1
|
Gebauer N, Wang SS. The Role of EBV in the Pathogenesis of Diffuse Large B-Cell Lymphoma. Curr Top Microbiol Immunol 2025. [PMID: 40399571 DOI: 10.1007/82_2025_296] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/23/2025]
Abstract
There are multiple established risk factors for DLBCL; these risk factors share an underlying biology, which generally cause immune dysfunction, spanning immunosuppression to chronic inflammation. EBV is an established risk factor for DLBCL and approximately 10% of DLBCLs are EBV-positive. EBV is a ubiquitous infection, and it is thus among populations that are immunocompromised, by age or medically defined, where EBV-positive DLBCLs arise. In this chapter, we review the current classification, epidemiology, clinical, pathology, and molecular characteristics of EBV-positive DLBCL, and discuss the role of EBV in lymphoma tumorigenesis. We further discuss current and novel treatments aimed at the NFκB pathway and other targets.
Collapse
Affiliation(s)
- Niklas Gebauer
- Klinik für Hämatologie und Onkologie, UKSH Campus Lübeck, Lübeck, Germany
| | - Sophia S Wang
- Division of Computational and Quantitative Medicine, City of Hope Comprehensive Cancer Center, Duarte, CA, USA.
| |
Collapse
|
2
|
Liao Y, Yan J, Kong I, Li Z, Ding W, Clark S, Giulino-Roth L, Gewurz BE. The Histone Demethylase LSD1/ZNF217/CoREST Complex is a Major Restriction Factor of Epstein-Barr Virus Lytic Reactivation. RESEARCH SQUARE 2025:rs.3.rs-5649616. [PMID: 39877093 PMCID: PMC11774438 DOI: 10.21203/rs.3.rs-5649616/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/31/2025]
Abstract
Epstein-Barr virus (EBV) contributes to ~1.5% of human cancers, including lymphomas, gastric and nasopharyngeal carcinomas. In most of these, nearly 80 viral lytic genes are silenced by incompletely understood epigenetic mechanisms, precluding use of antiviral agents such as ganciclovir to treat the 200,000 EBV-associated cancers/year. To identify host factors critical for EBV latency, we performed a human genome-wide CRISPR-Cas9 screen in Burkitt B-cells. Top hits included the lysine-specific histone demethylase LSD1 and its co-repressors ZNF217 and CoREST. LSD1 removes histone 3 lysine 4 (H3K4) and histone 3 lysine 9 (H3K9) methylation marks to downmodulate chromatin activation. LSD1, ZNF217 or CoREST knockout triggered EBV reactivation, as did a LSD1 small molecule antagonist, whose effects were additive with histone deacetylase inhibition. LSD1 blockade reactivated EBV in Burkitt lymphoma, gastric carcinoma and nasopharyngeal carcinoma models, sensitized cells to ganciclovir cytotoxicity and induced EBV reactivation in murine xenografts. ZNF217 and LSD1 co-occupied the EBV immediate early gene BZLF1 promoter, which drives B-cell lytic cycle, as well as to the oriLyt enhancer regions recently implicated in EBV reactivation. LSD1 depletion increased levels of activating histone 3 lysine 4 (H3K4) methylation but not repressive histone lysine 9 methylation marks at BZLF1 and oriLyt and induced their interaction by long-range DNA looping. An orthogonal CRISPR screen highlighted a key H3K4 methyltransferase KMT2D role in driving EBV reactivation. Our results highlight H3K4 methylation as a major EBV lytic switch regulator and suggest novel therapeutic approaches.
Collapse
Affiliation(s)
- Yifei Liao
- Division of Infectious Disease, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
- Center for Integrated Solutions to Infectious Diseases, Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA
- Department of Microbiology, Harvard Medical School, Boston, MA 02115
- Harvard Program in Virology, Harvard Medical School, Boston, MA 02115
| | - Jinjie Yan
- Division of Infectious Disease, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
- Center for Integrated Solutions to Infectious Diseases, Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA
- Department of Microbiology, Harvard Medical School, Boston, MA 02115
- Harvard Program in Virology, Harvard Medical School, Boston, MA 02115
| | - Isabella Kong
- Division of Pediatric Hematology/Oncology, Weill Cornell Medical College, New York, NY 10021, USA
| | - Zhixuan Li
- Division of Infectious Disease, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
- Center for Integrated Solutions to Infectious Diseases, Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA
- Department of Microbiology, Harvard Medical School, Boston, MA 02115
- Harvard Program in Virology, Harvard Medical School, Boston, MA 02115
| | - Weiyue Ding
- Division of Infectious Disease, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Sarah Clark
- Division of Pediatric Hematology/Oncology, Weill Cornell Medical College, New York, NY 10021, USA
| | - Lisa Giulino-Roth
- Division of Pediatric Hematology/Oncology, Weill Cornell Medical College, New York, NY 10021, USA
| | - Benjamin E Gewurz
- Division of Infectious Disease, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
- Center for Integrated Solutions to Infectious Diseases, Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA
- Department of Microbiology, Harvard Medical School, Boston, MA 02115
- Harvard Program in Virology, Harvard Medical School, Boston, MA 02115
| |
Collapse
|
3
|
Ding K, Liu H, Sheng L, Ma J, Zhang X, Huang H, Shi W, Peng H, Cao L, Wu W, Li J, Fan L. Anti-PD-1 antibody (Tislelizumab) combined with gemcitabine and oxaliplatin for extranodal NK/T-cell lymphoma failing asparaginase: A multicenter phase II trial. Eur J Cancer 2025; 214:115155. [PMID: 39644534 DOI: 10.1016/j.ejca.2024.115155] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2024] [Revised: 10/08/2024] [Accepted: 11/25/2024] [Indexed: 12/09/2024]
Abstract
BACKGROUND Extranodal natural killer/T-cell lymphoma (ENKTCL) is almost always fatal after the failure of asparaginase. This phase II study aimed to investigate the efficacy and safety of tislelizumab combined with gemcitabine and oxaliplatin (Tisle-GemOx) in patients with ENKTCL failing asparaginase. METHODS Eligible patients received Tisle-GemOx as initial induction for 6-8 cycles at 21-day intervals. Responders continued tislelizumab maintenance every two months for two years. The primary endpoint was the best complete response rate (CRR). RESULTS As of September 2023, 32 patients were enrolled in our study. Among the 30 efficacy-evaluable patients, the best CRR was 60 %, meeting the primary efficacy endpoint. With a median follow-up of 22.6 months, the median progression-free survival (PFS) was 7.4 months and the 1-year PFS rate was 46.4 %. Subgroup analyses showed that shorter PFS was associated with previous lines of chemotherapy ≥ 2 (P = 0.034) and concomitant hemophagocytic lymphohistiocytosis (P = 0.040). Pseudo-progression was observed in three patients (10 %). The most common grade ≥ 3 toxicities were lymphopenia (25 %) and anemia (15.6 %). CONCLUSIONS Tisle-GemOx exhibits promising anti-tumor activity and manageable toxicities as a salvage therapy for ENKTCL failing asparaginase. Further long-term follow-up is necessary to evaluate the durability of the response with tislelizumab maintenance in this patient population.
Collapse
Affiliation(s)
- Kaiyang Ding
- Department of Hematology, Anhui Provincial Hospital, the First Affiliated Hospital of USTC, Hefei 230002, Anhui, China
| | - Hailing Liu
- Department of Hematology, Jiangsu Province Hospital and Nanjing Medical University First Affiliated Hospital, Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Personalized Cancer Medicine, Nanjing Medical University, Nanjing 210029, Jiangsu, China
| | - Lixia Sheng
- Department of Hematology, the First Affiliated Hospital of Ningbo University, Ningbo, Zhejiang 315010, China
| | - Jie Ma
- Department of Hematology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan, China
| | - Xiaohui Zhang
- Department of Hematology, the Second Affiliated Hospital of Soochow University, Suzhou 215004, Jiangsu, China
| | - Hongming Huang
- Department of Hematology, the Affiliated Hospital of Nantong University, Nantong 226006, Jiangsu, China
| | - Wei Shi
- Department of Hematology, the Friendship Hospital of Ili Kazakh Autonomous Prefecture, Yining 835000, Xinjiang, China
| | - Hongling Peng
- Department of Hematology, the Second Xiangya Hospital, Central South University, Changsha 410011, Hunan, China
| | - Lei Cao
- Department of Hematology, Jiangsu Province Hospital and Nanjing Medical University First Affiliated Hospital, Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Personalized Cancer Medicine, Nanjing Medical University, Nanjing 210029, Jiangsu, China
| | - Wei Wu
- Department of Hematology, Jiangsu Province Hospital and Nanjing Medical University First Affiliated Hospital, Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Personalized Cancer Medicine, Nanjing Medical University, Nanjing 210029, Jiangsu, China
| | - Jianyong Li
- Department of Hematology, Jiangsu Province Hospital and Nanjing Medical University First Affiliated Hospital, Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Personalized Cancer Medicine, Nanjing Medical University, Nanjing 210029, Jiangsu, China.
| | - Lei Fan
- Department of Hematology, Jiangsu Province Hospital and Nanjing Medical University First Affiliated Hospital, Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Personalized Cancer Medicine, Nanjing Medical University, Nanjing 210029, Jiangsu, China.
| |
Collapse
|
4
|
Willman GH, Xu H, Zeigler TM, McIntosh MT, Bhaduri-McIntosh S. Polymerase theta is a synthetic lethal target for killing Epstein-Barr virus lymphomas. J Virol 2024; 98:e0057224. [PMID: 38860782 PMCID: PMC11265443 DOI: 10.1128/jvi.00572-24] [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: 04/02/2024] [Accepted: 05/27/2024] [Indexed: 06/12/2024] Open
Abstract
Treatment options for Epstein-Barr virus (EBV)-cancers are limited, underscoring the need for new therapeutic approaches. We have previously shown that EBV-transformed cells and cancers lack homologous recombination (HR) repair, a prominent error-free pathway that repairs double-stranded DNA breaks; instead, EBV-transformed cells demonstrate genome-wide scars of the error-prone microhomology-mediated end joining (MMEJ) repair pathway. This suggests that EBV-cancers are vulnerable to synthetic lethal therapeutic approaches that target MMEJ repair. Indeed, we have previously found that targeting PARP, an enzyme that contributes to MMEJ, results in the death of EBV-lymphoma cells. With the emergence of clinical resistance to PARP inhibitors and the recent discovery of inhibitors of Polymerase theta (POLθ), the polymerase essential for MMEJ, we investigated the role of POLθ in EBV-lymphoma cells. We report that EBV-transformed cell lines, EBV-lymphoma cell lines, and EBV-lymphomas in AIDS patients demonstrate greater abundance of POLθ, driven by the EBV protein EBNA1, compared to EBV-uninfected primary lymphocytes and EBV-negative lymphomas from AIDS patients (a group that also abundantly expresses POLθ). We also find POLθ enriched at cellular DNA replication forks and exposure to the POLθ inhibitor Novobiocin impedes replication fork progress, impairs MMEJ-mediated repair of DNA double-stranded breaks, and kills EBV-lymphoma cells. Notably, cell killing is not due to Novobiocin-induced activation of the lytic/replicative phase of EBV. These findings support a role for POLθ not just in DNA repair but also DNA replication and as a therapeutic target in EBV-lymphomas and potentially other EBV-cancers as EBNA1 is expressed in all EBV-cancers.IMPORTANCEEpstein-Barr virus (EBV) contributes to ~2% of the global cancer burden. With a recent estimate of >200,000 deaths a year, identifying molecular vulnerabilities will be key to the management of these frequently aggressive and treatment-resistant cancers. Building on our earlier work demonstrating reliance of EBV-cancers on microhomology-mediated end-joining repair, we now report that EBV lymphomas and transformed B cell lines abundantly express the MMEJ enzyme POLθ that likely protects cellular replication forks and repairs replication-related cellular DNA breaks. Importantly also, we show that a newly identified POLθ inhibitor kills EBV-cancer cells, revealing a novel strategy to block DNA replication and repair of these aggressive cancers.
Collapse
Affiliation(s)
- Griffin H. Willman
- Division of Infectious Diseases, Department of Pediatrics, University of Florida, Gainesville, Florida, USA
| | - Huanzhou Xu
- Division of Infectious Diseases, Department of Pediatrics, University of Florida, Gainesville, Florida, USA
| | - Travis M. Zeigler
- Division of Infectious Diseases, Department of Pediatrics, University of Florida, Gainesville, Florida, USA
| | - Michael T. McIntosh
- Child Health Research Institute, Department of Pediatrics, University of Florida, Gainesville, Florida, USA
- Department of Molecular Genetics and Microbiology, University of Florida, Gainesville, Florida, USA
| | - Sumita Bhaduri-McIntosh
- Division of Infectious Diseases, Department of Pediatrics, University of Florida, Gainesville, Florida, USA
- Department of Molecular Genetics and Microbiology, University of Florida, Gainesville, Florida, USA
| |
Collapse
|
5
|
Pociupany M, Snoeck R, Dierickx D, Andrei G. Treatment of Epstein-Barr Virus infection in immunocompromised patients. Biochem Pharmacol 2024; 225:116270. [PMID: 38734316 DOI: 10.1016/j.bcp.2024.116270] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2024] [Revised: 05/06/2024] [Accepted: 05/06/2024] [Indexed: 05/13/2024]
Abstract
Epstein-Barr Virus (EBV), is a ubiquitous γ-Herpesvirus that infects over 95% of the human population and can establish a life-long infection without causing any clinical symptoms in healthy individuals by residing in memory B-cells. Primary infection occurs in childhood and is mostly asymptomatic, however in some young adults it can result in infectious mononucleosis (IM). In immunocompromised individuals however, EBV infection has been associated with many different malignancies. Since EBV can infect both epithelial and B-cells and very rarely NK cells and T-cells, it is associated with both epithelial cancers like nasopharyngeal carcinoma (NPC) and gastric carcinoma (GC), with lymphomas including Burkitt Lymphoma (BL) or Post-transplant Lymphoproliferative Disorder (PTLD) and rarely with NK/T-cell lymphomas. Currently there are no approved antivirals active in PTLD nor in any other malignancy. Moreover, lytic phase disease almost never requires antiviral treatment. Although many novel therapies against EBV have been described, the management and/or prevention of EBV primary infections or reactivations remains difficult. In this review, we discuss EBV infection, therapies targeting EBV in both lytic and latent state with novel therapeutics developed that show anti-EBV activity as well as EBV-associated malignancies both, epithelial and lymphoproliferative malignancies and emerging therapies targeting the EBV-infected cells.
Collapse
Affiliation(s)
- Martyna Pociupany
- Laboratory of Virology and Chemotherapy, Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, KU Leuven, Leuven, Belgium
| | - Robert Snoeck
- Laboratory of Virology and Chemotherapy, Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, KU Leuven, Leuven, Belgium
| | - Daan Dierickx
- Laboratory of Experimental Hematology, Department of Oncology, KU Leuven, Leuven, Belgium; Department of Hematology, University Hospitals Leuven, Leuven, Belgium
| | - Graciela Andrei
- Laboratory of Virology and Chemotherapy, Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, KU Leuven, Leuven, Belgium.
| |
Collapse
|
6
|
Murray-Nerger LA, Lozano C, Burton EM, Liao Y, Ungerleider NA, Guo R, Gewurz BE. The nucleic acid binding protein SFPQ represses EBV lytic reactivation by promoting histone H1 expression. Nat Commun 2024; 15:4156. [PMID: 38755141 PMCID: PMC11099029 DOI: 10.1038/s41467-024-48333-x] [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: 10/24/2023] [Accepted: 04/29/2024] [Indexed: 05/18/2024] Open
Abstract
Epstein-Barr virus (EBV) uses a biphasic lifecycle of latency and lytic reactivation to infect >95% of adults worldwide. Despite its central role in EBV persistence and oncogenesis, much remains unknown about how EBV latency is maintained. We used a human genome-wide CRISPR/Cas9 screen to identify that the nuclear protein SFPQ was critical for latency. SFPQ supported expression of linker histone H1, which stabilizes nucleosomes and regulates nuclear architecture, but has not been previously implicated in EBV gene regulation. H1 occupied latent EBV genomes, including the immediate early gene BZLF1 promoter. Upon reactivation, SFPQ was sequestered into sub-nuclear puncta, and EBV genomic H1 occupancy diminished. Enforced H1 expression blocked EBV reactivation upon SFPQ knockout, confirming it as necessary downstream of SFPQ. SFPQ knockout triggered reactivation of EBV in B and epithelial cells, as well as of Kaposi's sarcoma-associated herpesvirus in B cells, suggesting a conserved gamma-herpesvirus role. These findings highlight SFPQ as a major regulator of H1 expression and EBV latency.
