1
|
Zhang W, Zeng M, Li Y, Yu L. Leveraging oncovirus-derived antigen against the viral malignancies in adoptive cell therapies. Biomark Res 2024; 12:71. [PMID: 39075601 PMCID: PMC11287861 DOI: 10.1186/s40364-024-00617-6] [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: 05/09/2024] [Accepted: 07/10/2024] [Indexed: 07/31/2024] Open
Abstract
Adoptive cell therapies (ACTs) have revolutionized cancer immunotherapy, prompting exploration into their application against oncoviruses. Oncoviruses such as human papillomavirus (HPV), hepatitis B virus (HBV), hepatitis C virus (HCV), and Epstein-Barr virus (EBV) contribute significantly (12-25%) to human malignancies through direct or indirect oncogenic mechanisms. These viruses persistently or latently infect cells, disrupt cellular homeostasis and pathways, challenging current antiviral treatment paradigms. Moreover, viral infections pose additional risks in the setting of long-term cancer therapy and lead to morbidity and mortality. Virally encoded oncoproteins, which are tumor-restricted, immunologically foreign, and even uniformly expressed, represent promising targets for patient-tailored ACTs. This review elucidates the rationale for leveraging viral antigen-specific ACTs in combating viral-associated malignancies. On this basis, ongoing preclinical studies consolidate our understanding of harnessing ACTs against viral malignancies, underscoring their potential to eradicate viruses implicated in cancer progression. Furthermore, we scrutinize the current landscape of clinical trials focusing on virus-specific ACTs and discuss their implications for therapeutic advancement.
Collapse
Affiliation(s)
- Wei Zhang
- Department of Hematology and Oncology, Shenzhen University General Hospital, International Cancer Center, Hematology Institution of Shenzhen University, Shenzhen University Medical School, Shenzhen University, Shenzhen, Guangdong, 518000, China
- Guangdong Key Laboratory for Biomedical Measurements and Ultrasound Imaging, National-Regional Key Technology Engineering Laboratory for Medical Ultrasound, School of Biomedical Engineering, Shenzhen University Medical school, Shenzhen, 518060, China
| | - Miao Zeng
- Department of Hematology and Oncology, Shenzhen University General Hospital, International Cancer Center, Hematology Institution of Shenzhen University, Shenzhen University Medical School, Shenzhen University, Shenzhen, Guangdong, 518000, China
- Guangdong Key Laboratory for Biomedical Measurements and Ultrasound Imaging, National-Regional Key Technology Engineering Laboratory for Medical Ultrasound, School of Biomedical Engineering, Shenzhen University Medical school, Shenzhen, 518060, China
| | - Yisheng Li
- Shenzhen Haoshi Biotechnology Co., Ltd, No. 155 Hongtian Road, Xinqiao Street, Bao'an District, Shenzhen, Guangdong, 518125, China
- Haoshi Cell Therapy Institute, Shenzhen University, Shenzhen, China
| | - Li Yu
- Department of Hematology and Oncology, Shenzhen University General Hospital, International Cancer Center, Hematology Institution of Shenzhen University, Shenzhen University Medical School, Shenzhen University, Shenzhen, Guangdong, 518000, China.
- Haoshi Cell Therapy Institute, Shenzhen University, Shenzhen, China.
| |
Collapse
|
2
|
Debelec-Butuner B, Quitt O, Schreiber S, Momburg F, Wisskirchen K, Protzer U. Activation of distinct antiviral T-cell immunity: A comparison of bi- and trispecific T-cell engager antibodies with a chimeric antigen receptor targeting HBV envelope proteins. Front Immunol 2022; 13:1029214. [DOI: 10.3389/fimmu.2022.1029214] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2022] [Accepted: 10/10/2022] [Indexed: 11/06/2022] Open
Abstract
Despite the availability of an effective prophylactic vaccine, 820,000 people die annually of hepatitis B virus (HBV)-related liver disease according to WHO. Since current antiviral therapies do not provide a curative treatment for the 296 million HBV carriers around the globe, novel strategies to cure HBV are urgently needed. A promising approach is the redirection of T cells towards HBV-infected hepatocytes employing chimeric antigen receptors or T-cell engager antibodies. We recently described the effective redirection of T cells employing a second-generation chimeric antigen receptor directed against the envelope protein of hepatitis B virus on the surface of infected cells (S-CAR) as well as bispecific antibodies that engage CD3 or CD28 on T cells employing the identical HBV envelope protein (HBVenv) binder. In this study, we added a trispecific antibody comprising all three moieties to the tool-box. Cytotoxic and non-cytolytic antiviral activities of these bi- and trispecific T-cell engager antibodies were assessed in co-cultures of human PBMC with HBV-positive hepatoma cells, and compared to that of S-CAR-grafted T cells. Activation of T cells via the S-CAR or by either a combination of the CD3- and CD28-targeting bispecific antibodies or the trispecific antibody allowed for specific elimination of HBV-positive target cells. While S-CAR-grafted effector T cells displayed faster killing kinetics, combinatory treatment with the bispecific antibodies or single treatment with the trispecific antibody was associated with a more pronounced cytokine release. Clearance of viral antigens and elimination of the HBV persistence form, the covalently closed circular (ccc) DNA, through cytolytic as well as cytokine-mediated activity was observed in all three settings with the combination of bispecific antibodies showing the strongest non-cytolytic, cytokine-mediated antiviral effect. Taken together, we demonstrate that bi- and trispecific T-cell engager antibodies can serve as a potent, off-the-shelf alternative to S-CAR-grafted T cells to cure HBV.
