1
|
Sun J, Wu G, Pastor F, Rahman N, Wang WH, Zhang Z, Merle P, Hui L, Salvetti A, Durantel D, Yang D, Andrisani O. RNA helicase DDX5 enables STAT1 mRNA translation and interferon signalling in hepatitis B virus replicating hepatocytes. Gut 2022; 71:991-1005. [PMID: 34021034 PMCID: PMC8606016 DOI: 10.1136/gutjnl-2020-323126] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/20/2020] [Accepted: 05/09/2021] [Indexed: 12/18/2022]
Abstract
OBJECTIVE RNA helicase DDX5 is downregulated during HBV replication and poor prognosis HBV-related hepatocellular carcinoma (HCC). The objective of this study is to investigate the role of DDX5 in interferon (IFN) signalling. We provide evidence of a novel mechanism involving DDX5 that enables translation of transcription factor STAT1 mediating the IFN response. DESIGN AND RESULTS Molecular, pharmacological and biophysical assays were used together with cellular models of HBV replication, HCC cell lines and liver tumours. We demonstrate that DDX5 regulates STAT1 mRNA translation by resolving a G-quadruplex (rG4) RNA structure, proximal to the 5' end of STAT1 5'UTR. We employed luciferase reporter assays comparing wild type (WT) versus mutant rG4 sequence, rG4-stabilising compounds, CRISPR/Cas9 editing of the STAT1-rG4 sequence and circular dichroism determination of the rG4 structure. STAT1-rG4 edited cell lines were resistant to the effect of rG4-stabilising compounds in response to IFN-α, while HCC cell lines expressing low DDX5 exhibited reduced IFN response. Ribonucleoprotein and electrophoretic mobility assays demonstrated direct and selective binding of RNA helicase-active DDX5 to the WT STAT1-rG4 sequence. Immunohistochemistry of normal liver and liver tumours demonstrated that absence of DDX5 corresponded to absence of STAT1. Significantly, knockdown of DDX5 in HBV infected HepaRG cells reduced the anti-viral effect of IFN-α. CONCLUSION RNA helicase DDX5 resolves a G-quadruplex structure in 5'UTR of STAT1 mRNA, enabling STAT1 translation. We propose that DDX5 is a key regulator of the dynamic range of IFN response during innate immunity and adjuvant IFN-α therapy.
Collapse
Affiliation(s)
- Jiazeng Sun
- Basic Medical Sciences, Purdue University, West Lafayette, Indiana, USA
| | - Guanhui Wu
- Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University, West Lafayette, Indiana, USA
| | - Florentin Pastor
- International Center for Infectiology Research (CIRI), INSERM U1111-CNRS UMR5308, Lyon, France
| | - Naimur Rahman
- Basic Medical Sciences, Purdue University System, West Lafayette, Indiana, USA
| | - Wen-Hung Wang
- Gene Editing Core, Bindley Biosciences Center, Purdue University, West Lafayette, Indiana, USA
| | - Zhengtao Zhang
- Department of Biochemistry and Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Shanghai, China
| | - Philippe Merle
- Service d'Hépatologie, Hôpital de La Croix-Rousse Centre Livet, Lyon, Rhône-Alpes, France
| | - Lijian Hui
- Department of Biochemistry and Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Shanghai, China
| | - Anna Salvetti
- International Center for Infectiology Research (CIRI), INSERM U1111-CNRS UMR5308, Lyon, France
| | - David Durantel
- INSERM U1111-CNRS UMR5308 International Center for Infectiology Research (CIRI), Lyon, France
| | - Danzhou Yang
- Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University, West Lafayette, Indiana, USA
| | - Ourania Andrisani
- Basic Medical Sciences, Purdue University, West Lafayette, Indiana, USA