Collapse
Affiliation(s)
- Laura A Murray-Nerger
- Division of Infectious Diseases, Department of Medicine, Brigham and Women's Hospital, 181 Longwood Avenue, Boston, MA, 02115, USA
- Department of Microbiology, Harvard Medical School, Boston, MA, 02115, USA
- Harvard Program in Virology, Boston, MA, 02115, USA
- Center for Integrated Solutions to Infectious Diseases, Broad Institute of Harvard and MIT, Cambridge, MA, 02142, USA
| | - Clarisel Lozano
- Division of Infectious Diseases, Department of Medicine, Brigham and Women's Hospital, 181 Longwood Avenue, Boston, MA, 02115, USA
| | - Eric M Burton
- Division of Infectious Diseases, Department of Medicine, Brigham and Women's Hospital, 181 Longwood Avenue, Boston, MA, 02115, USA
- Department of Microbiology, Harvard Medical School, Boston, MA, 02115, USA
- Harvard Program in Virology, Boston, MA, 02115, USA
- Center for Integrated Solutions to Infectious Diseases, Broad Institute of Harvard and MIT, Cambridge, MA, 02142, USA
| | - Yifei Liao
- Division of Infectious Diseases, Department of Medicine, Brigham and Women's Hospital, 181 Longwood Avenue, Boston, MA, 02115, USA
- Department of Microbiology, Harvard Medical School, Boston, MA, 02115, USA
- Harvard Program in Virology, Boston, MA, 02115, USA
- Center for Integrated Solutions to Infectious Diseases, Broad Institute of Harvard and MIT, Cambridge, MA, 02142, USA
| | | | - Rui Guo
- Department of Molecular Biology and Microbiology, Tufts University, Medford, MA, 02155, USA
| | - Benjamin E Gewurz
- Division of Infectious Diseases, Department of Medicine, Brigham and Women's Hospital, 181 Longwood Avenue, Boston, MA, 02115, USA.
- Department of Microbiology, Harvard Medical School, Boston, MA, 02115, USA.
- Harvard Program in Virology, Boston, MA, 02115, USA.
- Center for Integrated Solutions to Infectious Diseases, Broad Institute of Harvard and MIT, Cambridge, MA, 02142, USA.
| |
Collapse
|
7
|
Haverkos B, Alpdogan O, Baiocchi R, Brammer JE, Feldman TA, Capra M, Brem EA, Nair S, Scheinberg P, Pereira J, Shune L, Joffe E, Young P, Spruill S, Katkov A, McRae R, Royston I, Faller DV, Rojkjaer L, Porcu P. Targeted therapy with nanatinostat and valganciclovir in recurrent EBV-positive lymphoid malignancies: a phase 1b/2 study. Blood Adv 2023; 7:6339-6350. [PMID: 37530631 PMCID: PMC10587711 DOI: 10.1182/bloodadvances.2023010330] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Revised: 06/30/2023] [Accepted: 07/20/2023] [Indexed: 08/03/2023] Open
Abstract
Lymphomas are not infrequently associated with the Epstein-Barr virus (EBV), and EBV positivity is linked to worse outcomes in several subtypes. Nanatinostat is a class-I selective oral histone deacetylase inhibitor that induces the expression of lytic EBV BGLF4 protein kinase in EBV+ tumor cells, activating ganciclovir via phosphorylation, resulting in tumor cell apoptosis. This phase 1b/2 study investigated the combination of nanatinostat with valganciclovir in patients aged ≥18 years with EBV+ lymphomas relapsed/refractory to ≥1 prior systemic therapy with no viable curative treatment options. In the phase 1b part, 25 patients were enrolled into 5 dose escalation cohorts to determine the recommended phase 2 dose (RP2D) for phase 2 expansion. Phase 2 patients (n = 30) received RP2D (nanatinostat 20 mg daily, 4 days per week with valganciclovir 900 mg orally daily) for 28-day cycles. The primary end points were safety, RP2D determination (phase 1b), and overall response rate (ORR; phase 2). Overall, 55 patients were enrolled (B-non-Hodgkin lymphoma [B-NHL], [n = 10]; angioimmunoblastic T-cell lymphoma-NHL, [n = 21]; classical Hodgkin lymphoma, [n = 11]; and immunodeficiency-associated lymphoproliferative disorders, [n = 13]). The ORR was 40% in 43 evaluable patients (complete response rate [CRR], 19% [n = 8]) with a median duration of response of 10.4 months. For angioimmunoblastic T-cell lymphoma-NHL (n = 15; all refractory to the last prior therapy), the ORR/CRR ratio was 60%/27%. The most common adverse events were nausea (38% any grade) and cytopenia (grade 3/4 neutropenia [29%], thrombocytopenia [20%], and anemia [20%]). This novel oral regimen provided encouraging efficacy across several EBV+ lymphoma subtypes and warrants further evaluation; a confirmatory phase 2 study (NCT05011058) is underway. This phase 1b/2 study is registered at www.clinicaltrials.gov as #NCT03397706.
Collapse
Affiliation(s)
| | - Onder Alpdogan
- Division of Hematologic Malignancies and Hematopoetic Stem Cell Transplantation, Department of Medical Oncology, Thomas Jefferson University Hospital, Philadelphia, PA
| | - Robert Baiocchi
- The Ohio State University James Comprehensive Cancer Center, Columbus, OH
| | | | - Tatyana A. Feldman
- John Theurer Cancer Center, Hackensack University Medical Center, Hackensack, NJ
| | - Marcelo Capra
- Centro Integrado de Hematologia e Oncologia - Hospital Mãe de Deus, Porto Alegre, Brazil
| | - Elizabeth A. Brem
- Division of Hematology/Oncology, Deptartment of Medicine, University of California, Irvine, Orange, CA
| | - Santosh Nair
- Mid Florida Hematology and Oncology Center, Orange City, FL
| | - Phillip Scheinberg
- Division of Hematology, Hospital A Beneficência Portuguesa, São Paulo, Brazil
| | - Juliana Pereira
- Division of Hematology, Hospital das Clínicas da Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil
| | - Leyla Shune
- University of Kansas Cancer Center, University of Kansas Medical Center, Kansas City, KS
| | - Erel Joffe
- Memorial Sloan Kettering Cancer Center, New York, NY
| | | | | | | | | | | | | | | | - Pierluigi Porcu
- Division of Hematologic Malignancies and Hematopoetic Stem Cell Transplantation, Department of Medical Oncology, Thomas Jefferson University Hospital, Philadelphia, PA
| |
Collapse
|
8
|
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: 7] [Impact Index Per Article: 3.5] [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
|
9
|
Cordes BLA, Bilger A, Kraus RJ, Ward-Shaw ET, Labott MR, Lee S, Lambert PF, Mertz JE. Drugs That Mimic Hypoxia Selectively Target EBV-Positive Gastric Cancer Cells. Cancers (Basel) 2023; 15:1846. [PMID: 36980731 PMCID: PMC10046841 DOI: 10.3390/cancers15061846] [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: 02/06/2023] [Revised: 03/13/2023] [Accepted: 03/16/2023] [Indexed: 03/30/2023] Open
Abstract
Latent infection of Epstein-Barr virus (EBV) is associated with lymphoid and epithelial cell cancers, including 10% of gastric carcinomas. We previously reported that hypoxia inducible factor-1α (HIF-1α) induces EBV's latent-to-lytic switch and identified several HIF-1α-stabilizing drugs that induce this viral reactivation. Here, we tested three classes of these drugs for preferential killing of the EBV-positive gastric cancer AGS-Akata cell line compared to its matched EBV-negative AGS control. We observed preferential killing with iron chelators [Deferoxamine (DFO); Deferasirox (DFX)] and a prolyl hydroxylase inhibitor (BAY 85-3934 (Molidustat)), but not with a neddylation inhibitor [MLN4924 (Pevonedistat)]. DFO and DFX also induced preferential killing of the EBV-positive gastric cancer AGS-BDneo and SNU-719 cell lines. Preferential killing was enhanced when low-dose DFX (10 μM) was combined with the antiviral prodrug ganciclovir. DFO and DFX induced lytic EBV reactivation in approximately 10% of SNU-719 and 20-30% of AGS-Akata and AGS-BDneo cells. However, neither DFO nor DFX significantly induced synthesis of lytic EBV proteins in xenografts grown in NSG mice from AGS-Akata cells above the level observed in control-treated mice. Therefore, these FDA-approved iron chelators are less effective than gemcitabine at promoting EBV reactivation in vivo despite their high specificity and efficiency in vitro.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Janet E. Mertz
- McArdle Laboratory for Cancer Research, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI 53705, USA; (B.-l.A.C.); (A.B.); (R.J.K.); (E.T.W.-S.); (M.R.L.); (S.L.); (P.F.L.)
| |
Collapse
|
10
|
Sil S, Bertilla J, Rupachandra S. A comprehensive review on RNA interference-mediated targeting of interleukins and its potential therapeutic implications in colon cancer. 3 Biotech 2023; 13:18. [PMID: 36568500 PMCID: PMC9768089 DOI: 10.1007/s13205-022-03421-x] [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: 04/18/2022] [Accepted: 12/05/2022] [Indexed: 12/24/2022] Open
Abstract
Colon cancer is the world's fourth leading cause of death. It is cancer of the latter part of the large intestine, i.e. the colon. Chronic inflammation over a long period also leads to the development of cancer. Cancer in the colon region is arduous to diagnose and is detected at a later stage when it metastasizes to other parts of the body like the liver, lungs, peritoneum, etc. Colon cancer is a great example of solid tumours associated with chronic inflammation. Although conventional therapies are effective, they lose their effectiveness beyond a certain point. Relapse of the disease occurs frequently. RNA interference (RNAi) is emerging as a great tool to specifically attack the cancer cells of a target site like the colon. RNAi deals with epigenetic changes made in the defective cells which ultimately leads to their death without harming the healthy cells. In this review, two types of epigenetic modulators have been considered, namely siRNA and miRNA, and their effect on interleukins. Interleukins, a class of cytokines, are major inflammatory responses of the body that are released by immune cells like leukocytes and macrophages. Some of these interleukins are pro-inflammatory, thereby promoting inflammation which eventually causes cancer. RNAi can prevent colon cancer by inhibiting pro-inflammatory interleukins.
Collapse
Affiliation(s)
- Sagari Sil
- Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Kattankulathur, Chengalpattu, Tamil Nadu 603 203 India
| | - Janet Bertilla
- Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Kattankulathur, Chengalpattu, Tamil Nadu 603 203 India
| | - S. Rupachandra
- Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Kattankulathur, Chengalpattu, Tamil Nadu 603 203 India
| |
Collapse
|
11
|
Lin L, Liu X, Yu H, Deng H, Peng K, Chen J, Zhang C, Jiang T, Liu X. Inhibitory effect and related mechanism of decitabine combined with gemcitabine on proliferation of NK/T cell lymphoma cells. Front Pharmacol 2023; 14:1134895. [PMID: 36937854 PMCID: PMC10014839 DOI: 10.3389/fphar.2023.1134895] [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: 12/31/2022] [Accepted: 02/14/2023] [Indexed: 03/05/2023] Open
Abstract
Background: EBV-associated lymphoma is a neoplasm with a poor prognosis, highly aggressive, and progressive rapidly. There is no standard clinical treatment protocol. Decitabine and gemcitabine are known to have anticancer properties against cells of various cancer, respectively. However, the effect of the combination medication on NK/T cell lymphoma cells and potential mechanisms have not been thoroughly investigated. Methods: Human NK/T cell lymphoma cells NK92MI were treated with decitabine and gemcitabine alone or in combination. Experiments, including the Cell Counting Kit-8 and flow cytometry, were performed to investigate how the combination of decitabine and gemcitabine affects the biological behavior of NK92MI cells in vitro. mRNA sequencing, RT-PCR, and western blotting were used to detect changes in the related signal pathway, mRNA, and protein expressions. Results: Decitabine and gemcitabine significantly inhibited the viability and proliferation of NK92MI cells in a dose-dependent manner. The combination index was less than 1 after treating with two drugs, which was a significant synergistic effect. The decitabine concentration with the best synergistic effect was 4.046 µM, and the gemcitabine concentration was 0.005 µM. Flow cytometry showed that combining two drugs could significantly promote apoptosis and arrest the cell cycle at the S phase. In the combined DAC and GEM group, caspase3 protein levels were higher than in either group alone or the control group. The transcriptome sequence, KEGG, and PPI analysis showed that the differential genes after combined treatment were mainly enriched in signal pathways related to cell proliferation, adhesion, and migration compared with using alone and control groups. Based on the sequencing results, we further investigated the role of DAC and GEM in ferroptosis-related signaling molecules using RT-PCR and Western blot techniques. RT-PCR and western blotting showed that the expression levels of HMOX1 and EBV cleavage gene BRLF1 were higher in the group with combined DAC and GEM than in the group alone and the control group, while the protein and mRNA expression levels of SLC7A11 were lower than the others. In addition, the GPX4 protein expression level in the combination group was lower than in the drug-alone and control groups. In addition, the combination treatment increased the ROS level of NK92MI cells. Conclusion: Our current findings suggested that decitabine had an inhibitory effect on the proliferation of NK92MI cells when co-treated with gemcitabine. This combination may increase the expression of ferroptosis-related signaling molecules, thus inhibiting the proliferation of NK92MI cells. It also promoted apoptosis in NK/T cell lymphoma. For patients with NK/T cell lymphoma, this novel combination may provide clinical benefits.
Collapse
Affiliation(s)
- Lanke Lin
- College of Medical Technology, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Xiangqin Liu
- Department of Laboratory Medicine and Sichuan Provincial Key Laboratory for Human Disease Gene Study, Sichuan Provincial People’s Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Hui Yu
- Department of Laboratory Medicine, The People’s Hospital of Leshan, Leshan, China
| | - Huan Deng
- College of Medical Technology, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Kun Peng
- Health Management Center, Sichuan Provincial People’s Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Jiang Chen
- The Department of Ophthalmology, Sichuan Provincial People’s Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Chunle Zhang
- Division of Nephrology, Kidney Research Institute, West China Hospital of Sichuan University, Chengdu, China
- *Correspondence: Xiaoqi Liu, ; Tao Jiang, ; Chunle Zhang,
| | - Tao Jiang
- Department of Hematology, Sichuan Academy of Medical Sciences & Sichuan Provincial People’s Hospital, Chengdu, China
- *Correspondence: Xiaoqi Liu, ; Tao Jiang, ; Chunle Zhang,
| | - Xiaoqi Liu
- Sichuan Provincial Key Laboratory for Human Disease Gene Study, Center for Medical Genetics, Sichuan Academy of Medical Sciences & Sichuan Provincial People’s Hospital, University of Electronic Science and Technology of China, Chengdu, China
- Research Unit for Blindness Prevention of Chinese Academy of Medical Sciences (2019RU026), Sichuan Academy of Medical Sciences & Sichuan Provincial People’s Hospital,, Chengdu, Sichuan, China
- *Correspondence: Xiaoqi Liu, ; Tao Jiang, ; Chunle Zhang,
| |
Collapse
|
12
|
Yiu SPT, Guo R, Zerbe C, Weekes MP, Gewurz BE. Epstein-Barr virus BNRF1 destabilizes SMC5/6 cohesin complexes to evade its restriction of replication compartments. Cell Rep 2022; 38:110411. [PMID: 35263599 PMCID: PMC8981113 DOI: 10.1016/j.celrep.2022.110411] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Revised: 12/29/2021] [Accepted: 01/28/2022] [Indexed: 11/11/2022] Open
Abstract
Epstein-Barr virus (EBV) persistently infects people worldwide. Delivery of ∼170-kb EBV genomes to nuclei and use of nuclear membrane-less replication compartments (RCs) for their lytic cycle amplification necessitate evasion of intrinsic antiviral responses. Proteomics analysis indicates that, upon B cell infection or lytic reactivation, EBV depletes the cohesin SMC5/6, which has major roles in chromosome maintenance and DNA damage repair. The major tegument protein BNRF1 targets SMC5/6 complexes by a ubiquitin proteasome pathway dependent on calpain proteolysis and Cullin-7. In the absence of BNRF1, SMC5/6 associates with R-loop structures, including at the viral lytic origin of replication, and interferes with RC formation and encapsidation. CRISPR analysis identifies RC restriction roles of SMC5/6 components involved in DNA entrapment and SUMOylation. Our study highlights SMC5/6 as an intrinsic immune sensor and restriction factor for a human herpesvirus RC and has implications for the pathogenesis of EBV-associated cancers.
