH 43614, USA; (Y.M.); (Y.L.)
| | - Yibo Luo
- Department of Cell and Cancer Biology, University of Toledo College of Medicine and Life Sciences, Toledo, OH 43614, USA; (Y.M.); (Y.L.)
| | - Phillip R. Musich
- Department of Biomedical Sciences, Quillen College of Medicine, East Tennessee State University, Johnson City, TN 37614, USA;
| | - Yue Zou
- Department of Cell and Cancer Biology, University of Toledo College of Medicine and Life Sciences, Toledo, OH 43614, USA; (Y.M.); (Y.L.)
| |
Collapse
|
6
|
Suraweera CD, Espinoza B, Hinds MG, Kvansakul M. Mastering Death: The Roles of Viral Bcl-2 in dsDNA Viruses. Viruses 2024; 16:879. [PMID: 38932171 PMCID: PMC11209288 DOI: 10.3390/v16060879] [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: 05/07/2024] [Revised: 05/25/2024] [Accepted: 05/27/2024] [Indexed: 06/28/2024] Open
Abstract
Proteins of the Bcl-2 family regulate cellular fate via multiple mechanisms including apoptosis, autophagy, senescence, metabolism, inflammation, redox homeostasis, and calcium flux. There are several regulated cell death (RCD) pathways, including apoptosis and autophagy, that use distinct molecular mechanisms to elicit the death response. However, the same proteins/genes may be deployed in multiple biochemical pathways. In apoptosis, Bcl-2 proteins control the integrity of the mitochondrial outer membrane (MOM) by regulating the formation of pores in the MOM and apoptotic cell death. A number of prosurvival genes populate the genomes of viruses including those of the pro-survival Bcl-2 family. Viral Bcl-2 proteins are sequence and structural homologs of their cellular counterparts and interact with cellular proteins in apoptotic and autophagic pathways, potentially allowing them to modulate these pathways and determine cellular fate.
Collapse
Affiliation(s)
- Chathura D. Suraweera
- Genome Sciences and Cancer Division, The John Curtin School of Medical Research, Australian National University, Canberra 2601, Australia;
| | - Benjamin Espinoza
- Department of Biochemistry and Chemistry, La Trobe University, Melbourne, VIC 3086, Australia;
| | - Mark G. Hinds
- Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Parkville, VIC 3052, Australia
| | - Marc Kvansakul
- Genome Sciences and Cancer Division, The John Curtin School of Medical Research, Australian National University, Canberra 2601, Australia;
| |
Collapse
|
7
|
Bristol JA, Nelson SE, Ohashi M, Casco A, Hayes M, Ranheim EA, Pawelski AS, Singh DR, Hodson DJ, Johannsen EC, Kenney SC. Latent Epstein-Barr virus infection collaborates with Myc over-expression in normal human B cells to induce Burkitt-like Lymphomas in mice. PLoS Pathog 2024; 20:e1012132. [PMID: 38620028 PMCID: PMC11045125 DOI: 10.1371/journal.ppat.1012132] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2023] [Revised: 04/25/2024] [Accepted: 03/18/2024] [Indexed: 04/17/2024] Open
Abstract
Epstein-Barr virus (EBV) is an important cause of human lymphomas, including Burkitt lymphoma (BL). EBV+ BLs are driven by Myc translocation and have stringent forms of viral latency that do not express either of the two major EBV oncoproteins, EBNA2 (which mimics Notch signaling) and LMP1 (which activates NF-κB signaling). Suppression of Myc-induced apoptosis, often through mutation of the TP53 (p53) gene or inhibition of pro-apoptotic BCL2L11 (BIM) gene expression, is required for development of Myc-driven BLs. EBV+ BLs contain fewer cellular mutations in apoptotic pathways compared to EBV-negative BLs, suggesting that latent EBV infection inhibits Myc-induced apoptosis. Here we use an EBNA2-deleted EBV virus (ΔEBNA2 EBV) to create the first in vivo model for EBV+ BL-like lymphomas derived from primary human B cells. We show that cord blood B cells infected with both ΔEBNA2 EBV and a Myc-expressing vector proliferate indefinitely on a CD40L/IL21 expressing feeder layer in vitro and cause rapid onset EBV+ BL-like tumors in NSG mice. These LMP1/EBNA2-negative Myc-driven lymphomas have wild type p53 and very low BIM, and express numerous germinal center B cell proteins (including TCF3, BACH2, Myb, CD10, CCDN3, and GCSAM) in the absence of BCL6 expression. Myc-induced activation of Myb mediates expression of many of these BL-associated proteins. We demonstrate that Myc blocks LMP1 expression both by inhibiting expression of cellular factors (STAT3 and Src) that activate LMP1 transcription and by increasing expression of proteins (DNMT3B and UHRF1) known to enhance DNA methylation of the LMP1 promoters in human BLs. These results show that latent EBV infection collaborates with Myc over-expression to induce BL-like human B-cell lymphomas in mice. As NF-κB signaling retards the growth of EBV-negative BLs, Myc-mediated repression of LMP1 may be essential for latent EBV infection and Myc translocation to collaboratively induce human BLs.
Collapse
Affiliation(s)
- Jillian A. Bristol
- Department of Oncology, McArdle Laboratory for Cancer Research, School of Medicine and Public Health, University of Wisconsin Madison, Madison, Wisconsin, United States of America
| | - Scott E. Nelson
- Department of Oncology, McArdle Laboratory for Cancer Research, School of Medicine and Public Health, University of Wisconsin Madison, Madison, Wisconsin, United States of America
| | - Makoto Ohashi
- Department of Oncology, McArdle Laboratory for Cancer Research, School of Medicine and Public Health, University of Wisconsin Madison, Madison, Wisconsin, United States of America
| | - Alejandro Casco
- Department of Oncology, McArdle Laboratory for Cancer Research, School of Medicine and Public Health, University of Wisconsin Madison, Madison, Wisconsin, United States of America
| | - Mitchell Hayes
- Department of Oncology, McArdle Laboratory for Cancer Research, School of Medicine and Public Health, University of Wisconsin Madison, Madison, Wisconsin, United States of America
| | - Erik A. Ranheim
- Department of Pathology, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Abigail S. Pawelski
- Department of Oncology, McArdle Laboratory for Cancer Research, School of Medicine and Public Health, University of Wisconsin Madison, Madison, Wisconsin, United States of America
| | - Deo R. Singh
- Department of Oncology, McArdle Laboratory for Cancer Research, School of Medicine and Public Health, University of Wisconsin Madison, Madison, Wisconsin, United States of America
| | - Daniel J. Hodson
- Wellcome-MRC Cambridge Stem Cell Institute, University of Cambridge, Cambridge, United Kingdom
| | - Eric C. Johannsen
- Department of Oncology, McArdle Laboratory for Cancer Research, School of Medicine and Public Health, University of Wisconsin Madison, Madison, Wisconsin, United States of America
- Department of Medicine, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Shannon C. Kenney
- Department of Oncology, McArdle Laboratory for Cancer Research, School of Medicine and Public Health, University of Wisconsin Madison, Madison, Wisconsin, United States of America
- Department of Medicine, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| |
Collapse
|
8
|
Yang Q, Meng D, Zhang Q, Wang J. Advances in research on the anti-tumor mechanism of Astragalus polysaccharides. Front Oncol 2024; 14:1334915. [PMID: 38515577 PMCID: PMC10955345 DOI: 10.3389/fonc.2024.1334915] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2023] [Accepted: 02/22/2024] [Indexed: 03/23/2024] Open
Abstract
The dry root of the soybean plant Astragalus membranaceus (Fisch) Bge. var. mongholicus (Bge) Hsiao or A. membranaceus (Fisch) Bge, Astragali Radix (AR) has a long medicinal history. Astragalus polysaccharide (APS), the natural macromolecule that exhibits immune regulatory, anti-inflammatory, anti-tumor, and other pharmacological activities, is an important active ingredient extracted from AR. Recently, APS has been increasingly used in cancer therapy owing to its anti-tumor ability as it prevents the progression of prostate, liver, cervical, ovarian, and non-small-cell lung cancer by suppressing tumor cell growth and invasion and enhancing apoptosis. In addition, APS enhances the sensitivity of tumors to antineoplastic agents and improves the body's immunity. This macromolecule has prospects for broad application in tumor therapy through various pathways. In this article, we present the latest progress in the research on the anti-tumor effects of APS and its underlying mechanisms, aiming to provide novel theoretical support and reference for its use in cancer therapy.
Collapse
Affiliation(s)
| | | | - Qinyuan Zhang
- School of Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Jin Wang
- School of Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| |
Collapse
|
9
|
Cheng C, Yuan Y, Yuan F, Li X. Acute kidney injury: exploring endoplasmic reticulum stress-mediated cell death. Front Pharmacol 2024; 15:1308733. [PMID: 38434710 PMCID: PMC10905268 DOI: 10.3389/fphar.2024.1308733] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2023] [Accepted: 01/31/2024] [Indexed: 03/05/2024] Open
Abstract
Acute kidney injury (AKI) is a global health problem, given its substantial morbidity and mortality rates. A better understanding of the mechanisms and factors contributing to AKI has the potential to guide interventions aimed at mitigating the risk of AKI and its subsequent unfavorable outcomes. Endoplasmic reticulum stress (ERS) is an intrinsic protective mechanism against external stressors. ERS occurs when the endoplasmic reticulum (ER) cannot deal with accumulated misfolded proteins completely. Excess ERS can eventually cause pathological reactions, triggering various programmed cell death (autophagy, ferroptosis, apoptosis, pyroptosis). This article provides an overview of the latest research progress in deciphering the interaction between ERS and different programmed cell death. Additionally, the report consolidates insights into the roles of ERS in AKI and highlights the potential avenues for targeting ERS as a treatment direction toward for AKI.