| |
Collapse
|
2
|
Lu B, Tang Q, Wang Q, Liu X, Peng H, Zhu B, Xie L, Li Z, Wang H, Zheng Z, Wang L, Li B. Recovery Infectious Enterovirus 71 by Bac-to-Bac Expression System in vitro and in vivo. Front Microbiol 2022; 13:825111. [PMID: 35356523 PMCID: PMC8959925 DOI: 10.3389/fmicb.2022.825111] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Accepted: 01/19/2022] [Indexed: 11/13/2022] Open
Abstract
Enterovirus 71 (EV71) is one of the most important etiological agents for hand-foot-mouth disease. Compared with coxsackievirus A16 infection, EV71 infection is often associated with severe central nervous system complications, such as encephalitis, encephalomyelitis, and acute flaccid paralysis in infants and young children. In this study, we constructed a recombinant baculovirus with T7 ribonucleic acid polymerase under the control of a cytomegalovirus promoter and simultaneously engineered the T7 promoter upstream of a full-length EV71 complementary deoxyribonucleic acid. After transduction into mammalian cells, typical cytopathic effects (CPEs) and VP1 signals were detected in cells transfected with recombinant baculovirus. Additionally, viral particles located in the cytoplasm of human rhabdomyosarcoma cells (Rd) and Vero cells were observed by electron microscope, indicating that EV71 was recovered using a Bac-to-Bac expression system in vitro. After four passages, the rescued virus had a growth curve and plaque morphology similar to those of the parental virus. Furthermore, the Vp1 gene and the protein from the mouse brain were detected by reverse transcription polymerase chain reaction and immunohistochemistry after intracerebral injection of purified recombinant baculovirus. Typical CPEs were observed after inoculation of the supernatant from mouse brain to Rd cells, revealing a reconstruction of EV71 in vivo. Thus, we established a new approach to rescue EV71 based on a baculovirus expression system in vitro and in vivo, which may provide a safe and convenient platform for fundamental research and a strategy to rescue viruses that currently lack suitable cell culture and animal models.
Collapse
Affiliation(s)
- Baojing Lu
- The Key Laboratory of Microbiology and Parasitology of Anhui Province, The Key Laboratory of Zoonoses of High Institutions in Anhui, Department of Microbiology and Parasitology, School of Basic Medical Sciences, Anhui Medical University, Hefei, China
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Hefei, China
| | - Qi Tang
- The Key Laboratory of Microbiology and Parasitology of Anhui Province, The Key Laboratory of Zoonoses of High Institutions in Anhui, Department of Microbiology and Parasitology, School of Basic Medical Sciences, Anhui Medical University, Hefei, China
| | - Qianyun Wang
- The Key Laboratory of Microbiology and Parasitology of Anhui Province, The Key Laboratory of Zoonoses of High Institutions in Anhui, Department of Microbiology and Parasitology, School of Basic Medical Sciences, Anhui Medical University, Hefei, China
| | - Xuejuan Liu
- The Key Laboratory of Microbiology and Parasitology of Anhui Province, The Key Laboratory of Zoonoses of High Institutions in Anhui, Department of Microbiology and Parasitology, School of Basic Medical Sciences, Anhui Medical University, Hefei, China
| | - Hui Peng
- The Key Laboratory of Microbiology and Parasitology of Anhui Province, The Key Laboratory of Zoonoses of High Institutions in Anhui, Department of Microbiology and Parasitology, School of Basic Medical Sciences, Anhui Medical University, Hefei, China