Collapse
Affiliation(s)
- 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; Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA; Department of Microbiology, Harvard Medical School, Boston, MA 02115, USA
| | - Rui Guo
- Division of Infectious Diseases, Department of Medicine, Brigham and Women's Hospital, 181 Longwood Avenue, Boston, MA 02115, USA; Broad Institute of Harvard and MIT, Cambridge, MA 02142, 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
| | - Michael P Weekes
- Cambridge Institute for Medical Research, University of Cambridge, Hills Road, Cambridge CB2 0XY, UK
| | - Benjamin E Gewurz
- 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; Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA; Department of Microbiology, Harvard Medical School, Boston, MA 02115, USA.
| |
Collapse
|
13
|
Nečasová I, Stojaspal M, Motyčáková E, Brom T, Janovič T, Hofr C. Transcriptional regulators of human oncoviruses: structural and functional implications for anticancer therapy. NAR Cancer 2022; 4:zcac005. [PMID: 35252867 PMCID: PMC8892037 DOI: 10.1093/narcan/zcac005] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Revised: 02/04/2022] [Accepted: 02/15/2022] [Indexed: 11/26/2022] Open
Abstract
Transcription is often the first biosynthetic event of viral infection. Viruses produce preferentially viral transcriptional regulators (vTRs) essential for expressing viral genes and regulating essential host cell proteins to enable viral genome replication. As vTRs are unique viral proteins that promote the transcription of viral nucleic acid, vTRs interact with host proteins to suppress detection and immune reactions to viral infection. Thus, vTRs are promising therapeutic targets that are sequentially and structurally distinct from host cell proteins. Here, we review vTRs of three human oncoviruses: HBx of hepatitis B virus, HBZ of human T-lymphotropic virus type 1, and Rta of Epstein-Barr virus. We present three cunningly exciting and dangerous transcription strategies that make viral infections so efficient. We use available structural and functional knowledge to critically examine the potential of vTRs as new antiviral-anticancer therapy targets. For each oncovirus, we describe (i) the strategy of viral genome transcription; (ii) vTRs' structure and binding partners essential for transcription regulation; and (iii) advantages and challenges of vTR targeting in antiviral therapies. We discuss the implications of vTR regulation for oncogenesis and perspectives on developing novel antiviral and anticancer strategies.
Collapse
Affiliation(s)
- Ivona Nečasová
- Institute of Biophysics of the Czech Academy of Sciences, Scientific Incubator, Královopolská 135, Brno 612 65, Czech Republic
| | - Martin Stojaspal
- Institute of Biophysics of the Czech Academy of Sciences, Scientific Incubator, Královopolská 135, Brno 612 65, Czech Republic
| | - Edita Motyčáková
- Institute of Biophysics of the Czech Academy of Sciences, Scientific Incubator, Královopolská 135, Brno 612 65, Czech Republic
| | - Tomáš Brom
- LifeB, Functional Genomics and Proteomics, National Centre for Biomolecular Research, Faculty of Science, Masaryk University, Kamenice 753/5, Brno 625 00, Czech Republic
| | - Tomáš Janovič
- LifeB, Functional Genomics and Proteomics, National Centre for Biomolecular Research, Faculty of Science, Masaryk University, Kamenice 753/5, Brno 625 00, Czech Republic
| | - Ctirad Hofr
- Institute of Biophysics of the Czech Academy of Sciences, Scientific Incubator, Královopolská 135, Brno 612 65, Czech Republic
| |
Collapse
|
14
|
Sheng L, Chen C, Chen Y, He Y, Zhuang R, Gu Y, Yan Q, Li W, Lu C. vFLIP-regulated competing endogenous RNA (ceRNA) networks targeting lytic induction for KSHV-associated malignancies. J Med Virol 2022; 94:2766-2775. [PMID: 35149992 DOI: 10.1002/jmv.27654] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2021] [Revised: 02/07/2022] [Accepted: 02/09/2022] [Indexed: 11/11/2022]
Abstract
Kaposi's sarcoma-associated herpesvirus (KSHV) causes life-long latent infection and malignancies, including Kaposi sarcoma (KS) commonly found in AIDS patients. Lytic replication can be induced to kill tumor cells harboring latent KSHV, through viral cytopathic effects and the subsequent antiviral immune responses. Viral FLICE-inhibitory protein (vFLIP), encoded by KSHV ORF K13, inhibits KSHV lytic reactivation, implying that the competing endogenous RNA (ceRNA) networks regulated by vFLIP can be modulated to induce the lytic reactivation of latent KSHV, a promising strategy for KSHV-associated malignancies. Here, we performed whole-transcriptome sequencing to reveal the global landscape of non-coding RNAs and mRNAs in iSLK-RGB-BAC16 cells and iSLK-RGB-K13 mutant cells. It showed that vFLIP regulated 227 differently expressed (DE) lncRNAs, 57 DE circRNAs, 20 DE miRNAs and 1371 DE mRNAs. Enrichment analysis verified that riboflavin metabolism was simultaneously enriched in DE genes related to miRNAs, lncRNAs, and circRNAs. The upregulated hsa-miR-378i and hsa-miR-3654, and downregulated miR-4467, miR-3163, miR-4451 and miR-4257 were significantly enriched in the ceRNA complex network, which contained 9 upregulated and 7 downregulated circRNAs, 5 upregulated and 85 downregulated lncRNAs, 5 upregulated and 35 downregulated mRNAs. Finally, we constructed and validated two vFLIP-regulated ceRNA networks: circRNA hsa_circ_0070049/hsa-miR-378i/SPEG/FOXQ1 and lncRNA AL031123.1/hsa-miR-378i/SPEG/FOXQ1. Taken together, the two ceRNA networks may mediate KSHV reactivation. These novel findings refreshed the present understanding of ceRNA network in KSHV lytic induction and provided potential therapeutic targets for KSHV-associated malignancies. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Liuxue Sheng
- State Key Laboratory of Reproductive Medicine, Department of Gynecology, Women's Hospital of Nanjing Medical University, Nanjing Maternity and Child Health Care Hospital, Nanjing Medical University, Nanjing, 210004, P. R. China.,Department of Microbiology, Nanjing Medical University, Nanjing, 211166, P. R. China
| | - Chen Chen
- Department of Microbiology, Nanjing Medical University, Nanjing, 211166, P. R. China
| | - Yuheng Chen
- Department of Microbiology, Nanjing Medical University, Nanjing, 211166, P. R. China
| | - Yujia He
- Department of Microbiology, Nanjing Medical University, Nanjing, 211166, P. R. China
| | - Ruoyu Zhuang
- Department of Microbiology, Nanjing Medical University, Nanjing, 211166, P. R. China
| | - Yang Gu
- Department of Microbiology, Nanjing Medical University, Nanjing, 211166, P. R. China
| | - Qin Yan
- Department of Microbiology, Nanjing Medical University, Nanjing, 211166, P. R. China.,Key Laboratory of Pathogen Biology of Jiangsu Province, Nanjing Medical University, Nanjing, 211166, P. R. China
| | - Wan Li
- State Key Laboratory of Reproductive Medicine, Department of Gynecology, Women's Hospital of Nanjing Medical University, Nanjing Maternity and Child Health Care Hospital, Nanjing Medical University, Nanjing, 210004, P. R. China.,Department of Microbiology, Nanjing Medical University, Nanjing, 211166, P. R. China.,Key Laboratory of Pathogen Biology of Jiangsu Province, Nanjing Medical University, Nanjing, 211166, P. R. China
| | - Chun Lu
- State Key Laboratory of Reproductive Medicine, Department of Gynecology, Women's Hospital of Nanjing Medical University, Nanjing Maternity and Child Health Care Hospital, Nanjing Medical University, Nanjing, 210004, P. R. China.,Department of Microbiology, Nanjing Medical University, Nanjing, 211166, P. R. China.,Key Laboratory of Pathogen Biology of Jiangsu Province, Nanjing Medical University, Nanjing, 211166, P. R. China
| |
Collapse
|
15
|
Yanagi Y, Hara Y, Mabuchi S, Watanabe T, Sato Y, Kimura H, Murata T. PD-L1 upregulation by lytic induction of Epstein-Barr Virus. Virology 2022; 568:31-40. [DOI: 10.1016/j.virol.2022.01.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Revised: 12/20/2021] [Accepted: 01/16/2022] [Indexed: 10/19/2022]
|
16
|
Fan Y, Shan Q, Gong J, Qin J, Lu H. Molecular and Clinical Characteristics of Primary Pulmonary Lymphoepithelioma-Like Carcinoma. Front Mol Biosci 2021; 8:736940. [PMID: 34760925 PMCID: PMC8573970 DOI: 10.3389/fmolb.2021.736940] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2021] [Accepted: 10/07/2021] [Indexed: 11/22/2022] Open
Abstract
Objectives: Primary pulmonary lymphoepithelioma-like carcinoma (PPLELC) is an extremely rare subtype of non-small cell lung cancer (NSCLC). Currently, there are no established treatment protocols due to rarity of the cancer. Thus, this study aimed to explore the molecular and clinical characteristics of PPLELC. Study design and setting: Data from patients with PPLELC who were admitted to Zhejiang Cancer Hospital from August 2009 to September 2020 were retrospectively collected. Next-generation sequencing was performed to obtain a genomic profile and tumor mutation burden (TMB) value of patients with adequate tissue and divided them into two groups according to the expression level of PD-L1. The correlation of PD-L1 expression and the clinicopathological characteristics was evaluated by Pearson Chi-square test. Kaplan-Meier curves was applied to present the probability of survival between PD-L1 expression level and overall survival (OS). Moreover, the literature on the immunotherapy of advanced PPLELC published in PubMed between 2016 and 2020 were reviewed and the efficacy of immunotherapy were analyzed. Results: A total of 18 patients pathologically diagnosed as PPLELC were included. After a follow-up period of 8.8–138 months, 14 patients survived, three patients died and one patient lost, the median OS was 45.3 months Seven samples (tissue-available) tested by NGS and the median TMB was 2.5 mutations/Mb. 19 somatic mutated genes were recognized and TP53 (43%) and CYLD (43%) were the two most commonly mutated genes. Only seven patients who underwent NGS were tested for PD-L1. Three patients with high PD-L1 expression (PD-L1≥ 50%) and four patients with low PD-L1 expression (PD-L1 <50%) were included. No significant correlation was observed between PD-L1 expression and clinical characteristics (age, gender, smoking status, tumor stage, lymph node metastasis) (p > 0.05) and OS (p = 1). What’s more, 10 PPLELC patients involved in previous studies and one patient received nivolumab in the current study were collected retrospectively. 4/11 (36.4%) patients achieved PR, 6/11 (54.5%) patients achieved SD, and 1/11 (9.1%) patients achieved PD and the disease control rate (DCR) was 90.9%. Conclusions: The prognosis of PPLELC is better than that of other NSCLC, and immunotherapy may be a promising treatment to prolong the survival of advanced PPLELC patients. Whether the immunotherapy efficacy of PPLELC can be predicted by PD-L1 and TMB needs further clinical investigation. CYLD genetic alterations may participate in Epstein–Barr virus-mediated tumorigenesis in PPLELC, providing a novel therapeutic target.
Collapse
Affiliation(s)
- Ying Fan
- Zhejiang Key Laboratory of Diagnosis and Treatment Technology on Thoracic Oncology (Lung and Esophagus), Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Hangzhou, China.,Department of Thoracic Medical Oncology, Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Hangzhou, China.,Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, China.,The First Clinical Medical College, Wenzhou Medical University, Wenzhou, China
| | - Qianyun Shan
- Zhejiang Key Laboratory of Diagnosis and Treatment Technology on Thoracic Oncology (Lung and Esophagus), Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Hangzhou, China.,Department of Thoracic Medical Oncology, Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Hangzhou, China.,Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, China.,The Second Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, China
| | - Jiali Gong
- Zhejiang Key Laboratory of Diagnosis and Treatment Technology on Thoracic Oncology (Lung and Esophagus), Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Hangzhou, China.,Department of Thoracic Medical Oncology, Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Hangzhou, China.,Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, China.,The Second Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, China
| | - Jing Qin
- Zhejiang Key Laboratory of Diagnosis and Treatment Technology on Thoracic Oncology (Lung and Esophagus), Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Hangzhou, China.,Department of Thoracic Medical Oncology, Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Hangzhou, China.,Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, China
| | - Hongyang Lu
- Zhejiang Key Laboratory of Diagnosis and Treatment Technology on Thoracic Oncology (Lung and Esophagus), Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Hangzhou, China.,Department of Thoracic Medical Oncology, Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Hangzhou, China.,Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, China.,The First Clinical Medical College, Wenzhou Medical University, Wenzhou, China.,The Second Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, China
| |
Collapse
|
17
|
Van Sciver N, Ohashi M, Nawandar DM, Pauly NP, Lee D, Makielski KR, Bristol JA, Tsao SW, Lambert PF, Johannsen EC, Kenney SC. ΔNp63α promotes Epstein-Barr virus latency in undifferentiated epithelial cells. PLoS Pathog 2021; 17:e1010045. [PMID: 34748616 PMCID: PMC8601603 DOI: 10.1371/journal.ppat.1010045] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2021] [Revised: 11/18/2021] [Accepted: 10/18/2021] [Indexed: 01/27/2023] Open
Abstract
Epstein-Barr virus (EBV) is a human herpesvirus that causes infectious mononucleosis and contributes to both B-cell and epithelial-cell malignancies. EBV-infected epithelial cell tumors, including nasopharyngeal carcinoma (NPC), are largely composed of latently infected cells, but the mechanism(s) maintaining viral latency are poorly understood. Expression of the EBV BZLF1 (Z) and BRLF1 (R) encoded immediate-early (IE) proteins induces lytic infection, and these IE proteins activate each other's promoters. ΔNp63α (a p53 family member) is required for proliferation and survival of basal epithelial cells and is over-expressed in NPC tumors. Here we show that ΔNp63α promotes EBV latency by inhibiting activation of the BZLF1 IE promoter (Zp). Furthermore, we find that another p63 gene splice variant, TAp63α, which is expressed in some Burkitt and diffuse large B cell lymphomas, also represses EBV lytic reactivation. We demonstrate that ΔNp63α inhibits the Z promoter indirectly by preventing the ability of other transcription factors, including the viral IE R protein and the cellular KLF4 protein, to activate Zp. Mechanistically, we show that ΔNp63α promotes viral latency in undifferentiated epithelial cells both by enhancing expression of a known Zp repressor protein, c-myc, and by decreasing cellular p38 kinase activity. Furthermore, we find that the ability of cis-platinum chemotherapy to degrade ΔNp63α contributes to the lytic-inducing effect of this agent in EBV-infected epithelial cells. Together these findings demonstrate that the loss of ΔNp63α expression, in conjunction with enhanced expression of differentiation-dependent transcription factors such as BLIMP1 and KLF4, induces lytic EBV reactivation during normal epithelial cell differentiation. Conversely, expression of ΔNp63α in undifferentiated nasopharyngeal carcinoma cells and TAp63α in Burkitt lymphoma promotes EBV latency in these malignancies.