Collapse
Affiliation(s)
- Cong Cheng
- Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Yuan Yuan
- Department of Emergency, Changsha Hospital of Traditional Chinese Medicine (Changsha Eighth Hospital), Changsha, Hunan, China
| | - Fang Yuan
- Department of Pharmacy, The Third Hospital of Changsha, Changsha, Hunan, China
- Hunan Provincial Key Laboratory of Anti-Resistance Microbial Drugs, Changsha, Hunan, China
| | - Xin Li
- Department of Pharmacy, The Third Hospital of Changsha, Changsha, Hunan, China
- Hunan Provincial Key Laboratory of Anti-Resistance Microbial Drugs, Changsha, Hunan, China
| |
Collapse
|
10
|
Silva JDM, Alves CEDC, Pontes GS. Epstein-Barr virus: the mastermind of immune chaos. Front Immunol 2024; 15:1297994. [PMID: 38384471 PMCID: PMC10879370 DOI: 10.3389/fimmu.2024.1297994] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Accepted: 01/23/2024] [Indexed: 02/23/2024] Open
Abstract
The Epstein-Barr virus (EBV) is a ubiquitous human pathogen linked to various diseases, including infectious mononucleosis and multiple types of cancer. To control and eliminate EBV, the host's immune system deploys its most potent defenses, including pattern recognition receptors, Natural Killer cells, CD8+ and CD4+ T cells, among others. The interaction between EBV and the human immune system is complex and multifaceted. EBV employs a variety of strategies to evade detection and elimination by both the innate and adaptive immune systems. This demonstrates EBV's mastery of navigating the complexities of the immunological landscape. Further investigation into these complex mechanisms is imperative to advance the development of enhanced therapeutic approaches with heightened efficacy. This review provides a comprehensive overview of various mechanisms known to date, employed by the EBV to elude the immune response, while establishing enduring latent infections or instigate its lytic replication.
Collapse
Affiliation(s)
- Jean de Melo Silva
- Laboratory of Virology and Immunology, National Institute of Amazonian Research (INPA), Manaus, AM, Brazil
- Post-Graduate Program in Basic and Applied Immunology, Institute of Biological Science, Federal University of Amazonas, Manaus, AM, Brazil
| | | | - Gemilson Soares Pontes
- Laboratory of Virology and Immunology, National Institute of Amazonian Research (INPA), Manaus, AM, Brazil
- Post-Graduate Program in Basic and Applied Immunology, Institute of Biological Science, Federal University of Amazonas, Manaus, AM, Brazil
| |
Collapse
|
11
|
Bednarska K, Chowdhury R, Tobin JWD, Swain F, Keane C, Boyle S, Khanna R, Gandhi MK. Epstein-Barr virus-associated lymphomas decoded. Br J Haematol 2024; 204:415-433. [PMID: 38155519 DOI: 10.1111/bjh.19255] [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: 07/28/2023] [Revised: 11/15/2023] [Accepted: 11/29/2023] [Indexed: 12/30/2023]
Abstract
Epstein-Barr virus (EBV)-associated lymphomas cover a range of histological B- and T-cell non-Hodgkin and Hodgkin lymphoma subtypes. The role of EBV on B-cell malignant pathogenesis and its impact on the tumour microenvironment are intriguing but incompletely understood. Both the International Consensus Classification (ICC) and 5th Edition of the World Health Organization (WHO-HAEM5) proposals give prominence to the distinct clinical, prognostic, genetic and tumour microenvironmental features of EBV in lymphoproliferative disorders. There have been major advances in our biological understanding, in how to harness features of EBV and its host immune response for targeted therapy, and in using EBV as a method to monitor disease response. In this article, we showcase the latest developments and how they may be integrated to stimulate new and innovative approaches for further lines of investigation and therapy.
Collapse
Affiliation(s)
- Karolina Bednarska
- Mater Research Institute, University of Queensland, Brisbane, Queensland, Australia
| | - Rakin Chowdhury
- Frazer Institute, University of Queensland, Brisbane, Queensland, Australia
- Department of Haematology, Princess Alexandra Hospital, Brisbane, Queensland, Australia
| | - Joshua W D Tobin
- Mater Research Institute, University of Queensland, Brisbane, Queensland, Australia
- Department of Haematology, Princess Alexandra Hospital, Brisbane, Queensland, Australia
| | - Fiona Swain
- Frazer Institute, University of Queensland, Brisbane, Queensland, Australia
- Department of Haematology, Princess Alexandra Hospital, Brisbane, Queensland, Australia
| | - Colm Keane
- Frazer Institute, University of Queensland, Brisbane, Queensland, Australia
- Department of Haematology, Princess Alexandra Hospital, Brisbane, Queensland, Australia
| | - Stephen Boyle
- QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | - Rajiv Khanna
- QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | - Maher K Gandhi
- Mater Research Institute, University of Queensland, Brisbane, Queensland, Australia
- Department of Haematology, Princess Alexandra Hospital, Brisbane, Queensland, Australia
| |
Collapse
|
12
|
Abstract
Apoptosis, necroptosis, and pyroptosis are genetically programmed cell death mechanisms that eliminate obsolete, damaged, infected, and self-reactive cells. Apoptosis fragments cells in a manner that limits immune cell activation, whereas the lytic death programs of necroptosis and pyroptosis release proinflammatory intracellular contents. Apoptosis fine-tunes tissue architecture during mammalian development, promotes tissue homeostasis, and is crucial for averting cancer and autoimmunity. All three cell death mechanisms are deployed to thwart the spread of pathogens. Disabling regulators of cell death signaling in mice has revealed how excessive cell death can fuel acute or chronic inflammation. Here we review strategies for modulating cell death in the context of disease. For example, BCL-2 inhibitor venetoclax, an inducer of apoptosis, is approved for the treatment of certain hematologic malignancies. By contrast, inhibition of RIPK1, NLRP3, GSDMD, or NINJ1 to limit proinflammatory cell death and/or the release of large proinflammatory molecules from dying cells may benefit patients with inflammatory diseases.
Collapse
Affiliation(s)
- Nobuhiko Kayagaki
- Physiological Chemistry Department, Genentech, South San Francisco, California, USA;
| | - Joshua D Webster
- Pathology Department, Genentech, South San Francisco, California, USA
| | - Kim Newton
- Physiological Chemistry Department, Genentech, South San Francisco, California, USA;
| |
Collapse
|
13
|
Sausen DG, Poirier MC, Spiers LM, Smith EN. Mechanisms of T cell evasion by Epstein-Barr virus and implications for tumor survival. Front Immunol 2023; 14:1289313. [PMID: 38179040 PMCID: PMC10764432 DOI: 10.3389/fimmu.2023.1289313] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Accepted: 11/27/2023] [Indexed: 01/06/2024] Open
Abstract
Epstein-Barr virus (EBV) is a prevalent oncogenic virus estimated to infect greater than 90% of the world's population. Following initial infection, it establishes latency in host B cells. EBV has developed a multitude of techniques to avoid detection by the host immune system and establish lifelong infection. T cells, as important contributors to cell-mediated immunity, make an attractive target for these immunoevasive strategies. Indeed, EBV has evolved numerous mechanisms to modulate T cell responses. For example, it can augment expression of programmed cell death ligand-1 (PD-L1), which inhibits T cell function, and downregulates the interferon response, which has a strong impact on T cell regulation. It also modulates interleukin secretion and can influence major histocompatibility complex (MHC) expression and presentation. In addition to facilitating persistent EBV infection, these immunoregulatory mechanisms have significant implications for evasion of the immune response by tumor cells. This review dissects the mechanisms through which EBV avoids detection by host T cells and discusses how these mechanisms play into tumor survival. It concludes with an overview of cancer treatments targeting T cells in the setting of EBV-associated malignancy.
Collapse
Affiliation(s)
- D. G. Sausen
- School of Medicine, Eastern Virginia Medical School, Norfolk, VA, United States
| | | | | | | |
Collapse
|
14
|
Morovati S, Mohammadi A, Masoudi R, Heidari AA, Asad Sangabi M. The power of mumps virus: Matrix protein activates apoptotic pathways in human colorectal cell lines. PLoS One 2023; 18:e0295819. [PMID: 38091318 PMCID: PMC10718445 DOI: 10.1371/journal.pone.0295819] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2023] [Accepted: 11/30/2023] [Indexed: 12/18/2023] Open
Abstract
New therapeutic approaches can significantly impact the control of colorectal cancer (CRC), which is increasing worldwide. In this study, we investigated the potential of targeting viral proteins to combat cancer cells. Specifically, we examined the anticancer potential of the matrix (M) protein of the mumps virus Hoshino strain in SW480 CRC cell lines. To begin, we individually transfected SW480 cells with pcDNA3 plasmids containing the mumps virus M gene. We then investigated the percentage of cell death, caspase activity, and the expression levels of genes involved in apoptosis pathways. Following this, we performed bioinformatics analysis on the M protein to identify any similarities with Bcl-2 family members and their viral homologs. Our diagnostic methods showed that treatment with the mumps M protein induced apoptosis and upregulated the expression and activity of pro-apoptotic proteins in SW480 CRC cells compared to the control and vector groups. Based on our bioinformatics studies, we proposed that the BH3 motif in the M protein may trigger apoptosis in CRC cells by interacting with cellular Bax. Overall, our study showed for the first time that the mumps virus M protein could be considered as a targeted treatment for CRC by inducing apoptotic pathways.