Collapse
|
3
|
Lefeuvre C, Le Guillou-Guillemette H, Ducancelle A. A Pleiotropic Role of the Hepatitis B Virus Core Protein in Hepatocarcinogenesis. Int J Mol Sci 2021; 22:ijms222413651. [PMID: 34948447 PMCID: PMC8707456 DOI: 10.3390/ijms222413651] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2021] [Revised: 12/18/2021] [Accepted: 12/19/2021] [Indexed: 02/07/2023] Open
Abstract
Chronic hepatitis B virus (HBV) infection is one of the most common factors associated with hepatocellular carcinoma (HCC), which is the sixth most prevalent cancer among all cancers worldwide. However, the pathogenesis of HBV-mediated hepatocarcinogenesis is unclear. Evidence currently available suggests that the HBV core protein (HBc) plays a potential role in the development of HCC, such as the HBV X protein. The core protein, which is the structural component of the viral nucleocapsid, contributes to almost every stage of the HBV life cycle and occupies diverse roles in HBV replication and pathogenesis. Recent studies have shown that HBc was able to disrupt various pathways involved in liver carcinogenesis: the signaling pathways implicated in migration and proliferation of hepatoma cells, apoptosis pathways, and cell metabolic pathways inducing the development of HCC; and the immune system, through the expression and production of proinflammatory cytokines. In addition, HBc can modulate normal functions of hepatocytes through disrupting human host gene expression by binding to promoter regions. This HBV protein also promotes HCC metastasis through epigenetic alterations, such as micro-RNA. This review focuses on the molecular pathogenesis of the HBc protein in HBV-induced HCC.
Collapse
Affiliation(s)
- Caroline Lefeuvre
- Laboratoire de Virologie, Département de Biologie des Agents Infectieux, CHU Angers, F-49000 Angers, France; (H.L.G.-G.); (A.D.)
- HIFIH Laboratory UPRES EA3859, SFR ICAT 4208, Angers University, F-49000 Angers, France
- Correspondence:
| | - Hélène Le Guillou-Guillemette
- Laboratoire de Virologie, Département de Biologie des Agents Infectieux, CHU Angers, F-49000 Angers, France; (H.L.G.-G.); (A.D.)
- HIFIH Laboratory UPRES EA3859, SFR ICAT 4208, Angers University, F-49000 Angers, France
| | - Alexandra Ducancelle
- Laboratoire de Virologie, Département de Biologie des Agents Infectieux, CHU Angers, F-49000 Angers, France; (H.L.G.-G.); (A.D.)
- HIFIH Laboratory UPRES EA3859, SFR ICAT 4208, Angers University, F-49000 Angers, France
| |
Collapse
|
4
|
Liu PJ, Harris JM, Marchi E, D'Arienzo V, Michler T, Wing PAC, Magri A, Ortega-Prieto AM, van de Klundert M, Wettengel J, Durantel D, Dorner M, Klenerman P, Protzer U, Giotis ES, McKeating JA. Hypoxic gene expression in chronic hepatitis B virus infected patients is not observed in state-of-the-art in vitro and mouse infection models. Sci Rep 2020; 10:14101. [PMID: 32839523 PMCID: PMC7445281 DOI: 10.1038/s41598-020-70865-7] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2020] [Accepted: 07/31/2020] [Indexed: 02/07/2023] Open
Abstract
Hepatitis B virus (HBV) is the leading cause of hepatocellular carcinoma (HCC) worldwide. The prolyl hydroxylase domain (PHD)-hypoxia inducible factor (HIF) pathway is a key mammalian oxygen sensing pathway and is frequently perturbed by pathological states including infection and inflammation. We discovered a significant upregulation of hypoxia regulated gene transcripts in patients with chronic hepatitis B (CHB) in the absence of liver cirrhosis. We used state-of-the-art in vitro and in vivo HBV infection models to evaluate a role for HBV infection and the viral regulatory protein HBx to drive HIF-signalling. HBx had no significant impact on HIF expression or associated transcriptional activity under normoxic or hypoxic conditions. Furthermore, we found no evidence of hypoxia gene expression in HBV de novo infection, HBV infected human liver chimeric mice or transgenic mice with integrated HBV genome. Collectively, our data show clear evidence of hypoxia gene induction in CHB that is not recapitulated in existing models for acute HBV infection, suggesting a role for inflammatory mediators in promoting hypoxia gene expression.