Collapse
Affiliation(s)
- Nicholas Van Sciver
- Department of Oncology, School of Medicine and Public Health, University of Wisconsin- Madison, Madison, Wisconsin, United States of America
| | - Makoto Ohashi
- Department of Oncology, School of Medicine and Public Health, University of Wisconsin- Madison, Madison, Wisconsin, United States of America
| | - Dhananjay M. Nawandar
- Department of Oncology, School of Medicine and Public Health, University of Wisconsin- Madison, Madison, Wisconsin, United States of America
- Currently at Ring Therapeutics, Cambridge, Massachusetts, United States of America
| | - Nicholas P. Pauly
- Department of Oncology, School of Medicine and Public Health, University of Wisconsin- Madison, Madison, Wisconsin, United States of America
| | - Denis Lee
- Department of Oncology, School of Medicine and Public Health, University of Wisconsin- Madison, Madison, Wisconsin, United States of America
| | - Kathleen R. Makielski
- Department of Oncology, School of Medicine and Public Health, University of Wisconsin- Madison, Madison, Wisconsin, United States of America
| | - Jillian A. Bristol
- Department of Oncology, School of Medicine and Public Health, University of Wisconsin- Madison, Madison, Wisconsin, United States of America
| | - Sai Wah Tsao
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Paul F. Lambert
- Department of Oncology, School of Medicine and Public Health, University of Wisconsin- Madison, Madison, Wisconsin, United States of America
| | - Eric C. Johannsen
- Department of Oncology, 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, 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
|
18
|
Van Sciver N, Ohashi M, Pauly NP, Bristol JA, Nelson SE, Johannsen EC, Kenney SC. Hippo signaling effectors YAP and TAZ induce Epstein-Barr Virus (EBV) lytic reactivation through TEADs in epithelial cells. PLoS Pathog 2021; 17:e1009783. [PMID: 34339458 PMCID: PMC8360610 DOI: 10.1371/journal.ppat.1009783] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2021] [Revised: 08/12/2021] [Accepted: 07/05/2021] [Indexed: 12/13/2022] Open
Abstract
The Epstein-Barr virus (EBV) human herpesvirus is associated with B-cell and epithelial-cell malignancies, and both the latent and lytic forms of viral infection contribute to the development of EBV-associated tumors. Here we show that the Hippo signaling effectors, YAP and TAZ, promote lytic EBV reactivation in epithelial cells. The transcriptional co-activators YAP/TAZ (which are inhibited by Hippo signaling) interact with DNA-binding proteins, particularly TEADs, to induce transcription. We demonstrate that depletion of either YAP or TAZ inhibits the ability of phorbol ester (TPA) treatment, cellular differentiation or the EBV BRLF1 immediate-early (IE) protein to induce lytic EBV reactivation in oral keratinocytes, and show that over-expression of constitutively active forms of YAP and TAZ reactivate lytic EBV infection in conjunction with TEAD family members. Mechanistically, we find that YAP and TAZ interact with, and activate, the EBV BZLF1 immediate-early promoter. Furthermore, we demonstrate that YAP, TAZ, and TEAD family members are expressed at much higher levels in epithelial cell lines in comparison to B-cell lines, and find that EBV infection of oral keratinocytes increases the level of activated (dephosphorylated) YAP and TAZ. Finally, we have discovered that lysophosphatidic acid (LPA), a known YAP/TAZ activator that plays an important role in inflammation, induces EBV lytic reactivation in epithelial cells through a YAP/TAZ dependent mechanism. Together these results establish that YAP/TAZ are powerful inducers of the lytic form of EBV infection and suggest that the ability of EBV to enter latency in B cells at least partially reflects the extremely low levels of YAP/TAZ and TEADs in this cell type.
Collapse
Affiliation(s)
- Nicholas Van Sciver
- Department of Oncology, School of Medicine and Public Health, University of Wisconsin- Madison, Madison, Wisconsin, United States of America
- Cellular and Molecular Pathology Graduate Training Program, University of Wisconsin- Madison, Madison, Wisconsin, United States of America
| | - Makoto Ohashi
- Department of Oncology, School of Medicine and Public Health, University of Wisconsin- Madison, Madison, Wisconsin, United States of America
| | - Nicholas P. Pauly
- Department of Oncology, School of Medicine and Public Health, University of Wisconsin- Madison, Madison, Wisconsin, United States of America
| | - Jillian A. Bristol
- Department of Oncology, School of Medicine and Public Health, University of Wisconsin- Madison, Madison, Wisconsin, United States of America
| | - Scott E. Nelson
- Department of Oncology, School of Medicine and Public Health, University of Wisconsin- Madison, Madison, Wisconsin, United States of America
| | - Eric C. Johannsen
- Department of Oncology, 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, 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
|
19
|
Asha K, Sharma-Walia N. Targeting Host Cellular Factors as a Strategy of Therapeutic Intervention for Herpesvirus Infections. Front Cell Infect Microbiol 2021; 11:603309. [PMID: 33816328 PMCID: PMC8017445 DOI: 10.3389/fcimb.2021.603309] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2020] [Accepted: 01/12/2021] [Indexed: 12/13/2022] Open
Abstract
Herpesviruses utilize various host factors to establish latent infection, survival, and spread disease in the host. These factors include host cellular machinery, host proteins, gene expression, multiple transcription factors, cellular signal pathways, immune cell activation, transcription factors, cytokines, angiogenesis, invasion, and factors promoting metastasis. The knowledge and understanding of host genes, protein products, and biochemical pathways lead to discovering safe and effective antivirals to prevent viral reactivation and spread infection. Here, we focus on the contribution of pro-inflammatory, anti-inflammatory, and resolution lipid metabolites of the arachidonic acid (AA) pathway in the lifecycle of herpesvirus infections. We discuss how various herpesviruses utilize these lipid pathways to their advantage and how we target them to combat herpesvirus infection. We also summarize recent development in anti-herpesvirus therapeutics and new strategies proposed or under clinical trials. These anti-herpesvirus therapeutics include inhibitors blocking viral life cycle events, engineered anticancer agents, epigenome influencing factors, immunomodulators, and therapeutic compounds from natural extracts.
Collapse
Affiliation(s)
| | - Neelam Sharma-Walia
- H. M. Bligh Cancer Research Laboratories, Department of Microbiology and Immunology, Chicago Medical School, Rosalind Franklin University of Medicine and Science, North Chicago, IL, United States
| |
Collapse
|
20
|
Suberoyl bis-hydroxamic acid reactivates Kaposi's sarcoma-associated herpesvirus through histone acetylation and induces apoptosis in lymphoma cells. J Virol 2021; 95:JVI.01785-20. [PMID: 33328303 PMCID: PMC8092814 DOI: 10.1128/jvi.01785-20] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Kaposi's sarcoma-associated herpesvirus (KSHV) is an etiologic agent of Kaposi's sarcoma as well as primary effusion lymphoma (PEL), an aggressive B-cell neoplasm which mostly arises in immunocompromised individuals. Lytic replication of KSHV is also associated with a subset of multicentric Castleman diseases. At present, there is no specific treatment available for PEL and its prognosis is poor. In this study, we found that the histone deacetylase inhibitor suberoyl bis-hydroxamic acid (SBHA) induced KSHV reactivation in PEL cells in a dose-dependent manner. Next-generation sequencing analysis showed that more than 40% of all transcripts expressed in SBHA-treated PEL cells originated from the KSHV genome compared with less than 1% in untreated cells. Chromatin immunoprecipitation assays demonstrated that SBHA induced histone acetylation targeting the promoter region of the KSHV replication and transcription activator gene. However, there was no significant change in methylation status of the promoter region of this gene. In addition to its effect of KSHV reactivation, this study revealed that SBHA induces apoptosis in PEL cells in a dose-dependent manner, inducing acetylation and phosphorylation of p53, cleavage of caspases, and expression of pro-apoptotic factors such as Bim and Bax. These findings suggest that SBHA reactivates KSHV from latency and induces apoptosis through the mitochondrial pathway in PEL cells. Therefore, SBHA can be considered a new tool for induction of KSHV reactivation, and could provide a novel therapeutic strategy against PEL.IMPORTANCE Kaposi's sarcoma and primary effusion lymphoma cells are latently infected with Kaposi's sarcoma-associated herpesvirus (KSHV), whereas KSHV replication is frequently observed in multicentric Castleman disease. Although KSHV replication can be induced by some chemical reagents (e.g. 12-O-tetradecanoylphorbol-13-acetate), the mechanism of KSHV replication is not fully understood. We found that the histone deacetylase inhibitor suberoyl bis-hydroxamic acid (SBHA) induced KSHV reactivation with high efficiency, through histone acetylation in the promoter of the replication and transcription activator gene, compared with 12-O-tetradecanoylphorbol-13-acetate. SBHA also induced apoptosis through the mitochondrial pathway in KSHV-infected cells, with a lower EC50 than measured for viral reactivation. SBHA could be used in a highly efficient replication system for KSHV in vitro, and as a tool to reveal the mechanism of replication and pathogenesis of KSHV. The ability of SBHA to induce apoptosis at lower levels than needed to stimulate KSHV reactivation, indicates its therapeutic potential.
Collapse
|
21
|
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: 80] [Impact Index Per Article: 16.0] [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
|
22
|
Yiu SPT, Dorothea M, Hui KF, Chiang AKS. Lytic Induction Therapy against Epstein-Barr Virus-Associated Malignancies: Past, Present, and Future. Cancers (Basel) 2020; 12:cancers12082142. [PMID: 32748879 PMCID: PMC7465660 DOI: 10.3390/cancers12082142] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Revised: 07/30/2020] [Accepted: 07/30/2020] [Indexed: 12/29/2022] Open
Abstract
Epstein-Barr virus (EBV) lytic induction therapy is an emerging virus-targeted therapeutic approach that exploits the presence of EBV in tumor cells to confer specific killing effects against EBV-associated malignancies. Efforts have been made in the past years to uncover the mechanisms of EBV latent-lytic switch and discover different classes of chemical compounds that can reactivate the EBV lytic cycle. Despite the growing list of compounds showing potential to be used in the lytic induction therapy, only a few are being tested in clinical trials, with varying degrees of success. This review will summarize the current knowledge on EBV lytic reactivation, the major hurdles of translating the lytic induction therapy into clinical settings, and highlight some potential strategies in the future development of this therapy for EBV-related lymphoid and epithelial malignancies.
Collapse
|
23
|
The Epstein-Barr Virus Major Tegument Protein BNRF1 Is a Common Target of Cytotoxic CD4 + T Cells. J Virol 2020; 94:JVI.00284-20. [PMID: 32461311 DOI: 10.1128/jvi.00284-20] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2020] [Accepted: 05/18/2020] [Indexed: 01/14/2023] Open
Abstract
Cellular immunotherapy is a proven approach against Epstein-Barr virus (EBV)-driven lymphoproliferation in recipients of hematopoietic stem cells. Extending the applicability and improving the response rates of such therapy demands improving the knowledge base. We studied 23 healthy donors for specific CD4+ T cell responses against the viral tegument protein BNRF1 and found such T cells in all seropositive donors, establishing BNRF1 as an important immune target in EBV. We identified 18 novel immune epitopes from BNRF1, all of them generated by natural processing of the full-length protein from virus-transformed lymphoblastoid cell lines (LCL). BNRF1-specific CD4+ T cells were measured directly ex vivo by a cytokine-based method, thus providing a tool to study the interaction between immunity and infection in health and disease. T cells of the cytotoxic Th1 type inhibited the proliferation of autologous LCL as well as virus-driven transformation. We infer that they are important in limiting reactivations to subclinical levels during health and reducing virus propagation during disease. The information obtained from this work will feed into data sets that are indispensable in the design of patient-tailored immunotherapeutic approaches, thereby enabling the stride toward broader application of T cell therapy and improving clinical response rates.IMPORTANCE Epstein-Barr virus is carried by most humans and can cause life-threatening diseases. Virus-specific T cells have been used in different clinical settings with variable success rates. One way to improve immunotherapy is to better suit T cell generation protocols to viral targets available in different diseases. BNRF1 is present in viral particles and therefore likely available as a target for T cells in diseases with virus amplification. Here, we studied healthy Epstein-Barr virus (EBV) carriers for BNRF1 immunogenicity and report our results indicating BNRF1 to be a dominant target of the EBV-specific CD4+ T cell response. BNRF1-specific CD4+ T cells were found to be cytotoxic and capable of limiting EBV-driven B cell transformation in vitro The findings of this work contribute to forwarding our understanding of host-virus interactions during health and disease and are expected to find direct application in the generation of specific T cells for immunotherapy.
Collapse
|
24
|
Abstract
Primary pulmonary lymphoepithelioma-like carcinoma (PPLELC) is an extremely rare subtype of non-small-cell lung cancer (NSCLC) but differs from other NSCLC types. Most patients with PPLELC have almost no obvious clinical manifestations, and are often diagnosed in the advanced stage. Complete resection is the standard treatment for cases with early stage disease, while chemotherapy and radiotherapy are effective to some extent in late stage disease. High expression of PD-L1 in PPLELC tumor cells confers immunotherapy an important role in the treatment of PPLELC, and programmed death-1 (PD-1) inhibitors such as nivolumab and pembrolizumab may be optimal options for PPLELC patients. This review focuses on the clinical and pathologic features, treatment, and prognosis of PPLELC.
Collapse
|
25
|
Epstein Barr Virus-associated Pediatric Neoplasms. ARCHIVES OF PEDIATRIC INFECTIOUS DISEASES 2020. [DOI: 10.5812/pedinfect.94371] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
26
|
Dysregulation of Dual-Specificity Phosphatases by Epstein-Barr Virus LMP1 and Its Impact on Lymphoblastoid Cell Line Survival. J Virol 2020; 94:JVI.01837-19. [PMID: 31776277 DOI: 10.1128/jvi.01837-19] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2019] [Accepted: 11/14/2019] [Indexed: 12/14/2022] Open
Abstract
The strongest evidence of the oncogenicity of Epstein-Barr virus (EBV) in vitro is its ability to immortalize human primary B lymphocytes into lymphoblastoid cell lines (LCLs). Yet the underlying mechanisms explaining how the virus tempers the growth program of the host cells have not been fully elucidated. The mitogen-activated protein kinases (MAPKs) are implicated in many cellular processes and are constitutively activated in LCLs. We questioned the expression and regulation of the dual-specificity phosphatases (DUSPs), the main negative regulator of MAPKs, during EBV infection and immortalization. Thirteen DUSPs, including 10 typical and 3 atypical types of DUSPs, were tested. Most of them were downregulated after EBV infection. Here, a role of viral oncogene latent membrane protein 1 (LMP1) in limiting DUSP6 and DUSP8 expression was identified. Using MAPK inhibitors, we found that LMP1 activates extracellular signal-regulated kinase (ERK) or p38 to repress the expression of DUSP6 and DUSP8, with corresponding substrate specificity. Morphologically, overexpression of DUSP6 and DUSP8 attenuates the ability of EBV-immortalized LCL cells to clump together. Mechanistically, apoptosis induced by restoring DUSP6 and DUSP8 in LCLs indicated a novel mechanism for LMP1 to provide a survival signal during EBV immortalization. Collectively, this report provides the first description of the interplay between EBV genes and DUSPs and contributes considerably to the interpretation of MAPK regulation in EBV immortalization.IMPORTANCE Infections by the ubiquitous Epstein-Barr virus (EBV) are associated with a wide spectrum of lymphomas and carcinomas. It has been well documented that activation levels of MAPKs are found in cancer cells to translate various external or intrinsic stimuli into cellular responses. Physiologically, the dual-specificity phosphates (DUSPs) exhibit great ability in regulating MAPK activities with respect to their capability of dephosphorylating MAPKs. In this study, we found that DUSPs were generally downregulated after EBV infection. EBV oncogenic latent membrane protein 1 (LMP1) suppressed DUSP6 and DUSP8 expression via MAPK pathway. In this way, LMP1-mediated MAPK activation was a continuous process. Furthermore, DUSP downregulation was found to contribute greatly to prevent apoptosis of EBV-infected cells. To sum up, this report sheds light on a novel molecular mechanism explaining how EBV maintains the unlimited proliferation status of the immortalized cells and provides a new link to understand EBV-induced B cell survival.
Collapse
|
27
|
Ding L, Zhu Q, Zhou F, Tan H, Xu W, Pan C, Zhu C, Wang Y, Zhang H, Fu W, Qian Z, Yuan Z, Xu H, Wei F, Cai Q. Identification of viral SIM-SUMO2-interaction inhibitors for treating primary effusion lymphoma. PLoS Pathog 2019; 15:e1008174. [PMID: 31830143 PMCID: PMC6932820 DOI: 10.1371/journal.ppat.1008174] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2019] [Revised: 12/26/2019] [Accepted: 10/30/2019] [Indexed: 12/19/2022] Open
Abstract
Primary effusion lymphoma (PEL) is an aggressive B-cell malignancy without effective treatment, and caused by the infection of Kaposi's sarcoma-associated herpesvirus (KSHV), predominantly in its latent form. Previously we showed that the SUMO2-interacting motif within the viral latency-associated nuclear antigen (LANASIM) is essential for establishment and maintenance of KSHV latency. Here, we developed a luciferase based live-cell reporter system to screen inhibitors selectively targeting the interaction between LANASIM and SUMO2. Cambogin, a bioactive natural product isolated from the Garcinia genus (a traditional herbal medicine used for cancer treatment), was obtained from the reporter system screening to efficiently inhibit the association of SUMO2 with LANASIM, in turn reducing the viral episome DNA copy number for establishment and maintenance of KSHV latent infection at a low concentration (nM). Importantly, Cambogin treatments not only specifically inhibited proliferation of KSHV-latently infected cells in vitro, but also induced regression of PEL tumors in a xenograft mouse model. This study has identified Cambogin as a novel therapeutic agent for treating PEL as well as eliminating persistent infection of oncogenic herpesvirus.