Collapse
Affiliation(s)
- Solmaz Morovati
- Department of Pathobiology, Division of Biotechnology, School of Veterinary Medicine, Shiraz University, Shiraz, Iran
| | - Ali Mohammadi
- Department of Pathobiology, Division of Virology, School of Veterinary Medicine, Shiraz University, Shiraz, Iran
| | - Ramin Masoudi
- Department of Pathobiology, Division of Biotechnology, School of Veterinary Medicine, Shiraz University, Shiraz, Iran
| | - Amir Ali Heidari
- Department of Clinical Sciences, Division of Aquatic Animal Health and Diseases, School of Veterinary Medicine, Shiraz University, Shiraz, Iran
| | - Mehdi Asad Sangabi
- Department of Pathobiology, Division of Virology, School of Veterinary Medicine, Shiraz University, Shiraz, Iran
| |
Collapse
|
15
|
Thomas OG, Olsson T. Mimicking the brain: Epstein-Barr virus and foreign agents as drivers of neuroimmune attack in multiple sclerosis. Front Immunol 2023; 14:1304281. [PMID: 38022632 PMCID: PMC10655090 DOI: 10.3389/fimmu.2023.1304281] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2023] [Accepted: 10/23/2023] [Indexed: 12/01/2023] Open
Abstract
T cells have an essential role in adaptive immunity against pathogens and cancer, but failure of thymic tolerance mechanisms can instead lead to escape of T cells with the ability to attack host tissues. Multiple sclerosis (MS) occurs when structures such as myelin and neurons in the central nervous system (CNS) are the target of autoreactive immune responses, resulting in lesions in the brain and spinal cord which cause varied and episodic neurological deficits. A role for autoreactive T cell and antibody responses in MS is likely, and mounting evidence implicates Epstein-Barr virus (EBV) in disease mechanisms. In this review we discuss antigen specificity of T cells involved in development and progression of MS. We examine the current evidence that these T cells can target multiple antigens such as those from pathogens including EBV and briefly describe other mechanisms through which viruses could affect disease. Unravelling the complexity of the autoantigen T cell repertoire is essential for understanding key events in the development and progression of MS, with wider implications for development of future therapies.
Collapse
Affiliation(s)
- Olivia G. Thomas
- Therapeutic Immune Design, Centre for Molecular Medicine, Department of Clinical Neuroscience, Karolinska Institute, Stockholm, Sweden
- Neuroimmunology Unit, Department of Clinical Neuroscience, Centre for Molecular Medicine, Karolinska Institute, Stockholm, Sweden
| | - Tomas Olsson
- Therapeutic Immune Design, Centre for Molecular Medicine, Department of Clinical Neuroscience, Karolinska Institute, Stockholm, Sweden
| |
Collapse
|
16
|
Yiu SPT, Zerbe C, Vanderwall D, Huttlin EL, Weekes MP, Gewurz BE. An Epstein-Barr virus protein interaction map reveals NLRP3 inflammasome evasion via MAVS UFMylation. Mol Cell 2023; 83:2367-2386.e15. [PMID: 37311461 PMCID: PMC10372749 DOI: 10.1016/j.molcel.2023.05.018] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Revised: 04/05/2023] [Accepted: 05/14/2023] [Indexed: 06/15/2023]
Abstract
Epstein-Barr virus (EBV) causes infectious mononucleosis, triggers multiple sclerosis, and is associated with 200,000 cancers/year. EBV colonizes the human B cell compartment and periodically reactivates, inducing expression of 80 viral proteins. However, much remains unknown about how EBV remodels host cells and dismantles key antiviral responses. We therefore created a map of EBV-host and EBV-EBV interactions in B cells undergoing EBV replication, uncovering conserved herpesvirus versus EBV-specific host cell targets. The EBV-encoded G-protein-coupled receptor BILF1 associated with MAVS and the UFM1 E3 ligase UFL1. Although UFMylation of 14-3-3 proteins drives RIG-I/MAVS signaling, BILF1-directed MAVS UFMylation instead triggered MAVS packaging into mitochondrial-derived vesicles and lysosomal proteolysis. In the absence of BILF1, EBV replication activated the NLRP3 inflammasome, which impaired viral replication and triggered pyroptosis. Our results provide a viral protein interaction network resource, reveal a UFM1-dependent pathway for selective degradation of mitochondrial cargo, and highlight BILF1 as a novel therapeutic target.
Collapse
Affiliation(s)
- Stephanie Pei Tung Yiu
- Division of Infectious Diseases, Brigham and Women's Hospital, 181 Longwood Avenue, Boston, MA 02115, USA; Harvard Graduate Program in Virology, Boston, MA 02115, USA; Center for Integrated Solutions to Infectious Diseases, Broad Institute and Harvard Medical School, Cambridge, MA 02115, USA; Department of Microbiology, Harvard Medical School, Boston, MA 02115, USA
| | - Cassie Zerbe
- Cambridge Institute for Medical Research, University of Cambridge, Hills Road, Cambridge CB2 0XY, UK
| | - David Vanderwall
- Department of Cell Biology, Harvard Medical School, Boston, MA 02115, USA
| | - Edward L Huttlin
- Department of Cell Biology, Harvard Medical School, Boston, MA 02115, USA
| | - Michael P Weekes
- Cambridge Institute for Medical Research, University of Cambridge, Hills Road, Cambridge CB2 0XY, UK.
| | - Benjamin E Gewurz
- Division of Infectious Diseases, Brigham and Women's Hospital, 181 Longwood Avenue, Boston, MA 02115, USA; Harvard Graduate Program in Virology, Boston, MA 02115, USA; Center for Integrated Solutions to Infectious Diseases, Broad Institute and Harvard Medical School, Cambridge, MA 02115, USA; Department of Microbiology, Harvard Medical School, Boston, MA 02115, USA.
| |
Collapse
|
17
|
Masoudi R, Mohammadi A, Morovati S, Heidari AA, Asad-Sangabi M. Induction of apoptosis in colorectal cancer cells by matrix protein of PPR virus as a novel anti-cancer agent. Int J Biol Macromol 2023:125536. [PMID: 37369256 DOI: 10.1016/j.ijbiomac.2023.125536] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Revised: 06/19/2023] [Accepted: 06/21/2023] [Indexed: 06/29/2023]
Abstract
Colorectal cancer (CRC) is a common and highly malignant neoplasm, ranking as the fourth most frequent cause of cancer-related deaths worldwide. Recently, non-human oncolytic viruses such as Peste des petits ruminants virus (PPRV) are considered as a potent candidate in the viral therapy of cancer. In the current study, the apoptotic effects of matrix (M) protein of PPRV was investigated on SW480 CRC cells. The M gene was cloned into the pcDNA™3.1/Hygro(+) expression vector and transfected into the cancer cells. The cytotoxic effects of the M protein on SW480 cells were confirmed using MTT assay. Furthermore, flow cytometry results showed that the M protein induces apoptosis in 91 % of CRC cells. Interestingly, the expression of the M gene in SW480 cells led to the up-regulation of genes including Bax, p53, and Caspase-9, as well as an increase in the Bax/Bcl-2 ratio. By using bioinformatics modeling, we hypothesized that the M protein could interact with Bax factor through its BH3-like motif and could further activate the intrinsic apoptosis pathway. Ultimately, this study provided the first evidence of the pro-apoptotic activity of PPRV M protein indicating its possible development as a promising novel anti-cancer agent.
Collapse
Affiliation(s)
- Ramin Masoudi
- Division of Biotechnology, Department of Pathobiology, School of Veterinary Medicine, Shiraz University, Shiraz, Iran
| | - Ali Mohammadi
- Division of Virology, Department of Pathobiology, School of Veterinary Medicine, Shiraz University, Shiraz, Iran.
| | - Solmaz Morovati
- Division of Biotechnology, Department of Pathobiology, School of Veterinary Medicine, Shiraz University, Shiraz, Iran
| | - Amir Ali Heidari
- Division of Aquatic Animal Health and Diseases, Department of Clinical Sciences, School of Veterinary Medicine, Shiraz University, Shiraz, Iran
| | - Mehdi Asad-Sangabi
- Division of Virology, Department of Pathobiology, School of Veterinary Medicine, Shiraz University, Shiraz, Iran
| |
Collapse
|
18
|
Tan H, Gong Y, Liu Y, Long J, Luo Q, Faleti OD, Lyu X. Advancing therapeutic strategies for Epstein-Barr virus-associated malignancies through lytic reactivation. Biomed Pharmacother 2023; 164:114916. [PMID: 37229802 DOI: 10.1016/j.biopha.2023.114916] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Revised: 05/17/2023] [Accepted: 05/18/2023] [Indexed: 05/27/2023] Open
Abstract
Epstein-Barr virus (EBV) is a widespread human herpes virus associated with lymphomas and epithelial cell cancers. It establishes two separate infection phases, latent and lytic, in the host. Upon infection of a new host cell, the virus activates several pathways, to induce the expression of lytic EBV antigens and the production of infectious virus particles. Although the carcinogenic role of latent EBV infection has been established, recent research suggests that lytic reactivation also plays a significant role in carcinogenesis. In this review, we summarize the mechanism of EBV reactivation and recent findings about the role of viral lytic antigens in tumor formation. In addition, we discuss the treatment of EBV-associated tumors with lytic activators and the targets that may be therapeutically effective in the future.