| | - Binbin Zhu
- The Key Laboratory of Microbiology and Parasitology of Anhui Province, The Key Laboratory of Zoonoses of High Institutions in Anhui, Department of Microbiology and Parasitology, School of Basic Medical Sciences, Anhui Medical University, Hefei, China
| | - Li Xie
- Department of Tuberculosis Prevention, Shenzhen Center for Chronic Disease Control, Shenzhen, China
| | - Zeng Li
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Hefei, China
| | - Hanzhong Wang
- CAS Key Laboratory of Special Pathogens and Biosafety, Center for Emerging Infectious Diseases, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, China
| | - Zhenhua Zheng
- CAS Key Laboratory of Special Pathogens and Biosafety, Center for Emerging Infectious Diseases, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, China
| | - Linding Wang
- The Key Laboratory of Microbiology and Parasitology of Anhui Province, The Key Laboratory of Zoonoses of High Institutions in Anhui, Department of Microbiology and Parasitology, School of Basic Medical Sciences, Anhui Medical University, Hefei, China
| | - Bao Li
- The Comprehensive Lab, School of Basic Medical Science, Anhui Medical University, Hefei, China
| |
Collapse
|
3
|
Rahman N, Sun J, Li Z, Pattnaik A, Mohallem R, Wang M, Kazemian M, Aryal UK, Andrisani O. The cytoplasmic LSm1-7 and nuclear LSm2-8 complexes exert opposite effects on Hepatitis B virus biosynthesis and interferon responses. Front Immunol 2022; 13:970130. [PMID: 36016928 PMCID: PMC9396650 DOI: 10.3389/fimmu.2022.970130] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Accepted: 07/15/2022] [Indexed: 12/12/2022] Open
Abstract
Despite many studies on host or viral gene expression, how the cellular proteome responds to internal or external cues during the infection process remains unclear. In this study, we used a Hepatitis B Virus (HBV) replication model and performed proteomic analyses to understand how HBV evades innate immunity as a function of cell cycle progression. Specifically, we performed proteomic analyses of HBV-replicating cells in G1/S and G2/M phases, as a function of IFN-α treatment. We identified that the conserved LSm (Like-Sm1-8) proteins were differentially regulated in HBV replicating cells treated with IFN-α. Specifically, in G2/M phase, IFN-α increased protein level of LSm1, the unique subunit of cytoplasmic LSm1-7 complex involved in mRNA decay. By contrast, IFN-α decreased LSm8, the unique subunit of nuclear LSm2-8 complex, a chaperone of U6 spliceosomal RNA, suggesting the cytoplasmic LSm1-7 complex is antiviral, whereas the nuclear LSm2-8 complex is pro-viral. In HBV replication and infection models, siRNA-mediated knockdown of LSm1 increased all viral RNAs. Conversely, LSm8 knockdown reduced viral RNA levels, dependent on N6-adenosine methylation (m6A) of the epsilon stem-loop at the 5' end of pre-Core/pregenomic (preC/pg) RNA. Methylated RNA immunoprecipitation (MeRIP) assays demonstrated reduced viral RNA methylation by LSm8 knockdown, dependent on the 5' m6A modification, suggesting the LSm2-8 complex has a role in mediating this modification. Interestingly, splicing inhibitor Cp028 acting upstream of the LSm2-8 complex suppressed viral RNA levels without reducing the 5' m6A modification. This observation suggests Cp028 has novel antiviral effects, likely potentiating IFN-α-mediated suppression of HBV biosynthesis.