Collapse
Affiliation(s)
- Ling Ding
- MOE& NHC&CAMS Key Laboratory of Medical Molecular Virology, School of Basic Medical Science, Shanghai Medical College, Fudan University, Shanghai, P. R. China
| | - Qing Zhu
- MOE& NHC&CAMS Key Laboratory of Medical Molecular Virology, School of Basic Medical Science, Shanghai Medical College, Fudan University, Shanghai, P. R. China
- ShengYushou Center of Cell Biology and Immunology, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, P. R. China
| | - Feng Zhou
- Baoji Affiliated Hospital of Xi’an Medical University, Baoji & MOE Key Laboratory of Western Resources and Modern Biotechnology, College of Life Sciences, Northwest University, Xi’an, Shaanxi, China
| | - Hongsheng Tan
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine & Engineering Research Center of Shanghai Colleges for TCM New Drug Discovery, Shanghai, China
| | - Wenjia Xu
- Unit of Herpesvirus and Molecular Virology, Key Laboratory of Molecular Virology & Immunology, Institut Pasteur of Shanghai, Chinese Academy of Sciences, University of the Chinese Academy of Sciences, Shanghai, P. R. China
| | - Chengling Pan
- Beijing Computing Center, Beijing Academy of Science and Technology & Beijing Beike Deyuan Bio-Pharm Technology Company, Beijing, P. R. China
| | - Caixia Zhu
- MOE& NHC&CAMS Key Laboratory of Medical Molecular Virology, School of Basic Medical Science, Shanghai Medical College, Fudan University, Shanghai, P. R. China
| | - Yuyan Wang
- MOE& NHC&CAMS Key Laboratory of Medical Molecular Virology, School of Basic Medical Science, Shanghai Medical College, Fudan University, Shanghai, P. R. China
| | - Hong Zhang
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine & Engineering Research Center of Shanghai Colleges for TCM New Drug Discovery, Shanghai, China
| | - Wenwei Fu
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine & Engineering Research Center of Shanghai Colleges for TCM New Drug Discovery, Shanghai, China
| | - Zhikang Qian
- Unit of Herpesvirus and Molecular Virology, Key Laboratory of Molecular Virology & Immunology, Institut Pasteur of Shanghai, Chinese Academy of Sciences, University of the Chinese Academy of Sciences, Shanghai, P. R. China
| | - Zhenghong Yuan
- MOE& NHC&CAMS Key Laboratory of Medical Molecular Virology, School of Basic Medical Science, Shanghai Medical College, Fudan University, Shanghai, P. R. China
| | - Hongxi Xu
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine & Engineering Research Center of Shanghai Colleges for TCM New Drug Discovery, Shanghai, China
| | - Fang Wei
- ShengYushou Center of Cell Biology and Immunology, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, P. R. China
| | - Qiliang Cai
- MOE& NHC&CAMS Key Laboratory of Medical Molecular Virology, School of Basic Medical Science, Shanghai Medical College, Fudan University, Shanghai, P. R. China
- Expert Workstation, Baoji Central Hospital, Baoji, P. R. China
| |
Collapse
|
28
|
Pharmacologic Activation of Lytic Epstein-Barr Virus Gene Expression without Virion Production. J Virol 2019; 93:JVI.00998-19. [PMID: 31341058 DOI: 10.1128/jvi.00998-19] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2019] [Accepted: 07/18/2019] [Indexed: 12/14/2022] Open
Abstract
Several therapeutic strategies targeting Epstein-Barr virus (EBV)-associated tumors involve upregulation of viral lytic gene expression. Evidence has been presented that the unfolded protein response (UPR) leads to EBV lytic gene expression. Clofoctol, an antibacterial antibiotic, has been reported to upregulate the UPR in prostate cancer cell lines and to slow their growth. We investigated the effects of clofoctol on an EBV-positive Burkitt lymphoma cell line and confirmed the upregulation of all three branches of the UPR and activation of EBV lytic gene expression. While immediate early, early, and late EBV RNAs were all upregulated, immediate early and early viral proteins but not late viral proteins were expressed. Furthermore, infectious virions were not produced. The use of clofoctol in combination with a protein kinase R-like endoplasmic reticulum kinase inhibitor led to expression of late viral proteins. The effects of clofoctol on EBV lytic protein upregulation were not limited to lymphoid tumor cell lines but also occurred in naturally infected epithelial gastric cancer and nasopharyngeal cancer cell lines. An agent that upregulates lytic viral protein expression but that does not lead to the production of infectious virions may have particular value for lytic induction strategies in the clinical setting.IMPORTANCE Epstein-Barr virus is associated with many different cancers. In these cancers the viral genome is predominantly latent; i.e., most viral genes are not expressed, most viral proteins are not synthesized, and new virions are not produced. Some strategies for treating these cancers involve activation of lytic viral gene expression. We identify an antibacterial antibiotic, clofoctol, that is an activator of EBV lytic RNA and protein expression but that does not lead to virion production.
Collapse
|
29
|
Gao L, Han H, Wang H, Cao L, Feng WH. IL-10 knockdown with siRNA enhances the efficacy of Doxorubicin chemotherapy in EBV-positive tumors by inducing lytic cycle via PI3K/p38 MAPK/NF-kB pathway. Cancer Lett 2019; 462:12-22. [PMID: 31352079 DOI: 10.1016/j.canlet.2019.07.016] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2019] [Revised: 07/02/2019] [Accepted: 07/22/2019] [Indexed: 12/19/2022]
Abstract
High levels of IL-10 expression in Epstein-Barr virus (EBV) associated tumors have been reported and it is likely to be important for maintaining EBV latency and EBV-associated tumors. The switch from the latent form of EBV to the lytic form in tumor cells can lead to tumor cell lysis. Here, we found that knockdown of IL-10 induced EBV lytic replication. Subsequently, we demonstrated that IL-10 knockdown activated BZLF1 promoter through PI3K-p38 MAPK-NF-κB signaling pathway. Interestingly, we verified that VEGF-A was required for IL-10 knockdown to activate PI3K signaling and the accompanying EBV lytic induction. Exogenous recombinant human VEGF-A induced PI3K activation and EBV lytic infection, and inhibition of VEGF-A signaling prevented the PI3K/AKT phosphorylation and EBV reactivation responded to IL-10 knockdown. Most importantly, IL-10 knockdown synergized with chemotherapeutic agent Doxorubicin to kill EBV associated tumor cells in vitro and repress EBV-positive tumor growth in vivo. Our results suggest that inhibition of IL-10 has the potential to serve as a new supplemental strategy for the treatment of EBV-associated tumors.
Collapse
Affiliation(s)
- Li Gao
- State Key Laboratory of Agrobiotechnology, Beijing, 100193, China; Department of Microbiology and Immunology, College of Biological Sciences, China Agricultural University, Beijing, 100193, China; China Academy of Medicine Sciences, Peking Union Medical College, Institute of Medicinal Plant Development, Beijing, 100193, China
| | - Haige Han
- State Key Laboratory of Agrobiotechnology, Beijing, 100193, China; Department of Microbiology and Immunology, College of Biological Sciences, China Agricultural University, Beijing, 100193, China
| | - Honglei Wang
- State Key Laboratory of Agrobiotechnology, Beijing, 100193, China; Department of Microbiology and Immunology, College of Biological Sciences, China Agricultural University, Beijing, 100193, China
| | - Li Cao
- China Academy of Medicine Sciences, Peking Union Medical College, Institute of Medicinal Plant Development, Beijing, 100193, China
| | - Wen-Hai Feng
- State Key Laboratory of Agrobiotechnology, Beijing, 100193, China; Department of Microbiology and Immunology, College of Biological Sciences, China Agricultural University, Beijing, 100193, China.
| |
Collapse
|
30
|
Hui KF, Yiu SPT, Tam KP, Chiang AKS. Viral-Targeted Strategies Against EBV-Associated Lymphoproliferative Diseases. Front Oncol 2019; 9:81. [PMID: 30873380 PMCID: PMC6400835 DOI: 10.3389/fonc.2019.00081] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2018] [Accepted: 01/29/2019] [Indexed: 12/14/2022] Open
Abstract
Epstein-Barr virus (EBV) is strongly associated with a spectrum of EBV-associated lymphoproliferative diseases (EBV-LPDs) ranging from post-transplant lymphoproliferative disorder, B cell lymphomas (e.g., endemic Burkitt lymphoma, Hodgkin lymphoma, and diffuse large B cell lymphoma) to NK or T cell lymphoma (e.g., nasal NK/T-cell lymphoma). The virus expresses a number of latent viral proteins which are able to manipulate cell cycle and cell death processes to promote survival of the tumor cells. Several FDA-approved drugs or novel compounds have been shown to induce killing of some of the EBV-LPDs by inhibiting the function of latent viral proteins or activating the viral lytic cycle from latency. Here, we aim to provide an overview on the mechanisms by which EBV employs to drive the pathogenesis of various EBV-LPDs and to maintain the survival of the tumor cells followed by a discussion on the development of viral-targeted strategies based on the understanding of the patho-mechanisms.
Collapse
Affiliation(s)
- Kwai Fung Hui
- Department of Paediatrics and Adolescent Medicine, Li Ka Shing Faculty of Medicine, Queen Mary Hospital, The University of Hong Kong, Hong Kong, Hong Kong
| | - Stephanie Pei Tung Yiu
- Department of Paediatrics and Adolescent Medicine, Li Ka Shing Faculty of Medicine, Queen Mary Hospital, The University of Hong Kong, Hong Kong, Hong Kong
| | - Kam Pui Tam
- Department of Paediatrics and Adolescent Medicine, Li Ka Shing Faculty of Medicine, Queen Mary Hospital, The University of Hong Kong, Hong Kong, Hong Kong
| | - Alan Kwok Shing Chiang
- Department of Paediatrics and Adolescent Medicine, Li Ka Shing Faculty of Medicine, Queen Mary Hospital, The University of Hong Kong, Hong Kong, Hong Kong.,Center for Nasopharyngeal Carcinoma Research, The University of Hong Kong, Hong Kong, Hong Kong
| |
Collapse
|
31
|
Visalli RJ, Schwartz AM, Patel S, Visalli MA. Identification of the Epstein Barr Virus portal. Virology 2019; 529:152-159. [PMID: 30710799 DOI: 10.1016/j.virol.2019.01.003] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2018] [Revised: 12/21/2018] [Accepted: 01/03/2019] [Indexed: 12/12/2022]
Abstract
Little is known about Epstein Barr Virus (EBV) proteins that participate in viral DNA cleavage and packaging. Genes encoding potential terminase subunit and portal protein homologs include BGRF1/BDRF1, BALF3, BFRF1A and BBRF1 respectively. EBV mutants with deletions in one or more of these genes were impaired for DNA packaging (Pavlova et al., 2013). In the current study, BBRF1 oligomers were purified from recombinant baculovirus infected insect cell extracts. Transmission electron microscopy revealed that purified EBV portals retained features typically found in other portals including a central channel with clip, stem and wing/crown domains. Although compounds have been identified that target DNA encapsidation in human cytomegalovirus, herpes simplex viruses and varicella-zoster virus, the identification of new EBV targets has lagged significantly. Characterization of the EBV portal will direct studies aimed at developing potential small molecular inhibitors of the EBV encapsidation process.
Collapse
Affiliation(s)
- Robert J Visalli
- Department of Biomedical Sciences, Mercer University School of Medicine, Savannah, GA 31404, USA.
| | - Adam M Schwartz
- Department of Biomedical Sciences, Mercer University School of Medicine, Savannah, GA 31404, USA
| | - Shivam Patel
- Department of Biomedical Sciences, Mercer University School of Medicine, Savannah, GA 31404, USA
| | - Melissa A Visalli
- Department of Biomedical Sciences, Mercer University School of Medicine, Savannah, GA 31404, USA
| |
Collapse
|
32
|
Intracellular Iron Chelation by a Novel Compound, C7, Reactivates Epstein⁻Barr Virus (EBV) Lytic Cycle via the ERK-Autophagy Axis in EBV-Positive Epithelial Cancers. Cancers (Basel) 2018; 10:cancers10120505. [PMID: 30544928 PMCID: PMC6316324 DOI: 10.3390/cancers10120505] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2018] [Revised: 12/01/2018] [Accepted: 12/06/2018] [Indexed: 12/21/2022] Open
Abstract
Pharmaceutical reactivation of lytic cycle of Epstein–Barr virus (EBV) represents a potential therapeutic strategy against EBV-associated epithelial malignancies, e.g., gastric carcinoma (GC) and nasopharyngeal carcinoma (NPC). A novel lytic-inducing compound, C7, which exhibits structural similarity to Di-2-Pyridyl Ketone 4, 4-Dimethyl-3-Thiosemicarbazone (Dp44mT), a known chelator of intracellular iron, is found to reactivate EBV lytic cycle in GC and NPC. This study aims to investigate the role of intracellular iron chelation by C7 and other iron chelators in lytic reactivation of EBV in GC and NPC. Testing of six structural analogs of C7 revealed only those which have high affinity towards transition metals could induce EBV lytic cycle. Precomplexing C7 and iron chelators to iron prior to treatment of the cells abolished EBV lytic reactivation. Though hypoxia signaling pathway was activated, it was not the only pathway associated with EBV reactivation. Specifically, C7 and iron chelators initiated autophagy by activating extracellular signal-regulated kinase (ERK1/2) to reactivate EBV lytic cycle since autophagy and EBV lytic reactivation were abolished in cells treated with ERK1/2 blockers whilst inhibition of autophagy by 3-Methyladenine (3-MA) and atg5 knockdown significantly abolished EBV lytic reactivation. In summary, we discovered a novel mechanism of reactivation of the EBV lytic cycle through intracellular iron chelation and induction of ERK-autophagy axis in EBV-positive epithelial malignancies, raising the question whether clinically available iron chelators can be incorporated into existing therapeutic regimens to treat these cancers.
Collapse
|
33
|
Hopcraft SE, Pattenden SG, James LI, Frye S, Dittmer DP, Damania B. Chromatin remodeling controls Kaposi's sarcoma-associated herpesvirus reactivation from latency. PLoS Pathog 2018; 14:e1007267. [PMID: 30212584 PMCID: PMC6136816 DOI: 10.1371/journal.ppat.1007267] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2018] [Accepted: 08/07/2018] [Indexed: 01/08/2023] Open
Abstract
Kaposi's sarcoma-associated herpesvirus (KSHV) is the etiologic agent of three human malignancies, the endothelial cell cancer Kaposi's sarcoma, and two B cell cancers, Primary Effusion Lymphoma and multicentric Castleman's disease. KSHV has latent and lytic phases of the viral life cycle, and while both contribute to viral pathogenesis, lytic proteins contribute to KSHV-mediated oncogenesis. Reactivation from latency is driven by the KSHV lytic gene transactivator RTA, and RTA transcription is controlled by epigenetic modifications. To identify host chromatin-modifying proteins that are involved in the latent to lytic transition, we screened a panel of inhibitors that target epigenetic regulatory proteins for their ability to stimulate KSHV reactivation. We found several novel regulators of viral reactivation: an inhibitor of Bmi1, PTC-209, two additional histone deacetylase inhibitors, Romidepsin and Panobinostat, and the bromodomain inhibitor (+)-JQ1. All of these compounds stimulate lytic gene expression, viral genome replication, and release of infectious virions. Treatment with Romidepsin, Panobinostat, and PTC-209 induces histone modifications at the RTA promoter, and results in nucleosome depletion at this locus. Finally, silencing Bmi1 induces KSHV reactivation, indicating that Bmi1, a member of the Polycomb repressive complex 1, is critical for maintaining KSHV latency.