Collapse
Affiliation(s)
- Haiqi Tan
- Department of Laboratory Medicine, The Third Affiliated Hospital, Southern Medical University, Guangzhou 510630, China; The Third School of Clinical Medicine, Southern Medical University, Guangzhou 510630, China
| | - Yibing Gong
- Department of Laboratory Medicine, The Third Affiliated Hospital, Southern Medical University, Guangzhou 510630, China; The Third School of Clinical Medicine, Southern Medical University, Guangzhou 510630, China
| | - Yi Liu
- Department of Laboratory Medicine, The Third Affiliated Hospital, Southern Medical University, Guangzhou 510630, China; The Third School of Clinical Medicine, Southern Medical University, Guangzhou 510630, China
| | - Jingyi Long
- Department of Laboratory Medicine, The Third Affiliated Hospital, Southern Medical University, Guangzhou 510630, China; The Third School of Clinical Medicine, Southern Medical University, Guangzhou 510630, China
| | - Qingshuang Luo
- Department of Laboratory Medicine, The Third Affiliated Hospital, Southern Medical University, Guangzhou 510630, China; The Third School of Clinical Medicine, Southern Medical University, Guangzhou 510630, China
| | - Oluwasijibomi Damola Faleti
- Department of Laboratory Medicine, The Third Affiliated Hospital, Southern Medical University, Guangzhou 510630, China; The Third School of Clinical Medicine, Southern Medical University, Guangzhou 510630, China; Department of Applied Biology and Chemical Technology, The Hong Kong Polytechnic University, 999000, Hong Kong Special Administrative Region of China
| | - Xiaoming Lyu
- Department of Laboratory Medicine, The Third Affiliated Hospital, Southern Medical University, Guangzhou 510630, China; The Third School of Clinical Medicine, Southern Medical University, Guangzhou 510630, China.
| |
Collapse
|
19
|
Dorothea M, Xie J, Yiu SPT, Chiang AKS. Contribution of Epstein–Barr Virus Lytic Proteins to Cancer Hallmarks and Implications from Other Oncoviruses. Cancers (Basel) 2023; 15:cancers15072120. [PMID: 37046781 PMCID: PMC10093119 DOI: 10.3390/cancers15072120] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2023] [Revised: 03/27/2023] [Accepted: 03/31/2023] [Indexed: 04/05/2023] Open
Abstract
Epstein–Barr virus (EBV) is a prevalent human gamma-herpesvirus that infects the majority of the adult population worldwide and is associated with several lymphoid and epithelial malignancies. EBV displays a biphasic life cycle, namely, latent and lytic replication cycles, expressing a diversity of viral proteins. Among the EBV proteins being expressed during both latent and lytic cycles, the oncogenic roles of EBV lytic proteins are largely uncharacterized. In this review, the established contributions of EBV lytic proteins in tumorigenesis are summarized according to the cancer hallmarks displayed. We further postulate the oncogenic properties of several EBV lytic proteins by comparing the evolutionary conserved oncogenic mechanisms in other herpesviruses and oncoviruses.
Collapse
Affiliation(s)
- Mike Dorothea
- Department of Paediatrics and Adolescent Medicine, School of Clinical Medicine, The University of Hong Kong, Hong Kong, China
| | - Jia Xie
- Department of Paediatrics and Adolescent Medicine, School of Clinical Medicine, The University of Hong Kong, Hong Kong, China
| | - Stephanie Pei Tung Yiu
- Division of Infectious Diseases, Department of Medicine, Brigham and Women’s Hospital, 181 Longwood Avenue, Boston, MA 02115, USA
- Harvard Graduate Program in Virology, Boston, MA 02115, USA
| | - Alan Kwok Shing Chiang
- Department of Paediatrics and Adolescent Medicine, School of Clinical Medicine, The University of Hong Kong, Hong Kong, China
| |
Collapse
|
20
|
Duart G, Elazar A, Weinstein JY, Gadea-Salom L, Ortiz-Mateu J, Fleishman SJ, Mingarro I, Martinez-Gil L. Computational design of BclxL inhibitors that target transmembrane domain interactions. Proc Natl Acad Sci U S A 2023; 120:e2219648120. [PMID: 36881618 PMCID: PMC10089226 DOI: 10.1073/pnas.2219648120] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Accepted: 02/03/2023] [Indexed: 03/08/2023] Open
Abstract
Several methods have been developed to explore interactions among water-soluble proteins or regions of proteins. However, techniques to target transmembrane domains (TMDs) have not been examined thoroughly despite their importance. Here, we developed a computational approach to design sequences that specifically modulate protein-protein interactions in the membrane. To illustrate this method, we demonstrated that BclxL can interact with other members of the B cell lymphoma 2 (Bcl2) family through the TMD and that these interactions are required for BclxL control of cell death. Next, we designed sequences that specifically recognize and sequester the TMD of BclxL. Hence, we were able to prevent BclxL intramembrane interactions and cancel its antiapoptotic effect. These results advance our understanding of protein-protein interactions in membranes and provide a means to modulate them. Moreover, the success of our approach may trigger the development of a generation of inhibitors targeting interactions between TMDs.
Collapse
Affiliation(s)
- Gerard Duart
- Department of Biochemistry and Molecular Biology, Institut de Biotecnologia i Biomedicina, Universitat de València, Burjassot46100, Spain
| | - Assaf Elazar
- Department of Biomolecular Sciences, Weizmann Institute of Science, Rehovot76100, Israel
| | - Jonathan Y. Weinstein
- Department of Biomolecular Sciences, Weizmann Institute of Science, Rehovot76100, Israel
| | - Laura Gadea-Salom
- Department of Biochemistry and Molecular Biology, Institut de Biotecnologia i Biomedicina, Universitat de València, Burjassot46100, Spain
| | - Juan Ortiz-Mateu
- Department of Biochemistry and Molecular Biology, Institut de Biotecnologia i Biomedicina, Universitat de València, Burjassot46100, Spain
| | - Sarel J. Fleishman
- Department of Biomolecular Sciences, Weizmann Institute of Science, Rehovot76100, Israel
| | - Ismael Mingarro
- Department of Biochemistry and Molecular Biology, Institut de Biotecnologia i Biomedicina, Universitat de València, Burjassot46100, Spain
| | - Luis Martinez-Gil
- Department of Biochemistry and Molecular Biology, Institut de Biotecnologia i Biomedicina, Universitat de València, Burjassot46100, Spain
| |
Collapse
|
21
|
Zhang H, Song Y, Yang Y, Gao Z, Song Z, Wang W. Effect of Fas and Bcl-2 DNA Damages Response Expression in Stem Cells on Apoptosis of Nucleus Pulposus of Intervertebral Disc. Stem Cells Int 2023; 2023:8103595. [PMID: 36818160 PMCID: PMC9934980 DOI: 10.1155/2023/8103595] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Revised: 10/16/2022] [Accepted: 11/24/2022] [Indexed: 02/11/2023] Open
Abstract
The nucleus pulposus is an elastic jelly composed of crisscross fibrous reticular structures, namely, chondrocytes and proteoglycan mucoid matrix. Embryo and adult SC can resist the accumulation of genetic damage and repair them through various DNA repair mechanisms, thus preventing them from spreading to daughter cells. Fresh medullary tissue was fixed with 10% formaldehyde solution, embedded in paraffin, and sectioned at 4 m. The nucleus pulposus was stained with HE, and its degeneration was observed under light microscope. The average apoptotic index (AI) of 20 denatured nuclei was 50.230, the percentage of Fas-positive cells was 74.255%, and the percentage of Bcl-2-positive cells was 55.370%. The average apoptotic index (AI) was 28.317. The percentage of Fas-positive cells, Fas protein-positive cells, and Bcl-2 protein-positive cells in six normal nuclei was 41.717%, 41.717%, and 27.167%, respectively. The average AI value, Fas protein expression, and Bcl-2 protein expression in the two groups were significantly different (P < 0.05).