Collapse
Affiliation(s)
- Naimur Rahman
- Department of Basic Medical Sciences, Purdue University, West Lafayette, IN, United States.,Purdue Center for Cancer Research, Purdue University, West Lafayette, IN, United States
| | - Jiazeng Sun
- Department of Basic Medical Sciences, Purdue University, West Lafayette, IN, United States.,Purdue Center for Cancer Research, Purdue University, West Lafayette, IN, United States
| | - Zhili Li
- Department of Basic Medical Sciences, Purdue University, West Lafayette, IN, United States.,Purdue Center for Cancer Research, Purdue University, West Lafayette, IN, United States
| | - Aryamav Pattnaik
- Purdue Center for Cancer Research, Purdue University, West Lafayette, IN, United States.,Department of Biochemistry, Purdue University, West Lafayette, IN, United States
| | - Rodrigo Mohallem
- Purdue Proteomics Facility, Bindley Bioscience Center, Purdue University, West Lafayette, IN, United States.,Department of Comparative Pathobiology, Purdue University, West Lafayette, IN, United States
| | - Mengbo Wang
- Purdue Center for Cancer Research, Purdue University, West Lafayette, IN, United States.,Department of Biochemistry, Purdue University, West Lafayette, IN, United States
| | - Majid Kazemian
- Purdue Center for Cancer Research, Purdue University, West Lafayette, IN, United States.,Department of Biochemistry, Purdue University, West Lafayette, IN, United States.,Department of Computer Science, Purdue University, West Lafayette, IN, United States
| | - Uma K Aryal
- Purdue Proteomics Facility, Bindley Bioscience Center, Purdue University, West Lafayette, IN, United States.,Department of Comparative Pathobiology, Purdue University, West Lafayette, IN, United States
| | - Ourania Andrisani
- Department of Basic Medical Sciences, Purdue University, West Lafayette, IN, United States.,Purdue Center for Cancer Research, Purdue University, West Lafayette, IN, United States
| |
Collapse
|
4
|
Singh P, Kairuz D, Arbuthnot P, Bloom K. Silencing hepatitis B virus covalently closed circular DNA: The potential of an epigenetic therapy approach. World J Gastroenterol 2021; 27:3182-3207. [PMID: 34163105 PMCID: PMC8218364 DOI: 10.3748/wjg.v27.i23.3182] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Revised: 03/23/2021] [Accepted: 05/07/2021] [Indexed: 02/06/2023] Open
Abstract
Global prophylactic vaccination programmes have helped to curb new hepatitis B virus (HBV) infections. However, it is estimated that nearly 300 million people are chronically infected and have a high risk of developing hepatocellular carcinoma. As such, HBV remains a serious health priority and the development of novel curative therapeutics is urgently needed. Chronic HBV infection has been attributed to the persistence of the covalently closed circular DNA (cccDNA) which establishes itself as a minichromosome in the nucleus of hepatocytes. As the viral transcription intermediate, the cccDNA is responsible for producing new virions and perpetuating infection. HBV is dependent on various host factors for cccDNA formation and the minichromosome is amenable to epigenetic modifications. Two HBV proteins, X (HBx) and core (HBc) promote viral replication by modulating the cccDNA epigenome and regulating host cell responses. This includes viral and host gene expression, chromatin remodeling, DNA methylation, the antiviral immune response, apoptosis, and ubiquitination. Elimination of the cccDNA minichromosome would result in a sterilizing cure; however, this may be difficult to achieve. Epigenetic therapies could permanently silence the cccDNA minichromosome and promote a functional cure. This review explores the cccDNA epigenome, how host and viral factors influence transcription, and the recent epigenetic therapies and epigenome engineering approaches that have been described.
Collapse
Affiliation(s)
- Prashika Singh
- Wits/SAMRC Antiviral Gene Therapy Research Unit, School of Pathology, University of the Witwatersrand, Johannesburg 2050, Gauteng, South Africa
| | - Dylan Kairuz
- Wits/SAMRC Antiviral Gene Therapy Research Unit, School of Pathology, University of the Witwatersrand, Johannesburg 2050, Gauteng, South Africa
| | - Patrick Arbuthnot
- Wits/SAMRC Antiviral Gene Therapy Research Unit, School of Pathology, University of the Witwatersrand, Johannesburg 2050, Gauteng, South Africa
| | - Kristie Bloom