Collapse
Affiliation(s)
- Peter Jianrui Liu
- Nuffield Department of Medicine Research Building, University of Oxford, Oxford, OX3 7LF, UK
| | - James M Harris
- Nuffield Department of Medicine Research Building, University of Oxford, Oxford, OX3 7LF, UK
| | - Emanuele Marchi
- Medawar Building, University of Oxford, South Parks Road, Oxford, OX1 3SY, UK
| | - Valentina D'Arienzo
- Nuffield Department of Medicine Research Building, University of Oxford, Oxford, OX3 7LF, UK
| | - Thomas Michler
- Institute of Virology, Technical University of Munich/Helmholtz Zentrum München, Trogerstrasse 30, 81675, Munich, Germany
| | - Peter A C Wing
- Nuffield Department of Medicine Research Building, University of Oxford, Oxford, OX3 7LF, UK
| | - Andrea Magri
- Nuffield Department of Medicine Research Building, University of Oxford, Oxford, OX3 7LF, UK
| | - Ana Maria Ortega-Prieto
- Section of Molecular Virology, Department of Infectious Diseases, Imperial College London, London, W2 1PG, UK
| | - Maarten van de Klundert
- Institute of Virology, Technical University of Munich/Helmholtz Zentrum München, Trogerstrasse 30, 81675, Munich, Germany
| | - Jochen Wettengel
- Institute of Virology, Technical University of Munich/Helmholtz Zentrum München, Trogerstrasse 30, 81675, Munich, Germany
| | - David Durantel
- Cancer Research Center of Lyon (CRCL), INSERM U1052, and University of Lyon (UCBL1), Lyon, France
| | - Marcus Dorner
- Section of Molecular Virology, Department of Infectious Diseases, Imperial College London, London, W2 1PG, UK
| | - Paul Klenerman
- Medawar Building, University of Oxford, South Parks Road, Oxford, OX1 3SY, UK
| | - Ulrike Protzer
- Institute of Virology, Technical University of Munich/Helmholtz Zentrum München, Trogerstrasse 30, 81675, Munich, Germany
| | - Efstathios S Giotis
- Section of Molecular Virology, Department of Infectious Diseases, Imperial College London, London, W2 1PG, UK
- School of Life Sciences, University of Essex, Colchester, C04 3SQ, UK
| | - Jane A McKeating
- Nuffield Department of Medicine Research Building, University of Oxford, Oxford, OX3 7LF, UK.
| |
Collapse
|
5
|
Zhang Y, Li J, Wang S, Yang F, Zhou Y, Liu Y, Zhu W, Shi X. HBx‑associated long non‑coding RNA activated by TGF‑β promotes cell invasion and migration by inducing autophagy in primary liver cancer. Int J Oncol 2019; 56:337-347. [PMID: 31746419 DOI: 10.3892/ijo.2019.4908] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2018] [Accepted: 03/07/2019] [Indexed: 12/25/2022] Open
Abstract
Hepatitis B virus (HBV) x protein (HBx) has been reported as the primary pathogenic factor involved in HBV‑related liver cancer; however, the mechanisms underlying how HBx promotes tumor‑associated invasion and metastasis remain unclear. Long noncoding RNA activated by transforming growth factor (TGF)‑β (lncRNA‑ATB) is a novel oncogenic lncRNA stimulated by TGF‑β, which is closely associated with the invasion and metastasis of liver cancer. In the present study, whether lncRNA‑ATB was involved in HBx‑mediated hepatocarcinogenesis was investigated. The expression of lncRNA‑ATB in 26 primary liver cancer tissues and lentivirus transfected HBx‑HepG2 cell lines was detected, and it was revealed that more advanced tumor‑node‑metastasis staging and increased expression of lncRNA‑ATB in liver cancer tissues were significantly associated with HBV infection. It was further demonstrated that the expression levels of lncRNA‑ATB and TGF‑β were elevated in HepG2 cells following HBx‑vector transfection, which was accompanied with increased autophagy. Conversely, knockdown of lncRNA‑ATB or TGF‑β could suppress this effect. Furthermore, such suppression on autophagy in HepG2 cells could be alleviated by the induction of starvation. In addition, the invasive and migration abilities of HBx‑HepG2 cells were increased compared with HepG2 cells, while knockdown of lncRNA‑ATB or TGF‑β could reduce these abilities. In conclusion, the results of the present study revealed that HBx was closely associated with oncogenic lncRNA‑ATB. HBx‑induced autophagy could upregulate the expression of TGF‑β and lncRNA‑ATB. This may be considered to be a potential mechanism underlying HBV‑induced hepatocarcinogenesis.