Collapse
Affiliation(s)
- Hui Zhang
- The First School of Clinical Medicine of Lanzhou University, The First Hospital of Lanzhou University, Lanzhou, 730000 Gansu, China
- Gansu Provincial Hospital, Lanzhou, 730000 Gansu, China
| | - Yuxin Song
- Gansu Provincial Hospital, Lanzhou, 730000 Gansu, China
| | - Yang Yang
- Gansu Provincial Hospital, Lanzhou, 730000 Gansu, China
| | - Zhao Gao
- The First School of Clinical Medicine of Lanzhou University, The First Hospital of Lanzhou University, Lanzhou, 730000 Gansu, China
| | - Zhengdong Song
- The First School of Clinical Medicine of Lanzhou University, The First Hospital of Lanzhou University, Lanzhou, 730000 Gansu, China
| | - Wenji Wang
- The First School of Clinical Medicine of Lanzhou University, The First Hospital of Lanzhou University, Lanzhou, 730000 Gansu, China
| |
Collapse
|
22
|
The lytic phase of Epstein-Barr virus plays an important role in tumorigenesis. Virus Genes 2023; 59:1-12. [PMID: 36242711 DOI: 10.1007/s11262-022-01940-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2022] [Accepted: 10/02/2022] [Indexed: 01/13/2023]
Abstract
Epstein-Barr virus (EBV) is a recognized oncogenic virus that is related to the occurrence of lymphoma, nasopharyngeal carcinoma (NPC), and approximately 10% of gastric cancer (GC). EBV is a herpesvirus, and like other herpesviruses, EBV has a biphasic infection mode made up of latent and lytic infections. It has been established that latent infection promotes tumorigenesis in previous research, but in recent years, there has been new evidence that suggests that the lytic infection mode could also promote tumorigenesis. In this review, we mainly discuss the contribution of the EBV lytic phase to tumorigenesis, and graphically illustrate their relationship in detail. In addition, we described the relationship between the lytic cycle of EBV and autophagy. Finally, we also preliminarily explored the influence of the tumorigenesis effect of the EBV lytic phase on the future treatment of EBV-associated tumors.
Collapse
|
23
|
Cheragh M, Sadeghizadeh M, Pouriayevali MH, Parsania M. Dendrosomal nanocurcumin prevents EBV-associated cell transformation by targeting the lytic cycle genes of the Epstein-Barr virus in the generation of lymphoblastoid cell line. IRANIAN JOURNAL OF BASIC MEDICAL SCIENCES 2023; 26:1220-1226. [PMID: 37736518 PMCID: PMC10510484 DOI: 10.22038/ijbms.2023.69839.15199] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Accepted: 05/15/2023] [Indexed: 09/23/2023]
Abstract
Objectives Targeting the lytic cycle of the Epstein-Barr virus (EBV) has been considered a new treatment strategy for malignancies caused by this virus. This study aimed to investigate the effect of Dendrosomal NanoCurcumin (DNC) to prevent cell transformation and inhibit the expression of viral lytic gene expression in the generation of lymphoblastoid cell line (LCL). Materials and Methods Cell viability of LCLs and PBMCs was performed by MTT assay, and flow cytometry (Annexin/PI) was used for evaluation of apoptosis. CD markers on the surface of generated LCL (CD19) cells were examined for cell validation. The effect of DNC on transformation was evaluated by examining cell morphology and determining the expression level of lytic genes BZLF1, Zta, BHRF1, and BRLF1 of EBV using Real-time PCR. Student's t-test was used for statistical analysis. Results The MTT assay showed that DNC can inhibit the proliferation of LCL in a dose-dependent manner. The 50% cytotoxic concentration (CC50) of DNC and curcumin for LCL was determined 38.8 µg/ml and 75 µg/ml, respectively after 72 hr. Also, Real-time PCR data analysis showed that DNC in 30 µg/ml concentration significantly inhibited cell transformation in the LCL and significantly reduced viral lytic genes such as BZLF1, Zta, BHRF1, and BRLF1expression compared to control. Conclusion Overall, these findings show that DNC reduces the expression of the viral lytic cycle genes and also the induction of cell apoptosis and finally prevents the generation of LCL.
Collapse
Affiliation(s)
- Mahboobeh Cheragh
- Department of Microbiology, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Majid Sadeghizadeh
- Department of Genetics, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran
| | - Mohammad Hassan Pouriayevali
- Department of Arboviruses and Viral Hemorrhagic Fevers (National Reference Laboratory), Pasteur Institute of Iran, Tehran, Iran
| | - Masoud Parsania
- Department of Microbiology, Faculty of Medicine, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
- Medical Genomics Research Center, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| |
Collapse
|
24
|
Death Receptor DR5 as a Proviral Factor for Viral Entry and Replication of Coronavirus PEDV. Viruses 2022; 14:v14122724. [PMID: 36560727 PMCID: PMC9783156 DOI: 10.3390/v14122724] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Revised: 11/29/2022] [Accepted: 11/30/2022] [Indexed: 12/12/2022] Open
Abstract
Porcine epidemic diarrhea virus (PEDV), a member of Coronaviridae, causes high mortality in newborn piglets, and has caused significant economic losses in the pig industry. PEDV infection can induce apoptosis, both caspase-dependent and caspase-independent, but the details of apoptosis remain clarified. This study investigated the effect of death receptor DR5 on PEDV infection and its relationship with PEDV-induced apoptosis. We found that DR5 knockdown reduced viral mRNA and protein levels of PEDV, and the viral titer decreased from 104.5 TCID50 to 103.4 TCID50 at 12 hpi. Overexpression of DR5 significantly increased the viral titer. Further studies showed that DR5 facilitates viral replication by regulating caspase-8-dependent apoptosis, and the knockdown of DR5 significantly reduced PEDV-induced apoptosis. Interestingly, we detected a biphasic upregulation expression of DR5 in both Vero cells and piglets in response to PEDV infection. We found that DR5 also facilitates viral entry of PEDV, especially, incubation with DR5 antibody can reduce the PEDV binding to Vero cells. Our study improves the understanding of the mechanism by which PEDV induces apoptosis and provides new insights into the biological function of DR5 in PEDV infection.
Collapse
|
25
|
O’Leary CA, Tompkins VS, Rouse WB, Nam G, Moss W. Thermodynamic and structural characterization of an EBV infected B-cell lymphoma transcriptome. NAR Genom Bioinform 2022; 4:lqac082. [PMID: 36285286 PMCID: PMC9585548 DOI: 10.1093/nargab/lqac082] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Revised: 09/30/2022] [Accepted: 10/06/2022] [Indexed: 11/12/2022] Open
Abstract
Epstein-Barr virus (EBV) is a widely prevalent human herpes virus infecting over 95% of all adults and is associated with a variety of B-cell cancers and induction of multiple sclerosis. EBV accomplishes this in part by expression of coding and noncoding RNAs and alteration of the host cell transcriptome. To better understand the structures which are forming in the viral and host transcriptomes of infected cells, the RNA structure probing technique Structure-seq2 was applied to the BJAB-B1 cell line (an EBV infected B-cell lymphoma). This resulted in reactivity profiles and secondary structural analyses for over 10000 human mRNAs and lncRNAs, along with 19 lytic and latent EBV transcripts. We report in-depth structural analyses for the human MYC mRNA and the human lncRNA CYTOR. Additionally, we provide a new model for the EBV noncoding RNA EBER2 and provide the first reported model for the EBV tandem terminal repeat RNA. In-depth thermodynamic and structural analyses were carried out with the motif discovery tool ScanFold and RNAfold prediction tool; subsequent covariation analyses were performed on resulting models finding various levels of support. ScanFold results for all analyzed transcripts are made available for viewing and download on the user-friendly RNAStructuromeDB.
Collapse
Affiliation(s)
- Collin A O’Leary
- Roy J. Carver Department of Biophysics, Biochemistry and Molecular Biology, Iowa State University, Ames, IA 50011, USA
| | - Van S Tompkins
- Roy J. Carver Department of Biophysics, Biochemistry and Molecular Biology, Iowa State University, Ames, IA 50011, USA
| | - Warren B Rouse
- Roy J. Carver Department of Biophysics, Biochemistry and Molecular Biology, Iowa State University, Ames, IA 50011, USA
| | - Gijong Nam
- Roy J. Carver Department of Biophysics, Biochemistry and Molecular Biology, Iowa State University, Ames, IA 50011, USA
| | - Walter N Moss
- Roy J. Carver Department of Biophysics, Biochemistry and Molecular Biology, Iowa State University, Ames, IA 50011, USA
| |
Collapse
|
26
|
Functional Implications of Epstein-Barr Virus Lytic Genes in Carcinogenesis. Cancers (Basel) 2022; 14:cancers14235780. [PMID: 36497262 PMCID: PMC9740547 DOI: 10.3390/cancers14235780] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Revised: 11/19/2022] [Accepted: 11/21/2022] [Indexed: 11/27/2022] Open
Abstract
Epstein-Barr virus (EBV) is associated with a diverse range of tumors of both lymphoid and epithelial origin. Similar to other herpesviruses, EBV displays a bipartite life cycle consisting of latent and lytic phases. Current dogma indicates that the latent genes are key drivers in the pathogenesis of EBV-associated cancers, while the lytic genes are primarily responsible for viral transmission. In recent years, evidence has emerged to show that the EBV lytic phase also plays an important role in EBV tumorigenesis, and the expression of EBV lytic genes is frequently detected in tumor tissues and cell lines. The advent of next generation sequencing has allowed the comprehensive profiling of EBV gene expression, and this has revealed the consistent expression of several lytic genes across various types of EBV-associated cancers. In this review, we provide an overview of the functional implications of EBV lytic gene expression to the oncogenic process and discuss possible avenues for future investigations.