- Wits/SAMRC Antiviral Gene Therapy Research Unit, School of Pathology, University of the Witwatersrand, Johannesburg 2050, Gauteng, South Africa
| |
Collapse
|
5
|
Mani SKK, Yan B, Cui Z, Sun J, Utturkar S, Foca A, Fares N, Durantel D, Lanman N, Merle P, Kazemian M, Andrisani O. Restoration of RNA helicase DDX5 suppresses hepatitis B virus (HBV) biosynthesis and Wnt signaling in HBV-related hepatocellular carcinoma. Theranostics 2020; 10:10957-10972. [PMID: 33042264 PMCID: PMC7532671 DOI: 10.7150/thno.49629] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Accepted: 08/11/2020] [Indexed: 02/07/2023] Open
Abstract
Rationale: RNA helicase DDX5 is downregulated during hepatitis B virus (HBV) replication, and poor prognosis HBV-related hepatocellular carcinoma (HCC). The aim of this study is to determine the mechanism and significance of DDX5 downregulation for HBV-driven HCC, and identify biologics to prevent DDX5 downregulation. Methods: Molecular approaches including immunoblotting, qRT-PCR, luciferase transfections, hepatosphere assays, Assay for Transposase-Accessible Chromatin sequencing (ATAC-seq), and RNA-seq were used with cellular models of HBV replication, HBV infection, and HBV-related liver tumors, as well as bioinformatic analyses of liver cancer cells from two independent cohorts. Results: We demonstrate that HBV infection induces expression of the proto-oncogenic miR17~92 and miR106b~25 clusters which target the downregulation of DDX5. Increased expression of these miRNAs is also detected in HBV-driven HCCs exhibiting reduced DDX5 mRNA. Stable DDX5 knockdown (DDX5KD) in HBV replicating hepatocytes increased viral replication, and resulted in hepatosphere formation, drug resistance, Wnt activation, and pluripotency gene expression. ATAC-seq of DDX5KD compared to DDX5 wild-type (WT) cells identified accessible chromatin regions enriched in regulation of Wnt signaling genes. RNA-seq analysis comparing WT versus DDX5KD cells identified enhanced expression of multiple genes involved in Wnt pathway. Additionally, expression of Disheveled, DVL1, a key regulator of Wnt pathway activation, was significantly higher in liver cancer cells with low DDX5 expression, from two independent cohorts. Importantly, inhibitors (antagomirs) to miR17~92 and miR106b~25 restored DDX5 levels, reduced DVL1 expression, and suppressed both Wnt activation and viral replication. Conclusion : DDX5 is a negative regulator of Wnt signaling and hepatocyte reprogramming in HCCs. Restoration of DDX5 levels by miR17~92 / miR106b~25 antagomirs in HBV-infected patients can be explored as both antitumor and antiviral strategy.
Collapse
|
6
|
In Vitro Systems for Studying Different Genotypes/Sub-Genotypes of Hepatitis B Virus: Strengths and Limitations. Viruses 2020; 12:v12030353. [PMID: 32210021 PMCID: PMC7150782 DOI: 10.3390/v12030353] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2020] [Revised: 03/04/2020] [Accepted: 03/06/2020] [Indexed: 12/11/2022] Open
Abstract
Hepatitis B virus (HBV) infects the liver resulting in end stage liver disease, cirrhosis, and hepatocellular carcinoma. Despite an effective vaccine, HBV poses a serious health problem globally, accounting for 257 million chronic carriers. Unique features of HBV, including its narrow virus-host range and its hepatocyte tropism, have led to major challenges in the development of suitable in vivo and in vitro model systems to recapitulate the HBV replication cycle and to test various antiviral strategies. Moreover, HBV is classified into at least nine genotypes and 35 sub-genotypes with distinct geographical distributions and prevalence, which have different natural histories of infection, clinical manifestation, and response to current antiviral agents. Here, we review various in vitro systems used to study the molecular biology of the different (sub)genotypes of HBV and their response to antiviral agents, and we discuss their strengths and limitations. Despite the advances made, no system is ideal for pan-genotypic HBV research or drug development and therefore further improvement is required. It is necessary to establish a centralized repository of HBV-related generated materials, which are readily accessible to HBV researchers, with international collaboration toward advancement and development of in vitro model systems for testing new HBV antivirals to ensure their pan-genotypic and/or customized activity.