Collapse
Affiliation(s)
- Yuheng Zhang
- Department of General Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, Jiangsu 210008, P.R. China
| | - Jun Li
- Department of General Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230022, P.R. China
| | - Shuai Wang
- Department of General Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, Jiangsu 210008, P.R. China
| | - Faji Yang
- Department of General Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, Jiangsu 210008, P.R. China
| | - Yuan Zhou
- Department of General Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, Jiangsu 210008, P.R. China
| | - Yang Liu
- Department of General Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, Jiangsu 210008, P.R. China
| | - Wei Zhu
- Department of Anesthesiology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, Jiangsu 210008, P.R. China
| | - Xiaolei Shi
- Department of General Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, Jiangsu 210008, P.R. China
| |
Collapse
|
6
|
Hepatitis B Virus X Protein Function Requires Zinc Binding. J Virol 2019; 93:JVI.00250-19. [PMID: 31167910 PMCID: PMC6675892 DOI: 10.1128/jvi.00250-19] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2019] [Accepted: 05/28/2019] [Indexed: 12/18/2022] Open
Abstract
The host structural maintenance of chromosomes 5/6 complex (Smc5/6) suppresses hepatitis B virus (HBV) transcription. HBV counters this restriction by expressing the X protein (HBx), which redirects the cellular DNA damage-binding protein 1 (DDB1)-containing E3 ubiquitin ligase to target Smc5/6 for degradation. However, the details of how HBx modulates the interaction between DDB1 and Smc5/6 remain to be determined. In this study, we performed biophysical analyses of recombinant HBx and functional analysis of HBx mutants in HBV-infected primary human hepatocytes (PHH) to identify key regions and residues that are required for HBx function. We determined that recombinant HBx is soluble and exhibits stoichiometric zinc binding when expressed in the presence of DDB1. Mass spectrometry-based hydrogen-deuterium exchange and cysteine-specific chemical footprinting of the HBx:DDB1 complex identified several HBx cysteine residues (located between amino acids 61 and 137) that are likely involved in zinc binding. These cysteine residues did not form disulfide bonds in HBx expressed in human cells. In line with the biophysical data, functional analysis demonstrated that HBx amino acids 45 to 140 are required for Smc6 degradation and HBV transcription in PHH. Furthermore, site-directed mutagenesis determined that C61, C69, C137, and H139 are necessary for HBx function, although they are likely not essential for DDB1 binding. This CCCH motif is highly conserved in HBV as well as in the X proteins from various mammalian hepadnaviruses. Collectively, our data indicate that the essential HBx cysteine and histidine residues form a zinc-binding motif that is required for HBx function.IMPORTANCE The structural maintenance of chromosomes 5/6 complex (Smc5/6) is a host restriction factor that suppresses HBV transcription. HBV counters this restriction by expressing HBV X protein (HBx), which redirects a host ubiquitin ligase to target Smc5/6 for degradation. Despite this recent advance in understanding HBx function, the key regions and residues of HBx required for Smc5/6 degradation have not been determined. In the present study, we performed biochemical, biophysical, and cell-based analyses of HBx. By doing so, we mapped the minimal functional region of HBx and identified a highly conserved CCCH motif in HBx that is likely responsible for coordinating zinc and is essential for HBx function. We also developed a method to produce soluble recombinant HBx protein that likely adopts a physiologically relevant conformation. Collectively, this study provides new insights into the HBx structure-function relationship and suggests a new approach for structural studies of this enigmatic viral regulatory protein.
Collapse
|
7
|
VEGF Upregulation in Viral Infections and Its Possible Therapeutic Implications. Int J Mol Sci 2018; 19:ijms19061642. [PMID: 29865171 PMCID: PMC6032371 DOI: 10.3390/ijms19061642] [Citation(s) in RCA: 57] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2018] [Revised: 05/28/2018] [Accepted: 05/29/2018] [Indexed: 12/12/2022] Open
Abstract
Several viruses are recognized as the direct or indirect causative agents of human tumors and other severe human diseases. Vascular endothelial growth factor (VEGF) is identified as a principal proangiogenic factor that enhances the production of new blood vessels from existing vascular network. Therefore, oncogenic viruses such as Kaposi’s sarcoma herpesvirus (KSHV) and Epstein-Barr virus (EBV) and non-oncogenic viruses such as herpes simplex virus (HSV-1) and dengue virus, which lack their own angiogenic factors, rely on the recruitment of cellular genes for angiogenesis in tumor progression or disease pathogenesis. This review summarizes how human viruses exploit the cellular signaling machinery to upregulate the expression of VEGF and benefit from its physiological functions for their own pathogenesis. Understanding the interplay between viruses and VEGF upregulation will pave the way to design targeted and effective therapeutic approaches for viral oncogenesis and severe diseases.