Collapse
Affiliation(s)
- Sharon E. Hopcraft
- Lineberger Comprehensive Cancer Center, Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States of America
| | - Samantha G. Pattenden
- Center for Integrative Chemical Biology and Drug Discovery, Division of Chemical Biology and Medicinal Chemistry, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States of America
| | - Lindsey I. James
- Center for Integrative Chemical Biology and Drug Discovery, Division of Chemical Biology and Medicinal Chemistry, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States of America
| | - Stephen Frye
- Center for Integrative Chemical Biology and Drug Discovery, Division of Chemical Biology and Medicinal Chemistry, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States of America
| | - Dirk P. Dittmer
- Lineberger Comprehensive Cancer Center, Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States of America
| | - Blossom Damania
- Lineberger Comprehensive Cancer Center, Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States of America
| |
Collapse
|
34
|
Zhang K, Lv DW, Li R. B Cell Receptor Activation and Chemical Induction Trigger Caspase-Mediated Cleavage of PIAS1 to Facilitate Epstein-Barr Virus Reactivation. Cell Rep 2018; 21:3445-3457. [PMID: 29262325 DOI: 10.1016/j.celrep.2017.11.071] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2017] [Revised: 10/21/2017] [Accepted: 11/17/2017] [Indexed: 12/16/2022] Open
Abstract
Epstein-Barr virus (EBV) in tumor cells is predominately in the latent phase, but the virus can undergo lytic reactivation in response to various stimuli. However, the cellular factors that control latency and lytic replication are poorly defined. In this study, we demonstrated that a cellular factor, PIAS1, restricts EBV lytic replication. PIAS1 depletion significantly facilitated EBV reactivation, while PIAS1 reconstitution had the opposite effect. Remarkably, we found that various lytic triggers promote caspase-dependent cleavage of PIAS1 to antagonize PIAS1-mediated restriction and that caspase inhibition suppresses EBV replication through blocking PIAS1 cleavage. We further demonstrated that a cleavage-resistant PIAS1 mutant suppresses EBV replication upon B cell receptor activation. Mechanistically, we demonstrated that PIAS1 acts as an inhibitor for transcription factors involved in lytic gene expression. Collectively, these results establish PIAS1 as a key regulator of EBV lytic replication and uncover a mechanism by which EBV exploits apoptotic caspases to antagonize PIAS1-mediated restriction.
Collapse
Affiliation(s)
- Kun Zhang
- Philips Institute for Oral Health Research, School of Dentistry, Virginia Commonwealth University, Richmond, VA 23298, USA
| | - Dong-Wen Lv
- Philips Institute for Oral Health Research, School of Dentistry, Virginia Commonwealth University, Richmond, VA 23298, USA
| | - Renfeng Li
- Philips Institute for Oral Health Research, School of Dentistry, Virginia Commonwealth University, Richmond, VA 23298, USA; Department of Microbiology and Immunology, School of Medicine, Virginia Commonwealth University, Richmond, VA 23298, USA; Massey Cancer Center, Virginia Commonwealth University, Richmond, VA 23298, USA.
| |
Collapse
|
35
|
Li H, Hu J, Luo X, Bode AM, Dong Z, Cao Y. Therapies based on targeting Epstein-Barr virus lytic replication for EBV-associated malignancies. Cancer Sci 2018; 109:2101-2108. [PMID: 29751367 PMCID: PMC6029825 DOI: 10.1111/cas.13634] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2018] [Revised: 04/24/2018] [Accepted: 04/25/2018] [Indexed: 12/01/2022] Open
Abstract
In recent years, Epstein‐Barr virus (EBV) lytic infection has been shown to significantly contribute to carcinogenesis. Thus, therapies aimed at targeting the EBV lytic cycle have been developed as novel strategies for treatment of EBV‐associated malignancies. In this review, focusing on the viral lytic proteins, we describe recent advances regarding the involvement of the EBV lytic cycle in carcinogenesis. Moreover, we further discuss 2 distinct EBV lytic cycle‐targeted therapeutic strategies against EBV‐induced malignancies. One of the strategies involves inhibition of the EBV lytic cycle by natural compounds known to have anti‐EBV properties; another is to intentionally induce EBV lytic replication in combination with nucleotide analogues. Recent advances in EBV lytic‐based strategies are beginning to show promise in the treatment and/or prevention of EBV‐related tumors.
Collapse
Affiliation(s)
- Hongde Li
- Key Laboratory of Carcinogenesis and Invasion, Chinese Ministry of Education, Xiangya Hospital, Central South University, Changsha, China.,Cancer Research Institute, Xiangya School of Medicine, Central South University, Changsha, China.,Key Laboratory of Carcinogenesis, Chinese Ministry of Health, Changsha, China
| | - Jianmin Hu
- Key Laboratory of Carcinogenesis and Invasion, Chinese Ministry of Education, Xiangya Hospital, Central South University, Changsha, China.,Cancer Research Institute, Xiangya School of Medicine, Central South University, Changsha, China.,Key Laboratory of Carcinogenesis, Chinese Ministry of Health, Changsha, China
| | - Xiangjian Luo
- Key Laboratory of Carcinogenesis and Invasion, Chinese Ministry of Education, Xiangya Hospital, Central South University, Changsha, China.,Cancer Research Institute, Xiangya School of Medicine, Central South University, Changsha, China.,Key Laboratory of Carcinogenesis, Chinese Ministry of Health, Changsha, China
| | - Ann M Bode
- The Hormel Institute, University of Minnesota, Austin, MN, USA
| | - Zigang Dong
- The Hormel Institute, University of Minnesota, Austin, MN, USA
| | - Ya Cao
- Key Laboratory of Carcinogenesis and Invasion, Chinese Ministry of Education, Xiangya Hospital, Central South University, Changsha, China.,Cancer Research Institute, Xiangya School of Medicine, Central South University, Changsha, China.,Key Laboratory of Carcinogenesis, Chinese Ministry of Health, Changsha, China.,Research Center for Technologies of Nucleic Acid-Based Diagnostics and Therapeutics, Hunan Province, Changsha, China
| |
Collapse
|
36
|
Dugan JP, Haverkos BM, Villagomez L, Martin LK, Lustberg M, Patton J, Martin M, Huang Y, Nuovo G, Yan F, Cavaliere R, Fingeroth J, Kenney SC, Ambinder RF, Lozanski G, Porcu P, Caligiuri MA, Baiocchi RA. Complete and Durable Responses in Primary Central Nervous System Posttransplant Lymphoproliferative Disorder with Zidovudine, Ganciclovir, Rituximab, and Dexamethasone. Clin Cancer Res 2018; 24:3273-3281. [PMID: 29632007 DOI: 10.1158/1078-0432.ccr-17-2685] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2017] [Revised: 01/31/2018] [Accepted: 04/02/2018] [Indexed: 01/08/2023]
Abstract
Purpose: Primary central nervous system posttransplant lymphoproliferative disorder (PCNS-PTLD) is a complication of solid organ transplantation with a poor prognosis and typically associated with Epstein-Barr virus (EBV). We hypothesized EBV lytic-phase protein expression would allow successful treatment with antiviral therapy.Patients and Methods: Thirteen patients were treated with zidovudine (AZT), ganciclovir (GCV), dexamethasone, and rituximab in EBV+ PCNS-PTLD. Twice-daily, intravenous AZT 1,500 mg, GCV 5 mg/kg, and dexamethasone 10 mg were given for 14 days. Weekly rituximab 375 mg/m2 was delivered for the first 4 weeks. Twice-daily valganciclovir 450 mg and AZT 300 mg started day 15. Lytic and latent protein expression was assessed using in situ hybridization and immunohistochemistry. Immunoblot assay assessed lytic gene activation. Cells transfected with lytic kinase vectors were assessed for sensitivity to our therapy using MTS tetrazolium and flow cytometry.Results: The median time to response was 2 months. Median therapy duration was 26.5 months. Median follow-up was 52 months. The estimated 2-year overall survival (OS) was 76.9% (95% CI, 44.2%-91.9%). Overall response rate (ORR) was 92% (95% CI, 64%-100%). BXLF1/vTK and BGLF4 expression was found in the seven tumor biopsies evaluated. Lytic gene expression was induced in vitro using the four-drug regimen. Transfection with viral kinase cDNA increased cellular sensitivity to antiviral therapy.Conclusions: EBV+ PCNS-PTLD expressed lytic kinases and therapy with AZT, GCV, rituximab and dexamethasone provided durable responses. Induction of the lytic protein expression and increased cellular sensitivity to antiviral therapy after transfection with viral kinase cDNA provides a mechanistic rationale for our approach. Clin Cancer Res; 24(14); 3273-81. ©2018 AACR.
Collapse
Affiliation(s)
- James P Dugan
- Division of Hematology, University of Colorado, Aurora, Colorado
| | | | - Lynda Villagomez
- Department of Internal Medicine, Mt Sinai School of Medicine, New York, New York
| | - Ludmila K Martin
- Division of Hematology, Department of Internal Medicine, The Ohio State University, Columbus, Ohio
| | - Mark Lustberg
- Division of Infectious Disease, The Ohio State University, Columbus, Ohio
| | - John Patton
- Division of Hematology, Department of Internal Medicine, The Ohio State University, Columbus, Ohio
| | - Marisa Martin
- Division of Hematology, Department of Internal Medicine, The Ohio State University, Columbus, Ohio
| | - Ying Huang
- Division of Hematology, Department of Internal Medicine, The Ohio State University, Columbus, Ohio
| | - Gerard Nuovo
- Department of Pathology, The Ohio State University, Columbus, Ohio
| | - Fengting Yan
- Division of Hematology, Department of Internal Medicine, The Ohio State University, Columbus, Ohio
| | - Robert Cavaliere
- Department of Neurosurgery, The Ohio State University, Columbus, Ohio
| | | | - Shannon C Kenney
- University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin
| | | | - Gerard Lozanski
- Department of Neurosurgery, The Ohio State University, Columbus, Ohio
| | - Pierluigi Porcu
- Division of Hematology, Department of Internal Medicine, The Ohio State University, Columbus, Ohio
| | - Michael A Caligiuri
- Division of Hematology, Department of Internal Medicine, The Ohio State University, Columbus, Ohio
| | - Robert A Baiocchi
- Division of Hematology, Department of Internal Medicine, The Ohio State University, Columbus, Ohio.
| |
Collapse
|
37
|
Kim DE, Jung S, Ryu HW, Choi M, Kang M, Kang H, Yuk HJ, Jeong H, Baek J, Song JH, Kim J, Kang H, Han SB, Oh SR, Cho S. Selective oncolytic effect in Epstein-Barr virus (EBV)-associated gastric carcinoma through efficient lytic induction by Euphorbia extracts. J Funct Foods 2018. [DOI: 10.1016/j.jff.2018.01.004] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
|
38
|
Wang M, Wu W, Zhang Y, Yao G, Gu B. Rapamycin enhances lytic replication of Epstein-Barr virus in gastric carcinoma cells by increasing the transcriptional activities of immediate-early lytic promoters. Virus Res 2018; 244:173-180. [PMID: 29169830 DOI: 10.1016/j.virusres.2017.11.021] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2017] [Revised: 11/18/2017] [Accepted: 11/18/2017] [Indexed: 12/12/2022]
Abstract
Epstein-Barr virus (EBV), a human herpesvirus, is linked to both epithelial and lymphoid malignancies. Induction of EBV reactivation is a potential therapeutic strategy for EBV-associated tumors. In this study, we assessed the effects of rapamycin on EBV reactivation in gastric carcinoma cells. We found that rapamycin upregulated expression of EBV lytic proteins and increased the viral proliferation triggered by the EBV lytic inducer sodium butyrate. Reverse transcription-qPCR, luciferase activity assays, chromatin immunoprecipitation and western blotting were employed to explore the mechanism by which rapamycin promotes EBV reactivation. Our results showed that rapamycin treatment resulted in increased mRNA levels of EBV immediate-early genes. Rapamycin also enhanced the transcriptional activities of the EBV immediate-early lytic promoters Zp and Rp by strengthening Sp1 binding. Repression of the cellular ataxia telangiectasia-mutated/p53 pathway by siRNA-mediated knockdown of the ataxia telangiectasia-mutated gene significantly abrogated virus reactivation by rapamycin/sodium butyrate treatment, indicating that the ataxia telangiectasia-mutated/p53 pathway is involved in rapamycin-promoted EBV reactivation. Taken together, these findings demonstrate that rapamycin might have the potential to enhance the effectiveness of oncolytic viral therapies developed for EBV-associated malignancies.
Collapse
MESH Headings
- Ataxia Telangiectasia Mutated Proteins/antagonists & inhibitors
- Ataxia Telangiectasia Mutated Proteins/genetics
- Ataxia Telangiectasia Mutated Proteins/metabolism
- Butyric Acid/pharmacology
- Cell Line, Tumor
- Cell Survival/drug effects
- DNA, Viral/genetics
- DNA, Viral/metabolism
- Gastric Mucosa/drug effects
- Gastric Mucosa/metabolism
- Gastric Mucosa/virology
- Gene Expression Regulation
- Genes, Reporter
- Herpesvirus 4, Human/drug effects
- Herpesvirus 4, Human/genetics
- Herpesvirus 4, Human/growth & development
- Herpesvirus 4, Human/metabolism
- Humans
- Immediate-Early Proteins/agonists
- Immediate-Early Proteins/genetics
- Immediate-Early Proteins/metabolism
- Luciferases/genetics
- Luciferases/metabolism
- Oncolytic Virotherapy/methods
- Promoter Regions, Genetic/drug effects
- Protein Binding
- RNA, Small Interfering/genetics
- RNA, Small Interfering/metabolism
- Sirolimus/pharmacology
- Sp1 Transcription Factor/genetics
- Sp1 Transcription Factor/metabolism
- Transcription, Genetic
- Tumor Suppressor Protein p53/antagonists & inhibitors
- Tumor Suppressor Protein p53/genetics
- Tumor Suppressor Protein p53/metabolism
- Virus Activation/drug effects
- Virus Replication/drug effects
Collapse
Affiliation(s)
- Man Wang
- Institute for Translational Medicine, Medical College of Qingdao University, Qingdao, 266021, China.
| | - Wei Wu
- Institute for Translational Medicine, Medical College of Qingdao University, Qingdao, 266021, China
| | - Yinfeng Zhang
- Institute for Translational Medicine, Medical College of Qingdao University, Qingdao, 266021, China
| | - Guoliang Yao
- Department of General Surgery, The First Affiliated Hospital of Henan University of Science and Technology, Luoyang, 471003, China
| | - Bianli Gu
- Henan Key Laboratory of Cancer Epigenetics, Cancer Hospital, The First Affiliated Hospital, College of Clinical Medicine, Medical College of Henan University of Science and Technology, Luoyang, 471003, China
| |
Collapse
|
39
|
DNA hypermethylation induced by Epstein-Barr virus in the development of Epstein-Barr virus-associated gastric carcinoma. Arch Pharm Res 2017; 40:894-905. [PMID: 28779374 DOI: 10.1007/s12272-017-0939-5] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2017] [Accepted: 07/27/2017] [Indexed: 12/12/2022]
Abstract
Epstein-Barr virus (EBV)-associated gastric carcinoma (EBVaGC) is a recently recognized disease entity defined by the presence of EBV in gastric carcinoma cells. EBV infection causes major epigenetic alterations in the EBV genome and its cellular host genome, suggesting that EBV acts as a direct epigenetic driver for EBVaGC. One of the major epigenetic events in the viral and cellular genomes to control transcription is DNA hypo- or hyper-methylation. Particularly, local and global hypermethylation have been reported in EBVaGC. It is therefore important to understand the molecular mechanisms of DNA hypermethylation during EBVaGC carcinogenesis. To understand the functional roles of DNA methylation and suggest therapeutic target candidates for EBVaGC, we reviewed recent literature reporting DNA hypermethylation in EBVaGC. We summarized the identified candidate genes that are markedly hypermethylated in EBVaGC, which can potentially be targets for chemotherapies with demethylating agents.