Collapse
|
7
|
Ruan J, Ping CY, Sun S, Cheng X, Han PY, Zhang YG, Sun DX. Construction of a replication-competent hepatitis B virus vector carrying secreted luciferase transgene and establishment of new hepatitis B virus replication and expression cell lines. World J Gastroenterol 2019; 25:5961-5972. [PMID: 31660033 PMCID: PMC6815792 DOI: 10.3748/wjg.v25.i39.5961] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/19/2019] [Revised: 08/08/2019] [Accepted: 09/13/2019] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Previously, we have successfully constructed replication-competent hepatitis B virus (HBV) vectors by uncoupling the P open reading frame (ORF) from the preC/C ORF to carefully design the transgene insertion site to overcome the compact organization of the HBV genome and maintain HBV replication competence. Consequently, the replication-competent HBV vectors carrying foreign genes, including pCH-BsdR, carrying blasticidin resistance gene (399 bp), and pCH-hrGFP, carrying humanized renilla green fluorescent protein gene (720 bp), were successfully obtained. However, the replication efficiency of the former is higher but it is tedious to use, while that of the latter is poor and cannot be quantified. Hence, we need to search for a new reporter gene that is convenient and quantifiable for further research.
AIM To establish a helpful tool for intracellular HBV replication and anti-viral drugs screening studies.
METHODS We utilized the replication-competent HBV viral vectors constructed by our laboratory, combined with the secreted luciferase reporter gene, to construct replication-competent HBV vectors expressing the reporter gene secretory Nanoluc Luciferase (SecNluc). HepG2.TA2-7 cells were transfected with this vector to obtain cell lines with stably secreted HBV particles carrying secNluc reporter gene.
RESULTS The replication-competent HBV vector carrying the SecNluc reporter gene pCH-sNLuc could produce all major viral RNAs and a full set of envelope proteins and achieve high-level secreted luciferase expression. HBV replication intermediates could be produced from this vector. Via transfection with pTRE-sNLuc and selection by hygromycin, we obtained isolated cell clones, named HBV-NLuc-35 cells, which could secrete secNLuc recombinant viruses, and were sensitive to existing anti-HBV drugs. Using differentiated HepaRG cells, it was verified that recombinant HBV possessed infectivity.
CONCLUSION Our research demonstrated that a replication-competent HBV vector carrying a secreted luciferase transgene possesses replication and expression ability, and the established HBV replication and expression cell lines could stably secrete viral particles carrying secNluc reporter gene. More importantly, the cell line and the secreted recombinant viral particles could be used to trace HBV replication or infection.
Collapse
Affiliation(s)
- Jie Ruan
- The Liver Disease Center of Chinese People’s Liberation Army, the 980th Hospital of Chinese People’s Liberation Army Joint Logistics Support Force, Shijiazhuang 050082, Hebei Province, China
- Department of Infection and Liver Disease, Shannxi University of Chinese Medicine Affiliated Hospital, Xianyang 712000, Shannxi Province, China
| | - Cai-Yan Ping
- The Liver Disease Center of Chinese People’s Liberation Army, the 980th Hospital of Chinese People’s Liberation Army Joint Logistics Support Force, Shijiazhuang 050082, Hebei Province, China
| | - Shuo Sun
- The Liver Disease Center of Chinese People’s Liberation Army, the 980th Hospital of Chinese People’s Liberation Army Joint Logistics Support Force, Shijiazhuang 050082, Hebei Province, China
| | - Xin Cheng
- The Liver Disease Center of Chinese People’s Liberation Army, the 980th Hospital of Chinese People’s Liberation Army Joint Logistics Support Force, Shijiazhuang 050082, Hebei Province, China