Collapse
|
8
|
Yılmaz Susluer S, Kayabasi C, Ozmen Yelken B, Asik A, Celik D, Balci Okcanoglu T, Serin Senger S, Biray Avci C, Kose S, Gunduz C. Analysis of long non-coding RNA (lncRNA) expression in hepatitis B patients. Bosn J Basic Med Sci 2018; 18:150-161. [PMID: 29669510 DOI: 10.17305/bjbms.2018.2800] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2017] [Revised: 02/16/2018] [Accepted: 02/17/2018] [Indexed: 12/28/2022] Open
Abstract
Long non-coding RNAs (lncRNAs) have been implicated in numerous biological processes, including epigenetic regulation, cell-cycle control, and transcriptional/translational regulation of gene expression. Differential expression of lncRNAs and disruption of the regulatory processes are recognized as critical steps in cancer development. The role of lncRNAs in hepatitis B virus (HBV) infection is not well understood. Here we analyzed the expression of 135 lncRNAs in plasma samples of 82 HBV patients (classified as chronic patients, inactive carriers, or resolved patients) at diagnosis and at 12 months of treatment in relation to control group (81 healthy volunteers). We also investigated the effect of small interfering RNA (siRNA)-mediated silencing of lincRNA-SFMBT2 on HBV-positive human liver cancer cell line. lncRNA expression was analyzed by quantitative reverse transcription-polymerase chain reaction (qRT-PCR). Chemically synthesized siRNAs were transfected into the cell lines using Lipofectamine 2000 Reagent (Thermo Fisher Scientific). HBV DNA and HBsAg and HBeAg were detected in transfected cultures by real-time PCR and ELISA, respectively, using commercial kits. We observed changes in lncRNA expression in all three HBV groups, compared to control group. Most notably, the expression of anti-NOS2A, lincRNA-SFMBT2, and Zfhx2as was significantly increased and expression of Y5 lncRNA was decreased in chronic HBV patients. A decreased Y5 expression and increased lincRNA-SFMBT2 expression were observed in inactive HBsAg carriers. The expression of HOTTIP, MEG9, and PCAT-32 was increased in resolved HBV patients, and no significant change in the expression of Y5 was observed, compared to control group. siRNA-mediated inhibition of lincRNA-SFMBT2 decreased the level of HBV DNA in human liver cancer cells. Further research is needed to confirm the prognostic as well as therapeutic role of these lncRNAs in HBV patients.
Collapse
Affiliation(s)
- Sunde Yılmaz Susluer
- Department of Medical Biology, Faculty of Medicine, Ege University, Izmir, Turkey.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
9
|
DNA Tumor Virus Regulation of Host DNA Methylation and Its Implications for Immune Evasion and Oncogenesis. Viruses 2018; 10:v10020082. [PMID: 29438328 PMCID: PMC5850389 DOI: 10.3390/v10020082] [Citation(s) in RCA: 53] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2018] [Revised: 02/07/2018] [Accepted: 02/07/2018] [Indexed: 12/11/2022] Open
Abstract
Viruses have evolved various mechanisms to evade host immunity and ensure efficient viral replication and persistence. Several DNA tumor viruses modulate host DNA methyltransferases for epigenetic dysregulation of immune-related gene expression in host cells. The host immune responses suppressed by virus-induced aberrant DNA methylation are also frequently involved in antitumor immune responses. Here, we describe viral mechanisms and virus–host interactions by which DNA tumor viruses regulate host DNA methylation to evade antiviral immunity, which may contribute to the generation of an immunosuppressive microenvironment during cancer development. Recent trials of immunotherapies have shown promising results to treat multiple cancers; however, a significant number of non-responders necessitate identifying additional targets for cancer immunotherapies. Thus, understanding immune evasion mechanisms of cancer-causing viruses may provide great insights for reversing immune suppression to prevent and treat associated cancers.
Collapse
|
10
|
Caccuri F, Giordano F, Barone I, Mazzuca P, Giagulli C, Andò S, Caruso A, Marsico S. HIV-1 matrix protein p17 and its variants promote human triple negative breast cancer cell aggressiveness. Infect Agent Cancer 2017; 12:49. [PMID: 29021819 PMCID: PMC5613317 DOI: 10.1186/s13027-017-0160-7] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2017] [Accepted: 09/14/2017] [Indexed: 12/22/2022] Open
Abstract
Background The introduction of cART has changed the morbidity and mortality patterns affecting HIV-infected (HIV+) individuals. The risk of breast cancer in HIV+ patients has now approached the general population risk. However, breast cancer has a more aggressive clinical course and poorer outcome in HIV+ patients than in general population, without correlation with the CD4 or virus particles count. These findings suggest a likely influence of HIV-1 proteins on breast cancer aggressiveness and progression. The HIV-1 matrix protein (p17) is expressed in different tissues and organs of successfully cART-treated patients and promotes migration of different cells. Variants of p17 (vp17s), characterized by mutations and amino acid insertions, differently from the prototype p17 (refp17), also promote B-cell proliferation and transformation. Methods Wound-healing assay, matrigel-based invasion assay, and anchorage-independent proliferation assay were employed to compare the biological activity exerted by refp17 and three different vp17s on the triple-negative human breast cancer cell line MDA-MB 231. Intracellular signaling was investigated by western blot analysis. Results Motility and invasiveness increased in cells treated with both refp17 and vp17s compared to untreated cells. The effects of the viral proteins were mediated by binding to the chemokine receptor CXCR2 and activation of the ERK1/2 signaling pathway. However, vp17s promoted MDA-MB 231 cell growth and proliferation in contrast to refp17-treated or not treated cells. Conclusions In the context of the emerging role of the microenvironment in promoting and supporting cancer cell growth and metastatic spreading, here we provide the first evidence that exogenous p17 may play a crucial role in sustaining breast cancer cell migration and invasiveness, whereas some p17 variants may also be involved in cancer cell growth and proliferation.