Collapse
|
40
|
Kraus RJ, Yu X, Cordes BLA, Sathiamoorthi S, Iempridee T, Nawandar DM, Ma S, Romero-Masters JC, McChesney KG, Lin Z, Makielski KR, Lee DL, Lambert PF, Johannsen EC, Kenney SC, Mertz JE. Hypoxia-inducible factor-1α plays roles in Epstein-Barr virus's natural life cycle and tumorigenesis by inducing lytic infection through direct binding to the immediate-early BZLF1 gene promoter. PLoS Pathog 2017; 13:e1006404. [PMID: 28617871 PMCID: PMC5487075 DOI: 10.1371/journal.ppat.1006404] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2017] [Revised: 06/27/2017] [Accepted: 05/08/2017] [Indexed: 12/17/2022] Open
Abstract
When confronted with poor oxygenation, cells adapt by activating survival signaling pathways, including the oxygen-sensitive transcriptional regulators called hypoxia-inducible factor alphas (HIF-αs). We report here that HIF-1α also regulates the life cycle of Epstein-Barr virus (EBV). Incubation of EBV-positive gastric carcinoma AGS-Akata and SNU-719 and Burkitt lymphoma Sal and KemIII cell lines with a prolyl hydroxylase inhibitor, L-mimosine or deferoxamine, or the NEDDylation inhibitor MLN4924 promoted rapid and sustained accumulation of both HIF-1α and lytic EBV antigens. ShRNA knockdown of HIF-1α significantly reduced deferoxamine-mediated lytic reactivation. HIF-1α directly bound the promoter of the EBV primary latent-lytic switch BZLF1 gene, Zp, activating transcription via a consensus hypoxia-response element (HRE) located at nt -83 through -76 relative to the transcription initiation site. HIF-1α did not activate transcription from the other EBV immediate-early gene, BRLF1. Importantly, expression of HIF-1α induced EBV lytic-gene expression in cells harboring wild-type EBV, but not in cells infected with variants containing base-pair substitution mutations within this HRE. Human oral keratinocyte (NOK) and gingival epithelial (hGET) cells induced to differentiate by incubation with either methyl cellulose or growth in organotypic culture accumulated both HIF-1α and Blimp-1α, another cellular factor implicated in lytic reactivation. HIF-1α activity also accumulated along with Blimp-1α during B-cell differentiation into plasma cells. Furthermore, most BZLF1-expressing cells observed in lymphomas induced by EBV in NSG mice with a humanized immune system were located distal to blood vessels in hypoxic regions of the tumors. Thus, we conclude that HIF-1α plays central roles in both EBV’s natural life cycle and EBV-associated tumorigenesis. We propose that drugs that induce HIF-1α protein accumulation are good candidates for development of a lytic-induction therapy for treating some EBV-associated malignancies. Most adults throughout the world are infected with Epstein-Barr virus (EBV), a human herpesvirus frequently associated in a latent state with some cancers of epithelial and B-cell origin such as nasopharyngeal carcinoma and Burkitt lymphoma, respectively. To develop an oncolytic therapy for treating patients with EBV-associated cancers, we need a method to efficiently induce synthesis of lytic EBV proteins. The EBV protein encoded by its immediate-early BZLF1 gene usually mediates the switch into lytic viral infection. We show here that HIF-1α, a cellular transcription factor that accumulates in cells when deprived of normal levels of oxygen, can induce lytic EBV infection. HIF-1α mediates this switch by directly binding to a specific sequence located within the BZLF1 gene promoter, activating its expression. Importantly, we also show that deferoxamine, an FDA-approved drug that inhibits degradation of HIF-1α, can induce synthesis of lytic EBV proteins in some EBV-positive epithelial and lymphocytic cell lines. These findings indicate that HIF-1α-stabilizing drugs, administered in combination with nucleoside analogues such as ganciclovir, may be helpful as part of a lytic-induction therapy for treating some patients with EBV-positive malignancies.
Collapse
Affiliation(s)
- Richard J. Kraus
- McArdle Laboratory for Cancer Research, Department of Oncology, University of Wisconsin-Madison School of Medicine and Public Health, Madison, Wisconsin, United States of America
| | - Xianming Yu
- McArdle Laboratory for Cancer Research, Department of Oncology, University of Wisconsin-Madison School of Medicine and Public Health, Madison, Wisconsin, United States of America
| | - Blue-leaf A. Cordes
- McArdle Laboratory for Cancer Research, Department of Oncology, University of Wisconsin-Madison School of Medicine and Public Health, Madison, Wisconsin, United States of America
| | - Saraniya Sathiamoorthi
- McArdle Laboratory for Cancer Research, Department of Oncology, University of Wisconsin-Madison School of Medicine and Public Health, Madison, Wisconsin, United States of America
| | - Tawin Iempridee
- National Nanotechnology Center, National Science and Technology Development Agency, Thailand Science Park, Pathum Thani, Thailand
| | - Dhananjay M. Nawandar
- McArdle Laboratory for Cancer Research, Department of Oncology, University of Wisconsin-Madison School of Medicine and Public Health, Madison, Wisconsin, United States of America
| | - Shidong Ma
- McArdle Laboratory for Cancer Research, Department of Oncology, University of Wisconsin-Madison School of Medicine and Public Health, Madison, Wisconsin, United States of America
| | - James C. Romero-Masters
- McArdle Laboratory for Cancer Research, Department of Oncology, University of Wisconsin-Madison School of Medicine and Public Health, Madison, Wisconsin, United States of America
| | - Kyle G. McChesney
- McArdle Laboratory for Cancer Research, Department of Oncology, University of Wisconsin-Madison School of Medicine and Public Health, Madison, Wisconsin, United States of America
| | - Zhen Lin
- Department of Pathology, Tulane University Health Sciences Center and Tulane Cancer Center, New Orleans, Louisiana, United States of America
| | - Kathleen R. Makielski
- McArdle Laboratory for Cancer Research, Department of Oncology, University of Wisconsin-Madison School of Medicine and Public Health, Madison, Wisconsin, United States of America
| | - Denis L. Lee
- McArdle Laboratory for Cancer Research, Department of Oncology, University of Wisconsin-Madison School of Medicine and Public Health, Madison, Wisconsin, United States of America
| | - Paul F. Lambert
- McArdle Laboratory for Cancer Research, Department of Oncology, University of Wisconsin-Madison School of Medicine and Public Health, Madison, Wisconsin, United States of America
| | - Eric C. Johannsen
- McArdle Laboratory for Cancer Research, Department of Oncology, University of Wisconsin-Madison School of Medicine and Public Health, Madison, Wisconsin, United States of America
- Department of Medicine, University of Wisconsin-Madison School of Medicine and Public Health, Madison, Wisconsin, United States of America
| | - Shannon C. Kenney
- McArdle Laboratory for Cancer Research, Department of Oncology, University of Wisconsin-Madison School of Medicine and Public Health, Madison, Wisconsin, United States of America
- Department of Medicine, University of Wisconsin-Madison School of Medicine and Public Health, Madison, Wisconsin, United States of America
| | - Janet E. Mertz
- McArdle Laboratory for Cancer Research, Department of Oncology, University of Wisconsin-Madison School of Medicine and Public Health, Madison, Wisconsin, United States of America
- * E-mail:
| |
Collapse
|
41
|
Keck KM, Moquin SA, He A, Fernandez SG, Somberg JJ, Liu SM, Martinez DM, Miranda JL. Bromodomain and extraterminal inhibitors block the Epstein-Barr virus lytic cycle at two distinct steps. J Biol Chem 2017; 292:13284-13295. [PMID: 28588024 PMCID: PMC5555189 DOI: 10.1074/jbc.m116.751644] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2016] [Revised: 05/15/2017] [Indexed: 12/17/2022] Open
Abstract
Lytic infection by the Epstein-Barr virus (EBV) poses numerous health risks, such as infectious mononucleosis and lymphoproliferative disorder. Proteins in the bromodomain and extraterminal (BET) family regulate multiple stages of viral life cycles and provide promising intervention targets. Synthetic small molecules can bind to the bromodomains and disrupt function by preventing recognition of acetylated lysine substrates. We demonstrate that JQ1 and other BET inhibitors block two different steps in the sequential cascade of the EBV lytic cycle. BET inhibitors prevent expression of the viral immediate-early protein BZLF1. JQ1 alters transcription of genes controlled by the host protein BACH1, and BACH1 knockdown reduces BZLF1 expression. BET proteins also localize to the lytic origin of replication (OriLyt) genetic elements, and BET inhibitors prevent viral late gene expression. There JQ1 reduces BRD4 recruitment during reactivation to preclude replication initiation. This represents a rarely observed dual mode of action for drugs.
Collapse
Affiliation(s)
- Kristin M Keck
- the Gladstone Institute of Virology and Immunology, San Francisco, California 94158
| | - Stephanie A Moquin
- the Gladstone Institute of Virology and Immunology, San Francisco, California 94158.,From the Department of Cellular and Molecular Pharmacology, University of California, San Francisco (UCSF), California 94158 and
| | - Amanda He
- the Gladstone Institute of Virology and Immunology, San Francisco, California 94158.,From the Department of Cellular and Molecular Pharmacology, University of California, San Francisco (UCSF), California 94158 and
| | - Samantha G Fernandez
- the Gladstone Institute of Virology and Immunology, San Francisco, California 94158
| | - Jessica J Somberg
- the Gladstone Institute of Virology and Immunology, San Francisco, California 94158
| | - Stephanie M Liu
- the Gladstone Institute of Virology and Immunology, San Francisco, California 94158
| | - Delsy M Martinez
- the Gladstone Institute of Virology and Immunology, San Francisco, California 94158.,From the Department of Cellular and Molecular Pharmacology, University of California, San Francisco (UCSF), California 94158 and
| | - Jj L Miranda
- the Gladstone Institute of Virology and Immunology, San Francisco, California 94158 .,From the Department of Cellular and Molecular Pharmacology, University of California, San Francisco (UCSF), California 94158 and
| |
Collapse
|
42
|
EBV reactivation as a target of luteolin to repress NPC tumorigenesis. Oncotarget 2017; 7:18999-9017. [PMID: 26967558 PMCID: PMC4951347 DOI: 10.18632/oncotarget.7967] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2015] [Accepted: 02/08/2016] [Indexed: 11/25/2022] Open
Abstract
Nasopharyngeal carcinoma (NPC) is a malignancy derived from the epithelial cells of the nasopharynx. Although a combination of radiotherapy with chemotherapy is effective for therapy, relapse and metastasis after remission remain major causes of mortality. Epstein-Barr virus (EBV) is believed to be one of causes of NPC development. We demonstrated previously that EBV reactivation is important for the carcinogenesis of NPC. We sought, therefore, to determine whether EBV reactivation can be a target for retardation of relapse of NPC. After screening, we found luteolin is able to inhibit EBV reactivation. It inhibited EBV lytic protein expression and repressed the promoter activities of two major immediate-early genes, Zta and Rta. Furthermore, luteolin was shown to reduce genomic instability induced by recurrent EBV reactivation in NPC cells. EBV reactivation-induced NPC cell proliferation and migration, as well as matrigel invasiveness, were also repressed by luteolin treatment. Tumorigenicity in mice, induced by EBV reactivation, was decreased profoundly following luteolin administration. Together, these results suggest that inhibition of EBV reactivation is a novel approach to prevent the relapse of NPC.
Collapse
|
43
|
Jones RJ, Iempridee T, Wang X, Lee HC, Mertz JE, Kenney SC, Lin HC, Baladandayuthapani V, Dawson CW, Shah JJ, Weber DM, Orlowski RZ. Lenalidomide, Thalidomide, and Pomalidomide Reactivate the Epstein-Barr Virus Lytic Cycle through Phosphoinositide 3-Kinase Signaling and Ikaros Expression. Clin Cancer Res 2016; 22:4901-4912. [PMID: 27297582 PMCID: PMC5050094 DOI: 10.1158/1078-0432.ccr-15-2242] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2015] [Accepted: 05/22/2016] [Indexed: 01/05/2023]
Abstract
PURPOSE Lenalidomide, thalidomide, and pomalidomide (LTP) are immunomodulatory agents approved for use in multiple myeloma, but in some settings, especially with alkylating agents, an increase in Hodgkin lymphoma and other secondary primary malignancies (SPM) has been noted. Some of these malignancies have been linked to Epstein-Barr virus (EBV), raising the possibility that immunomodulatory drugs disrupt latent EBV infection. EXPERIMENTAL DESIGN We studied the ability of LTP to reactivate latently infected EBV-positive cell lines in vitro and in vivo, and evaluated the EBV viral load in archived serum samples from patients who received a lenalidomide, thalidomide, and dexamethasone (LTD) combination. RESULTS Treatment of EBV-infected B-cell lines with LTP at physiologically relevant concentrations induced the immediate early gene BZLF1, the early gene BMRF1, and the late proteins VCA and BCFR1. This occurred in the potency order pomalidomide > lenalidomide > thalidomide, and the nucleoside analogue ganciclovir enhanced the cytotoxic effects of lenalidomide and pomalidomide in Burkitt lymphoma cells in vitro and in vivo EBV reactivation was related to PI3K stimulation and Ikaros suppression, and blocked by the PI3Kδ inhibitor idelalisib. Combinations of lenalidomide with dexamethasone or rituximab increased EBV reactivation compared with lenalidomide alone and, importantly, lenalidomide with melphalan produced even greater reactivation. CONCLUSIONS We conclude LTP may reactivate EBV-positive resting memory B cells thereby enhancing EBV lytic cycle and host immune suppression. Clin Cancer Res; 22(19); 4901-12. ©2016 AACR.
Collapse
Affiliation(s)
- Richard J Jones
- Department of Lymphoma and Myeloma, The University of Texas MD Anderson Cancer Center, Houston, Texas.
| | - Tawin Iempridee
- National Nanotechnology Center (NANOTEC), National Science and Technology Development Agency (NSTDA), Pathum Thani, Thailand
| | - Xiaobin Wang
- Urology Department, ShengJing Hospital, China Medical University, ShenYang, China
| | - Hans C Lee
- Department of Lymphoma and Myeloma, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Janet E Mertz
- McArdle Laboratory for Cancer Research, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin
| | - Shannon C Kenney
- McArdle Laboratory for Cancer Research, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin
| | - Heather C Lin
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | | | - Christopher W Dawson
- Birmingham Cancer Research UK Cancer Centre, School of Cancer Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Jatin J Shah
- Department of Lymphoma and Myeloma, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Donna M Weber
- Department of Lymphoma and Myeloma, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Robert Z Orlowski
- Department of Lymphoma and Myeloma, The University of Texas MD Anderson Cancer Center, Houston, Texas. Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, Texas
| |
Collapse
|
44
|
Topalis D, Gillemot S, Snoeck R, Andrei G. Distribution and effects of amino acid changes in drug-resistant α and β herpesviruses DNA polymerase. Nucleic Acids Res 2016; 44:9530-9554. [PMID: 27694307 PMCID: PMC5175367 DOI: 10.1093/nar/gkw875] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2016] [Revised: 09/13/2016] [Accepted: 09/21/2016] [Indexed: 12/15/2022] Open
Abstract
Emergence of drug-resistance to all FDA-approved antiherpesvirus agents is an increasing concern in immunocompromised patients. Herpesvirus DNA polymerase (DNApol) is currently the target of nucleos(t)ide analogue-based therapy. Mutations in DNApol that confer resistance arose in immunocompromised patients infected with herpes simplex virus 1 (HSV-1) and human cytomegalovirus (HCMV), and to lesser extent in herpes simplex virus 2 (HSV-2), varicella zoster virus (VZV) and human herpesvirus 6 (HHV-6). In this review, we present distinct drug-resistant mutational profiles of herpesvirus DNApol. The impact of specific DNApol amino acid changes on drug-resistance is discussed. The pattern of genetic variability related to drug-resistance differs among the herpesviruses. Two mutational profiles appeared: one favoring amino acid changes in the Palm and Finger domains of DNApol (in α-herpesviruses HSV-1, HSV-2 and VZV), and another with mutations preferentially in the 3′-5′ exonuclease domain (in β-herpesvirus HCMV and HHV-6). The mutational profile was also related to the class of compound to which drug-resistance emerged.
Collapse
Affiliation(s)
- D Topalis
- Rega Institute for Medical Research, Department Microbiology and Immunology, KU Leuven, Minderbroedersstraat 10, 3000, Leuven, Belgium
| | - S Gillemot
- Rega Institute for Medical Research, Department Microbiology and Immunology, KU Leuven, Minderbroedersstraat 10, 3000, Leuven, Belgium
| | - R Snoeck
- Rega Institute for Medical Research, Department Microbiology and Immunology, KU Leuven, Minderbroedersstraat 10, 3000, Leuven, Belgium
| | - G Andrei
- Rega Institute for Medical Research, Department Microbiology and Immunology, KU Leuven, Minderbroedersstraat 10, 3000, Leuven, Belgium
| |
Collapse
|
45
|
Hsu CL, Kuo YC, Huang Y, Huang YC, Lui KW, Chang KP, Lin TL, Fan HC, Lin AC, Hsieh CH, Lee LY, Wang HM, Li HP, Chang YS. Application of a patient-derived xenograft model in cytolytic viral activation therapy for nasopharyngeal carcinoma. Oncotarget 2016; 6:31323-34. [PMID: 26416517 PMCID: PMC4741608 DOI: 10.18632/oncotarget.5544] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2015] [Accepted: 09/11/2015] [Indexed: 01/08/2023] Open
Abstract
Nasopharyngeal carcinoma (NPC) is an Epstein Barr virus (EBV)-related malignancy in which the tumor microenvironment plays a pivotal role in tumor progression. Here, we developed two patient-derived xenograft (PDX) mouse lines from engrafted NPC metastatic tumors. Positive staining for EBV-encoded small RNAs confirmed that these tumors harbored EBV, and gene expression profile analyses further showed that the PDX was highly similar to the primary parent tumor. In vivo drug screening using the PDX system demonstrated that gemcitabine had the best antitumor effect among the tested drugs. The donor of this PDX also showed excellent responsiveness to gemcitabine treatment. The combination of gemcitabine and valproic acid exerted synergistic antitumor effects. Further addition of ganciclovir to this two-drug combination regimen enhanced cytolytic viral activation, yielding the best antitumor response among tested regimens. Treatment with this three-drug combination regimen decreased plasma EBV-DNA load, tumor viral concentration, and the number of viable tumor cells to a greater extent than the two-drug gemcitabine and valproic acid combination. These results highlight the value of PDX models in the development of EBV-targeted strategies to treat NPC.