Collapse
|
27
|
Suraweera CD, Hinds MG, Kvansakul M. Crystal Structures of Epstein-Barr Virus Bcl-2 Homolog BHRF1 Bound to Bid and Puma BH3 Motif Peptides. Viruses 2022; 14:v14102222. [PMID: 36298777 PMCID: PMC9609553 DOI: 10.3390/v14102222] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Revised: 09/27/2022] [Accepted: 10/04/2022] [Indexed: 11/09/2022] Open
Abstract
Apoptosis is a powerful defense mechanism used by multicellular organisms to counteract viral infection. In response to premature host cell suicide, viruses have evolved numerous countermeasures to ensure cell viability to optimize their replication by encoding proteins homologous in structure and function to cellular pro-survival Bcl-2 proteins. Epstein-Barr virus (EBV), a member of the Gammaherpesviridae, encodes the Bcl-2 homolog BHRF1, a potent inhibitor of Bcl-2-mediated apoptosis. BHRF1 acts by directly targeting Bid and Puma, two proapoptotic proteins of the Bcl-2 family. Here, we determined the crystal structures of BHRF1 bound to peptides spanning the Bcl-2 binding motifs (Bcl-2 homology 3 motif, BH3) of Bid and Puma. BHRF1 engages BH3 peptides using the canonical ligand-binding groove of its Bcl-2 fold and maintains a salt bridge between an Arg residue with a conserved Asp residue in the BH3 motif mimicking the canonical ionic interaction seen in host Bcl-2:BH3 motif complexes. Furthermore, both Bid and Puma utilize a fifth binding pocket in the canonical ligand binding groove of BHRF1 to provide an additional hydrophobic interaction distinct from the interactions previously seen with Bak and Bim. These findings provide a structural basis for EBV-mediated suppression of host cell apoptosis and reveal the flexibility of virus encoded Bcl-2 proteins in mimicking key interactions from the endogenous host signaling pathways.
Collapse
Affiliation(s)
- Chathura D. Suraweera
- Department of Biochemistry & Chemistry, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, VIC 3086, Australia
| | - Mark G. Hinds
- Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Parkville, VIC 3010, Australia
- Correspondence: (M.G.H.); (M.K.)
| | - Marc Kvansakul
- Department of Biochemistry & Chemistry, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, VIC 3086, Australia
- Correspondence: (M.G.H.); (M.K.)
| |
Collapse
|
28
|
Wyżewski Z, Mielcarska MB, Gregorczyk-Zboroch KP, Myszka A. Virus-Mediated Inhibition of Apoptosis in the Context of EBV-Associated Diseases: Molecular Mechanisms and Therapeutic Perspectives. Int J Mol Sci 2022; 23:ijms23137265. [PMID: 35806271 PMCID: PMC9266970 DOI: 10.3390/ijms23137265] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2022] [Revised: 06/25/2022] [Accepted: 06/27/2022] [Indexed: 12/10/2022] Open
Abstract
Epstein-Barr virus (EBV), the representative of the Herpesviridae family, is a pathogen extensively distributed in the human population. One of its most characteristic features is the capability to establish latent infection in the host. The infected cells serve as a sanctuary for the dormant virus, and therefore their desensitization to apoptotic stimuli is part of the viral strategy for long-term survival. For this reason, EBV encodes a set of anti-apoptotic products. They may increase the viability of infected cells and enhance their resistance to chemotherapy, thereby contributing to the development of EBV-associated diseases, including Burkitt’s lymphoma (BL), Hodgkin’s lymphoma (HL), gastric cancer (GC), nasopharyngeal carcinoma (NPC) and several other malignancies. In this paper, we have described the molecular mechanism of anti-apoptotic actions of a set of EBV proteins. Moreover, we have reviewed the pro-survival role of non-coding viral transcripts: EBV-encoded small RNAs (EBERs) and microRNAs (miRNAs), in EBV-carrying malignant cells. The influence of EBV on the expression, activity and/or intracellular distribution of B-cell lymphoma 2 (Bcl-2) protein family members, has been presented. Finally, we have also discussed therapeutic perspectives of targeting viral anti-apoptotic products or their molecular partners.
Collapse
Affiliation(s)
- Zbigniew Wyżewski
- Institute of Biological Sciences, Cardinal Stefan Wyszyński University, Dewajtis 5, 01-815 Warsaw, Poland;
- Correspondence: ; Tel.: +48-728-208-338
| | - Matylda Barbara Mielcarska
- Institute of Veterinary Medicine, Warsaw University of Life Sciences—SGGW, Nowoursynowska 166, 02-787 Warsaw, Poland; (M.B.M.); (K.P.G.-Z.)
| | | | - Anna Myszka
- Institute of Biological Sciences, Cardinal Stefan Wyszyński University, Dewajtis 5, 01-815 Warsaw, Poland;
| |
Collapse
|
29
|
Suraweera CD, Hinds MG, Kvansakul M. Structural Insight into KsBcl-2 Mediated Apoptosis Inhibition by Kaposi Sarcoma Associated Herpes Virus. Viruses 2022; 14:v14040738. [PMID: 35458468 PMCID: PMC9027176 DOI: 10.3390/v14040738] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2022] [Revised: 03/23/2022] [Accepted: 03/29/2022] [Indexed: 02/05/2023] Open
Abstract
Numerous large DNA viruses have evolved sophisticated countermeasures to hijack the premature programmed cell death of host cells post-infection, including the expression of proteins homologous in sequence, structure, or function to cellular Bcl-2 proteins. Kaposi sarcoma herpes virus (KSHV), a member of the gammaherpesvirinae, has been shown to encode for KsBcl-2, a potent inhibitor of Bcl-2 mediated apoptosis. KsBcl-2 acts by directly engaging host pro-apoptotic Bcl-2 proteins including Bak, Bax and Bok, the BH3-only proteins; Bim, Bid, Bik, Hrk, Noxa and Puma. Here we determined the crystal structures of KsBcl-2 bound to the BH3 motif of pro-apoptotic proteins Bid and Puma. The structures reveal that KsBcl-2 engages pro-apoptotic BH3 motif peptides using the canonical ligand binding groove. Thus, the presence of the readily identifiable conserved BH1 motif sequence “NWGR” of KsBcl-2, as well as highly conserved Arg residue (R86) forms an ionic interaction with the conserved Asp in the BH3 motif in a manner that mimics the canonical ionic interaction seen in host Bcl-2:BH3 motif complexes. These findings provide a structural basis for KSHV mediated inhibition of host cell apoptosis and reveal the flexibility of virus encoded Bcl-2 proteins to mimic key interactions from endogenous host signalling pathways.
Collapse
Affiliation(s)
- Chathura D. Suraweera
- Department of Biochemistry and Chemistry, La Trobe Institute for Molecular Science, La Trobe University, Bundoora, VIC 3086, Australia;
| | - Mark G. Hinds
- Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Parkville, VIC 3052, Australia
- Correspondence: (M.G.H.); (M.K.)
| | - Marc Kvansakul
- Department of Biochemistry and Chemistry, La Trobe Institute for Molecular Science, La Trobe University, Bundoora, VIC 3086, Australia;
- Correspondence: (M.G.H.); (M.K.)
| |
Collapse
|
30
|
Li YS, Ren HC, Cao JH. Correlation of SARS‑CoV‑2 to cancer: Carcinogenic or anticancer? (Review). Int J Oncol 2022; 60:42. [PMID: 35234272 PMCID: PMC8923649 DOI: 10.3892/ijo.2022.5332] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2021] [Accepted: 12/15/2021] [Indexed: 11/05/2022] Open
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is highly infectious and pathogenic. Among patients with severe SARS-CoV-2-caused by corona virus disease 2019 (COVID-19), those complicated with malignant tumor are vulnerable to COVID-19 due to compromised immune function caused by tumor depletion, malnutrition and anti-tumor treatment. Cancer is closely related to the risk of severe illness and mortality in patients with COVID-19. SARS-CoV-2 could promote tumor progression and stimulate metabolism switching in tumor cells to initiate tumor metabolic modes with higher productivity efficiency, such as glycolysis, for facilitating the massive replication of SARS-CoV-2. However, it has been shown that infection with SARS-CoV-2 leads to a delay in tumor progression of patients with natural killer cell (NK cell) lymphoma and Hodgkin's lymphoma, while SARS-CoV-2 elicited anti-tumor immune response may exert a potential oncolytic role in lymphoma patients. The present review briefly summarized potential carcinogenicity and oncolytic characteristics of SARS-CoV-2 as well as strategies to protect patients with cancer during the COVID-19 pandemic.