Collapse
Affiliation(s)
- Francesca Caccuri
- Section of Microbiology, Department of Molecular and Translational Medicine, University of Brescia , Brescia, Italy
| | - Francesca Giordano
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Arcavacata di Rende, Italy
| | - Ines Barone
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Arcavacata di Rende, Italy
| | - Pietro Mazzuca
- Section of Microbiology, Department of Molecular and Translational Medicine, University of Brescia , Brescia, Italy
| | - Cinzia Giagulli
- Section of Microbiology, Department of Molecular and Translational Medicine, University of Brescia , Brescia, Italy
| | - Sebastiano Andò
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Arcavacata di Rende, Italy
| | - Arnaldo Caruso
- Section of Microbiology, Department of Molecular and Translational Medicine, University of Brescia , Brescia, Italy
| | - Stefania Marsico
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Arcavacata di Rende, Italy
| |
Collapse
|
11
|
Hepatitis B Virus Activates Signal Transducer and Activator of Transcription 3 Supporting Hepatocyte Survival and Virus Replication. Cell Mol Gastroenterol Hepatol 2017; 4:339-363. [PMID: 28884137 PMCID: PMC5581872 DOI: 10.1016/j.jcmgh.2017.07.003] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/04/2016] [Accepted: 07/13/2017] [Indexed: 02/07/2023]
Abstract
BACKGROUND & AIMS The human hepatitis B virus (HBV) is a major cause of chronic hepatitis and hepatocellular carcinoma, but molecular mechanisms driving liver disease and carcinogenesis are largely unknown. We therefore studied cellular pathways altered by HBV infection. METHODS We performed gene expression profiling of primary human hepatocytes infected with HBV and proved the results in HBV-replicating cell lines and human liver tissue using real-time polymerase chain reaction and Western blotting. Activation of signal transducer and activator of transcription (STAT3) was examined in HBV-replicating human hepatocytes, HBV-replicating mice, and liver tissue from HBV-infected individuals using Western blotting, STAT3-luciferase reporter assay, and immunohistochemistry. The consequences of STAT3 activation on HBV infection and cell survival were studied by chemical inhibition of STAT3 phosphorylation and small interfering RNA-mediated knockdown of STAT3. RESULTS Gene expression profiling of HBV-infected primary human hepatocytes detected no interferon response, while genes encoding for acute phase and antiapoptotic proteins were up-regulated. This gene regulation was confirmed in liver tissue samples of patients with chronic HBV infection and in HBV-related hepatocellular carcinoma. Pathway analysis revealed activation of STAT3 to be the major regulator. Interleukin-6-dependent and -independent activation of STAT3 was detected in HBV-replicating hepatocytes in cell culture and in vivo. Prevention of STAT3 activation by inhibition of Janus tyrosine kinases as well as small interfering RNA-mediated knockdown of STAT3-induced apoptosis and reduced HBV replication and gene expression. CONCLUSIONS HBV activates STAT3 signaling in hepatocytes to foster its own replication but also to prevent apoptosis of infected cells. This very likely supports HBV-related carcinogenesis.