Collapse
Affiliation(s)
- Cheng-Lung Hsu
- Division of Hematology-Oncology, Department of Internal Medicine, Chang Gung Memorial Hospital, Chang Gung University, Taoyuan 333, Taiwan, ROC
| | - Yung-Chia Kuo
- Division of Hematology-Oncology, Department of Internal Medicine, Chang Gung Memorial Hospital, Chang Gung University, Taoyuan 333, Taiwan, ROC
| | - Yenlin Huang
- Department of Pathology, Chang Gung Memorial Hospital, Chang Gung University, Taoyuan 333, Taiwan, ROC
| | - Yin-Cheng Huang
- Division of Neurologic Surgery, Department of Surgery, Chang Gung Memorial Hospital, Chang Gung University, Taoyuan 333, Taiwan, ROC
| | - Kar-Wai Lui
- Department of Medical Imaging and Intervention, Chang Gung Memorial Hospital, Chang Gung University, Taoyuan 333, Taiwan, ROC
| | - Kai-Ping Chang
- Department of Otolaryngology-Head and Neck Surgery, Chang Gung Memorial Hospital, Chang Gung University, Taoyuan 333, Taiwan, ROC
| | - Tung-Liang Lin
- Division of Hematology-Oncology, Department of Internal Medicine, Chang Gung Memorial Hospital, Chang Gung University, Taoyuan 333, Taiwan, ROC
| | - Hsien-Chi Fan
- Division of Hematology-Oncology, Department of Internal Medicine, Chang Gung Memorial Hospital, Chang Gung University, Taoyuan 333, Taiwan, ROC
| | - An-Chi Lin
- Division of Hematology-Oncology, Department of Internal Medicine, Chang Gung Memorial Hospital, Chang Gung University, Taoyuan 333, Taiwan, ROC
| | - Chia-Hsun Hsieh
- Division of Hematology-Oncology, Department of Internal Medicine, Chang Gung Memorial Hospital, Chang Gung University, Taoyuan 333, Taiwan, ROC
| | - Li-Yu Lee
- Department of Pathology, Chang Gung Memorial Hospital, Chang Gung University, Taoyuan 333, Taiwan, ROC
| | - Hung-Ming Wang
- Division of Hematology-Oncology, Department of Internal Medicine, Chang Gung Memorial Hospital, Chang Gung University, Taoyuan 333, Taiwan, ROC
| | - Hsin-Pai Li
- Department of Cell and Molecular Biology, Chang Gung University, Taoyuan 333, Taiwan, ROC
| | - Yu-Sun Chang
- Department of Cell and Molecular Biology, Chang Gung University, Taoyuan 333, Taiwan, ROC
| |
Collapse
|
46
|
Lee HG, Kim H, Kim EJ, Park PG, Dong SM, Choi TH, Kim H, Chong CR, Liu JO, Chen J, Ambinder RF, Hayward SD, Park JH, Lee JM. Targeted therapy for Epstein-Barr virus-associated gastric carcinoma using low-dose gemcitabine-induced lytic activation. Oncotarget 2016; 6:31018-29. [PMID: 26427042 PMCID: PMC4741585 DOI: 10.18632/oncotarget.5041] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2015] [Accepted: 08/24/2015] [Indexed: 12/26/2022] Open
Abstract
The constant presence of the viral genome in Epstein-Barr virus (EBV)-associated gastric cancers (EBVaGCs) suggests the applicability of novel EBV-targeted therapies. The antiviral nucleoside drug, ganciclovir (GCV), is effective only in the context of the viral lytic cycle in the presence of EBV-encoded thymidine kinase (TK)/protein kinase (PK) expression. In this study, screening of the Johns Hopkins Drug Library identified gemcitabine as a candidate for combination treatment with GCV. Pharmacological induction of EBV-TK or PK in EBVaGC-originated tumor cells were used to study combination treatment with GCV in vitro and in vivo. Gemcitabine was found to be a lytic inducer via activation of the ataxia telangiectasia-mutated (ATM)/p53 genotoxic stress pathway in EBVaGC. Using an EBVaGC mouse model and a [125I] fialuridine (FIAU)-based lytic activation imaging system, we evaluated gemcitabine-induced lytic activation in an in vivo system and confirmed the efficacy of gemcitabine-GCV combination treatment. This viral enzyme-targeted anti-tumor strategy may provide a new therapeutic approach for EBVaGCs.
Collapse
Affiliation(s)
- Hyun Gyu Lee
- Department of Microbiology and Immunology, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Hyemi Kim
- Department of Microbiology and Immunology, Yonsei University College of Medicine, Seoul, Republic of Korea.,Brain Korea 21 PLUS Project for Medical Sciences, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Eun Jung Kim
- Radiopharmaceutical Research Team, Korea Institute of Radiological and Medical Sciences, Seoul, Republic of Korea
| | - Pil-Gu Park
- Department of Microbiology and Immunology, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Seung Myung Dong
- Research Institute, National Cancer Center, Goyang, Gyeonggi-do, Republic of Korea
| | - Tae Hyun Choi
- Radiopharmaceutical Research Team, Korea Institute of Radiological and Medical Sciences, Seoul, Republic of Korea
| | - Hyunki Kim
- Department of Pathology, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Curtis R Chong
- Lowe Center for Thoracic Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, MA, USA.,Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts, MA, USA
| | - Jun O Liu
- Department of Pharmacology and Molecular Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA.,Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Jianmeng Chen
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Richard F Ambinder
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - S Diane Hayward
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Jeon Han Park
- Department of Microbiology and Immunology, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Jae Myun Lee
- Department of Microbiology and Immunology, Yonsei University College of Medicine, Seoul, Republic of Korea.,Brain Korea 21 PLUS Project for Medical Sciences, Yonsei University College of Medicine, Seoul, Republic of Korea
| |
Collapse
|
47
|
Astakhova L, Ngara M, Babich O, Prosekov A, Asyakina L, Dyshlyuk L, Midtvedt T, Zhou X, Ernberg I, Matskova L. Short Chain Fatty Acids (SCFA) Reprogram Gene Expression in Human Malignant Epithelial and Lymphoid Cells. PLoS One 2016; 11:e0154102. [PMID: 27441625 PMCID: PMC4956219 DOI: 10.1371/journal.pone.0154102] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2015] [Accepted: 04/09/2016] [Indexed: 11/25/2022] Open
Abstract
The effect of short chain fatty acids (SCFAs) on gene expression in human, malignant cell lines was investigated, with a focus on signaling pathways. The commensal microbial flora produce high levels of SCFAs with established physiologic effects in humans. The most abundant SCFA metabolite in the human microflora is n-butyric acid. It is well known to activate endogenous latent Epstein-Barr virus (EBV), that was used as a reference read out system and extended to EBV+ epithelial cancer cell lines. N-butyric acid and its salt induced inflammatory and apoptotic responses in tumor cells of epithelial and lymphoid origin. Epithelial cell migration was inhibited. The n-butyric gene activation was reduced by knock-down of the cell membrane transporters MCT-1 and -4 by siRNA. N-butyric acid show biologically significant effects on several important cellular functions, also with relevance for tumor cell phenotype.
Collapse
Affiliation(s)
- Lidiia Astakhova
- Institute of Food Science and Technology, Kemerovo, Russia
- Department of Microbiology, Tumor and Cell Biology (MTC), Karolinska Institutet, Stockholm, Sweden
| | - Mtakai Ngara
- Department of Cell and Molecular Biology (CMB), Ludwig Institute for Cancer Research (LICR), Karolinska Institutet, Stockholm, Sweden
| | - Olga Babich
- Institute of Food Science and Technology, Kemerovo, Russia
| | | | | | | | - Tore Midtvedt
- Department of Microbiology, Tumor and Cell Biology (MTC), Karolinska Institutet, Stockholm, Sweden
| | - Xiaoying Zhou
- Department of Microbiology, Tumor and Cell Biology (MTC), Karolinska Institutet, Stockholm, Sweden
| | - Ingemar Ernberg
- Department of Microbiology, Tumor and Cell Biology (MTC), Karolinska Institutet, Stockholm, Sweden
| | - Liudmila Matskova
- Department of Microbiology, Tumor and Cell Biology (MTC), Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
48
|
Du Y, Yu J, Du L, Tang J, Feng WH. Cordycepin enhances Epstein–Barr virus lytic infection and Epstein–Barr virus-positive tumor treatment efficacy by doxorubicin. Cancer Lett 2016; 376:240-8. [DOI: 10.1016/j.canlet.2016.04.001] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2015] [Revised: 03/31/2016] [Accepted: 04/01/2016] [Indexed: 12/12/2022]
|
49
|
van Diemen FR, Kruse EM, Hooykaas MJG, Bruggeling CE, Schürch AC, van Ham PM, Imhof SM, Nijhuis M, Wiertz EJHJ, Lebbink RJ. CRISPR/Cas9-Mediated Genome Editing of Herpesviruses Limits Productive and Latent Infections. PLoS Pathog 2016; 12:e1005701. [PMID: 27362483 PMCID: PMC4928872 DOI: 10.1371/journal.ppat.1005701] [Citation(s) in RCA: 198] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2015] [Accepted: 05/23/2016] [Indexed: 12/15/2022] Open
Abstract
Herpesviruses infect the majority of the human population and can cause significant morbidity and mortality. Herpes simplex virus (HSV) type 1 causes cold sores and herpes simplex keratitis, whereas HSV-2 is responsible for genital herpes. Human cytomegalovirus (HCMV) is the most common viral cause of congenital defects and is responsible for serious disease in immuno-compromised individuals. Epstein-Barr virus (EBV) is associated with infectious mononucleosis and a broad range of malignancies, including Burkitt’s lymphoma, nasopharyngeal carcinoma, Hodgkin’s disease, and post-transplant lymphomas. Herpesviruses persist in their host for life by establishing a latent infection that is interrupted by periodic reactivation events during which replication occurs. Current antiviral drug treatments target the clinical manifestations of this productive stage, but they are ineffective at eliminating these viruses from the infected host. Here, we set out to combat both productive and latent herpesvirus infections by exploiting the CRISPR/Cas9 system to target viral genetic elements important for virus fitness. We show effective abrogation of HCMV and HSV-1 replication by targeting gRNAs to essential viral genes. Simultaneous targeting of HSV-1 with multiple gRNAs completely abolished the production of infectious particles from human cells. Using the same approach, EBV can be almost completely cleared from latently infected EBV-transformed human tumor cells. Our studies indicate that the CRISPR/Cas9 system can be effectively targeted to herpesvirus genomes as a potent prophylactic and therapeutic anti-viral strategy that may be used to impair viral replication and clear latent virus infection. Herpesviruses are large DNA viruses that are carried by almost 100% of the adult human population. Herpesviruses include several important human pathogens, such as herpes simplex viruses (HSV) type 1 and 2 (causing cold sores and genital herpes, respectively), human cytomegalovirus (HCMV; the most common viral cause of congenital defects, and responsible for serious disease in immuno-compromised individuals), and Epstein-Barr virus (EBV; associated with infectious mononucleosis and a wide range of malignancies). Current antiviral drug treatments are not effective in clearing herpesviruses from infected individuals. Therefore, there is a need for alternative strategies to combat these pathogenic viruses and prevent or cure herpesvirus-associated diseases. Here, we have assessed whether a direct attack of herpesvirus genomes within virus-infected cells can inactivate these viruses. For this, we have made use of the recently developed CRISPR/Cas9 genome-engineering system to target and alter specific regions within the genome of these viruses. By targeting sites in the genomes of three different herpesviruses (HSV-1, HCMV, and EBV), we show complete inhibition of viral replication and in some cases even eradication of the viral genomes from infected cells. The findings presented in this study open new avenues for the development of therapeutic strategies to combat pathogenic human herpesviruses using novel genome-engineering technologies.
Collapse
Affiliation(s)
- Ferdy R. van Diemen
- Medical Microbiology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Elisabeth M. Kruse
- Medical Microbiology, University Medical Center Utrecht, Utrecht, The Netherlands
| | | | | | - Anita C. Schürch
- Medical Microbiology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Petra M. van Ham
- Medical Microbiology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Saskia M. Imhof
- Department of Ophthalmology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Monique Nijhuis
- Medical Microbiology, University Medical Center Utrecht, Utrecht, The Netherlands
| | | | - Robert Jan Lebbink
- Medical Microbiology, University Medical Center Utrecht, Utrecht, The Netherlands
- * E-mail:
| |
Collapse
|
50
|
Treece AL, Duncan DL, Tang W, Elmore S, Morgan DR, Dominguez RL, Speck O, Meyers MO, Gulley ML. Gastric adenocarcinoma microRNA profiles in fixed tissue and in plasma reveal cancer-associated and Epstein-Barr virus-related expression patterns. J Transl Med 2016; 96:661-71. [PMID: 26950485 PMCID: PMC5767475 DOI: 10.1038/labinvest.2016.33] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2015] [Revised: 12/09/2015] [Accepted: 01/12/2016] [Indexed: 12/27/2022] Open
Abstract
MicroRNA expression in formalin-fixed paraffin-embedded tissue (FFPE) or plasma may add value for cancer management. The GastroGenus miR Panel was developed to measure 55 cancer-specific human microRNAs, Epstein-Barr virus (EBV)-encoded microRNAs, and controls. This Q-rtPCR panel was applied to 100 FFPEs enriched for adenocarcinoma or adjacent non-malignant mucosa, and to plasma of 31 patients. In FFPE, microRNAs upregulated in malignant versus adjacent benign gastric mucosa were hsa-miR-21, -155, -196a, -196b, -185, and -let-7i. Hsa-miR-18a, 34a, 187, -200a, -423-3p, -484, and -744 were downregulated. Plasma of cancer versus non-cancer controls had upregulated hsa-miR-23a, -103, and -221 and downregulated hsa-miR-378, -346, -486-5p, -200b, -196a, -141, and -484. EBV-infected versus uninfected cancers expressed multiple EBV-encoded microRNAs, and concomitant dysregulation of four human microRNAs suggests that viral infection may alter cellular biochemical pathways. Human microRNAs were dysregulated between malignant and benign gastric mucosa and between plasma of cancer patients and non-cancer controls. Strong association of EBV microRNA expression with known EBV status underscores the ability of microRNA technology to reflect disease biology. Expression of viral microRNAs in concert with unique human microRNAs provides novel insights into viral oncogenesis and reinforces the potential for microRNA profiles to aid in classifying gastric cancer subtypes. Pilot studies of plasma suggest the potential for a noninvasive addition to cancer diagnostics.
Collapse
MESH Headings
- Adenocarcinoma/genetics
- Adenocarcinoma/metabolism
- Adenocarcinoma/virology
- Aged
- Aged, 80 and over
- Case-Control Studies
- Epstein-Barr Virus Infections/genetics
- Epstein-Barr Virus Infections/metabolism
- Epstein-Barr Virus Infections/virology
- Female
- Gene Expression Profiling
- Gene Expression Regulation, Neoplastic
- Herpesvirus 4, Human/genetics
- Herpesvirus 4, Human/isolation & purification
- Humans
- Male
- MicroRNAs/blood
- MicroRNAs/genetics
- MicroRNAs/metabolism
- Middle Aged
- Pilot Projects
- RNA, Neoplasm/blood
- RNA, Neoplasm/genetics
- RNA, Neoplasm/metabolism
- RNA, Viral/blood
- RNA, Viral/genetics
- RNA, Viral/metabolism
- Stomach Neoplasms/genetics
- Stomach Neoplasms/metabolism
- Stomach Neoplasms/virology
Collapse
Affiliation(s)
- Amanda L Treece
- Department of Pathology and Laboratory Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Daniel L Duncan
- Department of Pathology and Laboratory Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Weihua Tang
- Department of Pathology and Laboratory Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Sandra Elmore
- Department of Pathology and Laboratory Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Douglas R Morgan
- Division of Gastroenterology, Hepatology, and Nutrition; Department of Medicine, Vanderbilt University, Nashville, TN, USA
| | - Ricardo L Dominguez
- Department of Gastroenterology, Western Regional Hospital, Santa Rosa de Copan, Honduras
| | - Olga Speck
- Department of Pathology and Laboratory Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Michael O Meyers
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Division of Surgical Oncology, Department of Surgery, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Margaret L Gulley
- Department of Pathology and Laboratory Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| |
Collapse
|