Collapse
Affiliation(s)
- Ying-Shuang Li
- Intravenous Drug Administration Center, Department of Pharmacy, The Third People's Hospital of Qingdao, Qingdao, Shandong 266041, P.R. China
| | - Hua-Cheng Ren
- Intravenous Drug Administration Center, Department of Pharmacy, The Third People's Hospital of Qingdao, Qingdao, Shandong 266041, P.R. China
| | - Jian-Hua Cao
- Intravenous Drug Administration Center, Department of Pharmacy, The Third People's Hospital of Qingdao, Qingdao, Shandong 266041, P.R. China
| |
Collapse
|
31
|
Verdu-Bou M, Tapia G, Hernandez-Rodriguez A, Navarro JT. Clinical and Therapeutic Implications of Epstein-Barr Virus in HIV-Related Lymphomas. Cancers (Basel) 2021; 13:5534. [PMID: 34771697 PMCID: PMC8583310 DOI: 10.3390/cancers13215534] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Accepted: 10/29/2021] [Indexed: 12/26/2022] Open
Abstract
The incidence of lymphomas is increased in people living with HIV (PLWH). Aggressive B-cell non-Hodgkin lymphomas (NHLs) are the most common and are considered an AIDS-defining cancer (ADC). Although Hodgkin lymphoma (HL) is not considered an ADC, its incidence is also increased in PLWH. Among all HIV-related lymphomas (HRL), the prevalence of Epstein-Barr virus (EBV) is high. It has been shown that EBV is involved in different lymphomagenic mechanisms mediated by some of its proteins, contributing to the development of different lymphoma subtypes. Additionally, cooperation between both HIV and EBV can lead to the proliferation of aberrant B-cells, thereby being an additional lymphomagenic mechanism in EBV-associated HRL. Despite the close relationship between EBV and HRL, the impact of EBV on clinical aspects has not been extensively studied. These lymphomas are treated with the same therapeutic regimens as the general population in combination with cART. Nevertheless, new therapeutic strategies targeting EBV are promising for these lymphomas. In this article, the different types of HRL are extensively reviewed, focusing on the influence of EBV on the epidemiology, pathogenesis, clinical presentation, and pathological characteristics of each lymphoma subtype. Moreover, novel therapies targeting EBV and future strategies to treat HRL harboring EBV are discussed.
Collapse
Affiliation(s)
- Miriam Verdu-Bou
- Lymphoid Neoplasms Group, Josep Carreras Leukaemia Research Institute, Can Ruti Campus, 08916 Badalona, Spain;
| | - Gustavo Tapia
- Department of Pathology, Germans Trias i Pujol Hospital, Universitat Autònoma de Barcelona, 08916 Badalona, Spain;
| | - Agueda Hernandez-Rodriguez
- Department of Microbiology, Germans Trias i Pujol Hospital, Universitat Autònoma de Barcelona, 08916 Badalona, Spain;
| | - Jose-Tomas Navarro
- Lymphoid Neoplasms Group, Josep Carreras Leukaemia Research Institute, Can Ruti Campus, 08916 Badalona, Spain;
- Department of Hematology, Institut Català d’Oncologia-Germans Trias i Pujol Hospital, 08916 Badalona, Spain
- Department of Medicine, Universitat Autònoma de Barcelona, 08916 Badalona, Spain
| |
Collapse
|
32
|
Delvecchio VS, Fierro C, Giovannini S, Melino G, Bernassola F. Emerging roles of the HECT-type E3 ubiquitin ligases in hematological malignancies. Discov Oncol 2021; 12:39. [PMID: 35201500 PMCID: PMC8777521 DOI: 10.1007/s12672-021-00435-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/18/2021] [Accepted: 09/13/2021] [Indexed: 02/07/2023] Open
Abstract
Ubiquitination-mediated proteolysis or regulation of proteins, ultimately executed by E3 ubiquitin ligases, control a wide array of cellular processes, including transcription, cell cycle, autophagy and apoptotic cell death. HECT-type E3 ubiquitin ligases can be distinguished from other subfamilies of E3 ubiquitin ligases because they have a C-terminal HECT domain that directly catalyzes the covalent attachment of ubiquitin to their substrate proteins. Deregulation of HECT-type E3-mediated ubiquitination plays a prominent role in cancer development and chemoresistance. Several members of this subfamily are indeed frequently deregulated in human cancers as a result of genetic mutations and altered expression or activity. HECT-type E3s contribute to tumorigenesis by regulating the ubiquitination rate of substrates that function as either tumour suppressors or oncogenes. While the pathological roles of the HECT family members in solid tumors are quite well established, their contribution to the pathogenesis of hematological malignancies has only recently emerged. This review aims to provide a comprehensive overview of the involvement of the HECT-type E3s in leukemogenesis.
Collapse
Affiliation(s)
- Vincenza Simona Delvecchio
- Department of Experimental Medicine, TOR, University of Rome “Tor Vergata”, Via Montpellier 1, 00133 Rome, Italy
| | - Claudia Fierro
- Department of Experimental Medicine, TOR, University of Rome “Tor Vergata”, Via Montpellier 1, 00133 Rome, Italy
| | - Sara Giovannini
- Department of Experimental Medicine, TOR, University of Rome “Tor Vergata”, Via Montpellier 1, 00133 Rome, Italy
| | - Gerry Melino
- Department of Experimental Medicine, TOR, University of Rome “Tor Vergata”, Via Montpellier 1, 00133 Rome, Italy
| | - Francesca Bernassola
- Department of Experimental Medicine, TOR, University of Rome “Tor Vergata”, Via Montpellier 1, 00133 Rome, Italy
| |
Collapse
|
33
|
Structural Investigation of Orf Virus Bcl-2 Homolog ORFV125 Interactions with BH3-Motifs from BH3-Only Proteins Puma and Hrk. Viruses 2021; 13:v13071374. [PMID: 34372579 PMCID: PMC8310162 DOI: 10.3390/v13071374] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Revised: 07/12/2021] [Accepted: 07/13/2021] [Indexed: 12/21/2022] Open
Abstract
Numerous viruses have evolved sophisticated countermeasures to hijack the early programmed cell death of host cells in response to infection, including the use of proteins homologous in sequence or structure to Bcl-2. Orf virus, a member of the parapoxviridae, encodes for the Bcl-2 homolog ORFV125, a potent inhibitor of Bcl-2-mediated apoptosis in the host. ORFV125 acts by directly engaging host proapoptotic Bcl-2 proteins including Bak and Bax as well as the BH3-only proteins Hrk and Puma. Here, we determined the crystal structures of ORFV125 bound to the BH3 motif of proapoptotic proteins Puma and Hrk. The structures reveal that ORFV125 engages proapoptotic BH3 motif peptides using the canonical ligand binding groove. An Arg located in the structurally equivalent BH1 region of ORFV125 forms an ionic interaction with the conserved Asp in the BH3 motif in a manner that mimics the canonical ionic interaction seen in host Bcl-2:BH3 motif complexes. These findings provide a structural basis for Orf virus-mediated inhibition of host cell apoptosis and reveal the flexibility of virus encoded Bcl-2 proteins to mimic key interactions from endogenous host signalling pathways.
Collapse
|
34
|
Prusinkiewicz MA, Mymryk JS. Metabolic Control by DNA Tumor Virus-Encoded Proteins. Pathogens 2021; 10:560. [PMID: 34066504 PMCID: PMC8148605 DOI: 10.3390/pathogens10050560] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Revised: 05/01/2021] [Accepted: 05/04/2021] [Indexed: 12/15/2022] Open
Abstract
Viruses co-opt a multitude of host cell metabolic processes in order to meet the energy and substrate requirements for successful viral replication. However, due to their limited coding capacity, viruses must enact most, if not all, of these metabolic changes by influencing the function of available host cell regulatory proteins. Typically, certain viral proteins, some of which can function as viral oncoproteins, interact with these cellular regulatory proteins directly in order to effect changes in downstream metabolic pathways. This review highlights recent research into how four different DNA tumor viruses, namely human adenovirus, human papillomavirus, Epstein-Barr virus and Kaposi's associated-sarcoma herpesvirus, can influence host cell metabolism through their interactions with either MYC, p53 or the pRb/E2F complex. Interestingly, some of these host cell regulators can be activated or inhibited by the same virus, depending on which viral oncoprotein is interacting with the regulatory protein. This review highlights how MYC, p53 and pRb/E2F regulate host cell metabolism, followed by an outline of how each of these DNA tumor viruses control their activities. Understanding how DNA tumor viruses regulate metabolism through viral oncoproteins could assist in the discovery or repurposing of metabolic inhibitors for antiviral therapy or treatment of virus-dependent cancers.
Collapse
Affiliation(s)
| | - Joe S. Mymryk
- Department of Microbiology and Immunology, Western University, London, ON N6A 3K7, Canada;
- Department of Otolaryngology, Head & Neck Surgery, Western University, London, ON N6A 3K7, Canada
- Department of Oncology, Western University, London, ON N6A 3K7, Canada
- London Regional Cancer Program, Lawson Health Research Institute, London, ON N6C 2R5, Canada
| |
Collapse
|
35
|
Effects of Epstein-Barr Virus Infection on the Risk and Prognosis of Primary Laryngeal Squamous Cell Carcinoma: A Hospital-Based Case-Control Study in Taiwan. Cancers (Basel) 2021; 13:cancers13071741. [PMID: 33917480 PMCID: PMC8038767 DOI: 10.3390/cancers13071741] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2021] [Revised: 04/01/2021] [Accepted: 04/01/2021] [Indexed: 12/15/2022] Open
Abstract
Mounting molecular evidence supports Epstein-Barr virus (EBV) involvement in the pathogenesis of laryngeal squamous cell carcinoma (LSCC); however, the epidemiological data are inconsistent. In this retrospective case-control study, we aimed to determine whether EBV infection underlies the risk and prognosis of LSCC. The prevalence of EBV infection, as analyzed using an EBV DNA polymerase chain reaction assay, was significantly higher in 42 Taiwanese patients with newly diagnosed primary LSCC, compared to 39 age- and sex-matched control patients without cancer (48% vs. 19%). Furthermore, most of the EBER signals detected using in situ hybridization were localized to the nuclei of tumor-infiltrating lymphocytes. In multivariate analysis, EBV DNA positivity, age ≥ 55 years, cigarette smoking, and high BCL-2, B2M, and CD161 expression (assessed using immunohistochemistry) were identified as independent risk factors for LSCC. Furthermore, five-year local recurrence and disease-free survival rates were 34% and 58%, respectively, with a high EBER signal and low CD3 expression independently predicting five-year local recurrence and disease-free survival. Our comprehensive profiling data accurately identified patients at risk for LSCC development, local recurrence, or disease-free survival. The information obtained in this study improves our understanding of EBV infection in LSCC, and may guide precision medicine for patients with LSCC.