Collapse
Key Words
- APR, acute phase response
- Apoptosis
- CRP, C-reactive protein
- DMSO, dimethyl sulfoxide
- FCS, fetal calf serum
- HBV pg RNA, hepatitis B pregenomic RNA
- HBV, Hepatitis B virus
- HBVtg, hepatitis B transgenic
- HBeAg, hepatitis B early antigen
- HCC, hepatocellular carcinoma
- HNF, hepatocyte nuclear factor
- Hepatitis B Virus Infection
- Hepatocellular Carcinoma
- IFN, interferon
- IL-6, interleukin 6
- IRF3, interferon regulatory factor 3
- NAC, N-acetyl-L-cysteine
- PCR, polymerase chain reaction
- PHH, primary human hepatocyte
- ROS, reactive oxygen species
- RT, reverse transcription
- STAT3 Signaling
- STAT3, signal transducer and activator of transcription 3
- cDNA, complementary DNA
- cRNA, complementary RNA
- cccDNA, covalently closed circular DNA
- mRNA, messenger RNA
- p.i., postinfection
- pSTAT3, phosphorylated signal transducer and activator of transcription 3
- pgRNA, pregenomic RNA
- siRNA, small interfering RNA
Collapse
|
12
|
Glycyrrhetic acid, but not glycyrrhizic acid, strengthened entecavir activity by promoting its subcellular distribution in the liver via efflux inhibition. Eur J Pharm Sci 2017. [PMID: 28627473 DOI: 10.1016/j.ejps.2017.06.015] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Entecavir (ETV) is a superior nucleoside analogue used to treat hepatitis B virus (HBV) infection. Although its advantages over other agents include low viral resistance and the elicitation of a sharp decrease in HBV DNA, adverse effects such as hepatic steatosis, hepatic damage and lactic acidosis have also been reported. Glycyrrhizin has long been used as hepato-protective medicine. The clinical combination of ETV plus glycyrrhizin in China displays better therapeutic effects and lower rates of liver damage. However, there is little evidence explaining the probable synergistic mechanism that exists between these two drugs from a pharmacokinetics view. Here, alterations in the plasma pharmacokinetics, tissue distribution, subcellular distribution, and in vitro and in vivo antiviral activity of ETV after combination with glycyrrhizic acid (GL) were analysed to determine the synergistic mechanisms of these two drugs. Specific efflux transporter membrane vesicles were also used to elucidate their interactions. The primary active GL metabolite, glycyrrhetic acid (GA), did not affect the plasma pharmacokinetics of ETV but promoted its accumulation in hepatocytes, increasing its distribution in the cytoplasm and nucleus and augmenting the antiviral efficiency of ETV. These synergistic actions were primarily due to the inhibitory effect of GA on MRP4 and BCRP, which transport ETV out of hepatocytes. In conclusion, GA interacted with ETV at cellular and subcellular levels in the liver through MRP4 and BCRP inhibition, which enhanced the antiviral activity of ETV. Our results partially explain the synergistic mechanism of ETV and GL from a pharmacokinetics view, providing more data to support the use of these compounds together in clinical HBV treatment.
Collapse
|
13
|
Ko C, Michler T, Protzer U. Novel viral and host targets to cure hepatitis B. Curr Opin Virol 2017; 24:38-45. [DOI: 10.1016/j.coviro.2017.03.019] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2016] [Revised: 03/06/2017] [Accepted: 03/30/2017] [Indexed: 02/07/2023]
|
14
|
Niu C, Livingston CM, Li L, Beran RK, Daffis S, Ramakrishnan D, Burdette D, Peiser L, Salas E, Ramos H, Yu M, Cheng G, Strubin M, Delaney IV WE, Fletcher SP. The Smc5/6 Complex Restricts HBV when Localized to ND10 without Inducing an Innate Immune Response and Is Counteracted by the HBV X Protein Shortly after Infection. PLoS One 2017; 12:e0169648. [PMID: 28095508 PMCID: PMC5240991 DOI: 10.1371/journal.pone.0169648] [Citation(s) in RCA: 110] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2016] [Accepted: 12/20/2016] [Indexed: 02/06/2023] Open
Abstract
The structural maintenance of chromosome 5/6 complex (Smc5/6) is a restriction factor that represses hepatitis B virus (HBV) transcription. HBV counters this restriction by expressing HBV X protein (HBx), which targets Smc5/6 for degradation. However, the mechanism by which Smc5/6 suppresses HBV transcription and how HBx is initially expressed is not known. In this study we characterized viral kinetics and the host response during HBV infection of primary human hepatocytes (PHH) to address these unresolved questions. We determined that Smc5/6 localizes with Nuclear Domain 10 (ND10) in PHH. Co-localization has functional implications since depletion of ND10 structural components alters the nuclear distribution of Smc6 and induces HBV gene expression in the absence of HBx. We also found that HBV infection and replication does not induce a prominent global host transcriptional response in PHH, either shortly after infection when Smc5/6 is present, or at later times post-infection when Smc5/6 has been degraded. Notably, HBV and an HBx-negative virus establish high level infection in PHH without inducing expression of interferon-stimulated genes or production of interferons or other cytokines. Our study also revealed that Smc5/6 is degraded in the majority of infected PHH by the time cccDNA transcription could be detected and that HBx RNA is present in cell culture-derived virus preparations as well as HBV patient plasma. Collectively, these data indicate that Smc5/6 is an intrinsic antiviral restriction factor that suppresses HBV transcription when localized to ND10 without inducing a detectable innate immune response. Our data also suggest that HBx protein may be initially expressed by delivery of extracellular HBx RNA into HBV-infected cells.