Collapse
|
36
|
Hutcheson RL, Chakravorty A, Sugden B. Burkitt Lymphomas Evolve to Escape Dependencies on Epstein-Barr Virus. Front Cell Infect Microbiol 2021; 10:606412. [PMID: 33505922 PMCID: PMC7829347 DOI: 10.3389/fcimb.2020.606412] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2020] [Accepted: 11/27/2020] [Indexed: 01/25/2023] Open
Abstract
Epstein-Barr Virus (EBV) can transform B cells and contributes to the development of Burkitt lymphoma and other cancers. Through decades of study, we now recognize that many of the viral genes required to transform cells are not expressed in EBV-positive Burkitt lymphoma (BL) tumors, likely due to the immune pressure exerted on infected cells. This recognition has led to the hypothesis that the loss of expression of these viral genes must be compensated through some mechanisms. Recent progress in genome-wide mutational analysis of tumors provides a wealth of data about the cellular mutations found in EBV-positive BLs. Here, we review common cellular mutations found in these tumors and consider how they may compensate for the viral genes that are no longer expressed. Understanding these mutations and how they may substitute for EBV's genes and contribute to lymphomagenesis can serve as a launchpad for more mechanistic studies, which will help us navigate the sea of genomic data available today, and direct the discoveries necessary to improve the treatment of EBV-positive BLs.
Collapse
|
37
|
García-Murria MJ, Duart G, Grau B, Diaz-Beneitez E, Rodríguez D, Mingarro I, Martínez-Gil L. Viral Bcl2s' transmembrane domain interact with host Bcl2 proteins to control cellular apoptosis. Nat Commun 2020; 11:6056. [PMID: 33247105 PMCID: PMC7695858 DOI: 10.1038/s41467-020-19881-9] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2020] [Accepted: 10/29/2020] [Indexed: 12/20/2022] Open
Abstract
Viral control of programmed cell death relies in part on the expression of viral analogs of the B-cell lymphoma 2 (Bcl2) protein known as viral Bcl2s (vBcl2s). vBcl2s control apoptosis by interacting with host pro- and anti-apoptotic members of the Bcl2 family. Here, we show that the carboxyl-terminal hydrophobic region of herpesviral and poxviral vBcl2s can operate as transmembrane domains (TMDs) and participate in their homo-oligomerization. Additionally, we show that the viral TMDs mediate interactions with cellular pro- and anti-apoptotic Bcl2 TMDs within the membrane. Furthermore, these intra-membrane interactions among viral and cellular proteins are necessary to control cell death upon an apoptotic stimulus. Therefore, their inhibition represents a new potential therapy against viral infections, which are characterized by short- and long-term deregulation of programmed cell death.
Collapse
Affiliation(s)
- Maria Jesús García-Murria
- Department of Biochemistry and Molecular Biology, Institut de Biotecnologia i Biomedicina, Universitat de València, 46100, Burjassot, Spain
| | - Gerard Duart
- Department of Biochemistry and Molecular Biology, Institut de Biotecnologia i Biomedicina, Universitat de València, 46100, Burjassot, Spain
| | - Brayan Grau
- Department of Biochemistry and Molecular Biology, Institut de Biotecnologia i Biomedicina, Universitat de València, 46100, Burjassot, Spain
| | - Elisabet Diaz-Beneitez
- Department of Molecular and Cell Biology, Centro Nacional de Biotecnología, Consejo Superior de Investigaciones Científicas, Campus Universidad Autónoma, 28049, Madrid, Spain
| | - Dolores Rodríguez
- Department of Molecular and Cell Biology, Centro Nacional de Biotecnología, Consejo Superior de Investigaciones Científicas, Campus Universidad Autónoma, 28049, Madrid, Spain
| | - Ismael Mingarro
- Department of Biochemistry and Molecular Biology, Institut de Biotecnologia i Biomedicina, Universitat de València, 46100, Burjassot, Spain
| | - Luis Martínez-Gil
- Department of Biochemistry and Molecular Biology, Institut de Biotecnologia i Biomedicina, Universitat de València, 46100, Burjassot, Spain.
| |
Collapse
|
38
|
Epstein-Barr Virus: How Its Lytic Phase Contributes to Oncogenesis. Microorganisms 2020; 8:microorganisms8111824. [PMID: 33228078 PMCID: PMC7699388 DOI: 10.3390/microorganisms8111824] [Citation(s) in RCA: 70] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Revised: 11/14/2020] [Accepted: 11/17/2020] [Indexed: 12/11/2022] Open
Abstract
Epstein–Barr Virus (EBV) contributes to the development of lymphoid and epithelial malignancies. While EBV’s latent phase is more commonly associated with EBV-associated malignancies, there is increasing evidence that EBV’s lytic phase plays a role in EBV-mediated oncogenesis. The lytic phase contributes to oncogenesis primarily in two ways: (1) the production of infectious particles to infect more cells, and (2) the regulation of cellular oncogenic pathways, both cell autonomously and non-cell autonomously. The production of infectious particles requires the completion of the lytic phase. However, the regulation of cellular oncogenic pathways can be mediated by an incomplete (abortive) lytic phase, in which early lytic gene products contribute substantially, whereas late lytic products are largely dispensable. In this review, we discuss the evidence of EBV’s lytic phase contributing to oncogenesis and the role it plays in tumor formation and progression, as well as summarize known mechanisms by which EBV lytic products regulate oncogenic pathways. Understanding the contribution of EBV’s lytic phase to oncogenesis will help design ways to target it to treat EBV-associated malignancies.
Collapse
|
39
|
The Bcl-2 Family: Ancient Origins, Conserved Structures, and Divergent Mechanisms. Biomolecules 2020; 10:biom10010128. [PMID: 31940915 PMCID: PMC7022251 DOI: 10.3390/biom10010128] [Citation(s) in RCA: 69] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2019] [Revised: 12/18/2019] [Accepted: 01/09/2020] [Indexed: 12/13/2022] Open
Abstract
Intrinsic apoptosis, the response to intracellular cell death stimuli, is regulated by the interplay of the B-cell lymphoma 2 (Bcl-2) family and their membrane interactions. Bcl-2 proteins mediate a number of processes including development, homeostasis, autophagy, and innate and adaptive immune responses and their dysregulation underpins a host of diseases including cancer. The Bcl-2 family is characterized by the presence of conserved sequence motifs called Bcl-2 homology motifs, as well as a transmembrane region, which form the interaction sites and intracellular location mechanism, respectively. Bcl-2 proteins have been recognized in the earliest metazoans including Porifera (sponges), Placozoans, and Cnidarians (e.g., Hydra). A number of viruses have gained Bcl-2 homologs and subvert innate immunity and cellular apoptosis for their replication, but they frequently have very different sequences to their host Bcl-2 analogs. Though most mechanisms of apoptosis initiation converge on activation of caspases that destroy the cell from within, the numerous gene insertions, deletions, and duplications during evolution have led to a divergence in mechanisms of intrinsic apoptosis. Currently, the action of the Bcl-2 family is best understood in vertebrates and nematodes but new insights are emerging from evolutionarily earlier organisms. This review focuses on the mechanisms underpinning the activity of Bcl-2 proteins including their structures and interactions, and how they have changed over the course of evolution.
Collapse
|
40
|
Mechanistic Insights into Chemoresistance Mediated by Oncogenic Viruses in Lymphomas. Viruses 2019; 11:v11121161. [PMID: 31888174 PMCID: PMC6950054 DOI: 10.3390/v11121161] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2019] [Revised: 12/09/2019] [Accepted: 12/12/2019] [Indexed: 12/17/2022] Open
Abstract
Viral lymphomagenesis induced by infection with oncogenic viruses, such as Kaposi’s sarcoma associated herpesvirus (KSHV), Epstein–Barr virus (EBV) and human T-cell leukemia virus (HTLV-1), represents a group of aggressive malignancies with a diverse range of pathological features. Combined chemotherapy remains the standard of care for these virus-associated lymphomas; however, frequent chemoresistance is a barrier to achieving successful long-term disease-free survival. There is increasing evidence that indicates virus-associated lymphomas display more resistance to cytotoxic chemotherapeutic agents than that observed in solid tumors. Although the tumor microenvironment and genetic changes, such as key oncogene mutations, are closely related to chemoresistance, some studies demonstrate that the components of oncogenic viruses themselves play pivotal roles in the multidrug chemoresistance of lymphoma cells. In this review, we summarize recent advances in the understanding of the mechanisms through which oncogenic viruses mediate lymphoma cell chemoresistance, with a particular focus on KSHV and EBV, two major oncogenic viruses. We also discuss the current challenges to overcome these obstacles in the treatment of virus-associated lymphomas.
Collapse
|