Collapse
Affiliation(s)
- Congrong Niu
- Gilead Sciences, Foster City, California, United States of America
| | | | - Li Li
- Gilead Sciences, Foster City, California, United States of America
| | - Rudolf K. Beran
- Gilead Sciences, Foster City, California, United States of America
| | - Stephane Daffis
- Gilead Sciences, Foster City, California, United States of America
| | | | - Dara Burdette
- Gilead Sciences, Foster City, California, United States of America
| | - Leanne Peiser
- Gilead Sciences, Foster City, California, United States of America
| | - Eduardo Salas
- Gilead Sciences, Foster City, California, United States of America
| | - Hilario Ramos
- Gilead Sciences, Foster City, California, United States of America
| | - Mei Yu
- Gilead Sciences, Foster City, California, United States of America
| | - Guofeng Cheng
- Gilead Sciences, Foster City, California, United States of America
| | - Michel Strubin
- Department of Microbiology and Molecular Medicine, University Medical Center (C.M.U.), Geneva, Switzerland
| | | | | |
Collapse
|
15
|
Lee SH, Park JW, Woo SH, Go DM, Kwon HJ, Jang JJ, Kim DY. Suppression of osteopontin inhibits chemically induced hepatic carcinogenesis by induction of apoptosis in mice. Oncotarget 2016; 7:87219-87231. [PMID: 27888617 PMCID: PMC5349983 DOI: 10.18632/oncotarget.13529] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2016] [Accepted: 11/01/2016] [Indexed: 12/16/2022] Open
Abstract
Previous clinical reports have found elevated osteopontin (OPN) levels in tumor tissues to be indicative of greater malignancy in human hepatocellular carcinoma (HCC). However, the role of OPN on carcinogenesis and its underlying mechanism remain unclear. In the present study, we investigated the oncogenic role of OPN in diethylnitrosamine (DEN)-induced hepatic carcinogenesis in mice. The overall incidence of hepatic tumors at 36 weeks was significantly lower in OPN knockout (KO) mice than in wild-type (WT) mice. Apoptosis was significantly enhanced in OPN KO mice, and was accompanied by the downregulation of epidermal growth factor receptor (EGFR). In the in vitro study, OPN suppression also led to lower mRNA and protein levels of EGFR associated with the downregulation of c-Jun in Hep3B and Huh7 human HCC cells lines, which resulted in increased apoptotic cell death in both cell lines. Moreover, a positive correlation was clearly identified between the expression of OPN and EGFR in human HCC tissues. These data demonstrate that the OPN deficiency reduced the incidence of chemically induced HCC by suppressing EGFR-mediated anti-apoptotic signaling. An important implication of our findings is that OPN positively contributes to hepatic carcinogenesis.
Collapse
Affiliation(s)
- Su-Hyung Lee
- Department of Veterinary Pathology, College of Veterinary Medicine, Seoul National University, Seoul 151-742, South Korea
| | - Jun-Won Park
- Biomolecular Function Research Branch, National Cancer Center, Goyang, Gyeonggi 410-769, South Korea
| | - Sang-Ho Woo
- Department of Veterinary Pathology, College of Veterinary Medicine, Seoul National University, Seoul 151-742, South Korea
| | - Du-Min Go
- Department of Veterinary Pathology, College of Veterinary Medicine, Seoul National University, Seoul 151-742, South Korea
| | - Hyo-Jung Kwon
- Department of Veterinary Pathology, College of Veterinary Medicine, Chungnam National University, Daejeon 305-764, South Korea
| | - Ja-June Jang
- Department of Pathology, College of Medicine, Seoul National University, Seoul 110-799, Korea
| | - Dae-Yong Kim
- Department of Veterinary Pathology, College of Veterinary Medicine, Seoul National University, Seoul 151-742, South Korea
| |
Collapse
|
16
|
Shi L, Peng F, Tao Y, Fan X, Li N. Roles of long noncoding RNAs in hepatocellular carcinoma. Virus Res 2016; 223:131-9. [PMID: 27374059 DOI: 10.1016/j.virusres.2016.06.008] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2016] [Revised: 05/19/2016] [Accepted: 06/15/2016] [Indexed: 12/11/2022]
Abstract
Hepatocellular carcinoma (HCC) is one of the most common cancers worldwide with high prevalence and lethality. Hepatitis B virus (HBV) and Hepatitis C virus (HCV) infection are the major risk factors for HCC. Long noncoding RNAs (lncRNAs) are involved in diverse biological processes, and aberrant lncRNA expression is relevant to many human diseases including HCC. Although many researches on HCC have been reported and lncRNAs roles in carcinogenesis have been highlighted recently, reports on roles of lncRNAs in HBV/HCV-induced HCC are limited. In this review, we concentrate on recent progress regarding the functional roles of lncRNAs in HCC and HBV/HCV-related HCC.
Collapse
Affiliation(s)
- Linxi Shi
- Department of Blood Transfusion, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, Hunan Province 410008, China
| | - Fang Peng
- Key Laboratory of Cancer Proteomics of Chinese Ministry of Health, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, Hunan 410008, China
| | - Yongguang Tao
- Cancer Research Institute, Central South University, 87 Xiangya Road, Changsha, Hunan 410078, China
| | - Xuegong Fan
- Hunan Key Laboratory of Viral Hepatitis,Xiangya Hospital, Central South University, Hunan Province, 87 Xiangya Road, Changsha 410008, China.
| | - Ning Li
- Department of Blood Transfusion, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, Hunan Province 410008, China.
| |
Collapse
|