| | - Peng-Yu Han
- The Liver Disease Center of Chinese People’s Liberation Army, the 980th Hospital of Chinese People’s Liberation Army Joint Logistics Support Force, Shijiazhuang 050082, Hebei Province, China
| | - Yin-Ge Zhang
- The Liver Disease Center of Chinese People’s Liberation Army, the 980th Hospital of Chinese People’s Liberation Army Joint Logistics Support Force, Shijiazhuang 050082, Hebei Province, China
| | - Dian-Xing Sun
- The Liver Disease Center of Chinese People’s Liberation Army, the 980th Hospital of Chinese People’s Liberation Army Joint Logistics Support Force, Shijiazhuang 050082, Hebei Province, China
| |
Collapse
|
8
|
Fu S, Li N, Zhou PC, Huang Y, Zhou RR, Fan XG. Detection of HBV DNA and antigens in HBsAg-positive patients with primary hepatocellular carcinoma. Clin Res Hepatol Gastroenterol 2017; 41:415-423. [PMID: 28286056 DOI: 10.1016/j.clinre.2017.01.009] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/14/2016] [Revised: 12/16/2016] [Accepted: 01/23/2017] [Indexed: 02/08/2023]
Abstract
BACKGROUND Hepatitis B virus (HBV) markers include HBV deoxyribonucleic acid (DNA) and HBV antigens. The former involves HBV covalently closed circular DNA (cccDNA) as well as total HBV DNA, whereas the latter involves HBsAg, HBcAg, and HBx. METHODS Samples of tumor and adjacent non-tumor liver tissue were collected from 28 HBV-associated HCC patients. Intrahepatic total HBV DNA and cccDNA were measured using the real-time PCR Taqman assay. HBV antigens in hepatocytes were detected using immunohistochemical staining. Intrahepatic levels of total HBV DNA or cccDNA in HCC patients with different intrahepatic HBV antigen expression patterns were compared, and the correlation between serum HBV DNA and intrahepatic HBV DNA was analyzed. RESULTS No significant differences in intrahepatic cccDNA levels were observed between tumor and non-tumor liver tissue (median -3.00 vs. -2.30 log copies/cell, P=0.298). However, the tumor tissue had significantly higher levels of total HBV DNA (median -0.60 vs. -1.24 log copies/cell, P=0.045) but significantly lower proportion of intrahepatic HBV DNA in the form of cccDNA (median 0.25% vs. 4%, P=0.023) than the corresponding values in the non-tumor tissue. Also, HBV antigen levels were lower in the tumor tissue than in the non-tumor tissue. Analysis of the correlation between serum HBV DNA and intrahepatic HBV DNA indicated that the viral status in the tumor tissue was more complicated in HBV-HCC patients-the detected serum HBV DNA failed to accurately reflect intrahepatic viral load. CONCLUSION HBV DNA may play an important role in hepatocarcinogenesis, and cccDNA was not the predominant form of HBV DNA in the tumor tissue.
Collapse
Affiliation(s)
- Sha Fu
- Department of Infectious Diseases, Key Laboratory of Viral Hepatitis of Hunan Province, Xiangya Hospital, Central South University, PO Box 410008, Changsha, China
| | - Ning Li
- Department of Blood Transfusion, Xiangya Hospital, Central South University, Changsha, China
| | - Peng-Cheng Zhou
- Department of Infectious Diseases, Key Laboratory of Viral Hepatitis of Hunan Province, Xiangya Hospital, Central South University, PO Box 410008, Changsha, China
| | - Yan Huang
- Department of Infectious Diseases, Key Laboratory of Viral Hepatitis of Hunan Province, Xiangya Hospital, Central South University, PO Box 410008, Changsha, China
| | - Rong-Rong Zhou
- Department of Infectious Diseases, Key Laboratory of Viral Hepatitis of Hunan Province, Xiangya Hospital, Central South University, PO Box 410008, Changsha, China.
| | - Xue-Gong Fan
- Department of Infectious Diseases, Key Laboratory of Viral Hepatitis of Hunan Province, Xiangya Hospital, Central South University, PO Box 410008, Changsha, China.
| |
Collapse
|
9
|
Alfaiate D, Lucifora J, Abeywickrama-Samarakoon N, Michelet M, Testoni B, Cortay JC, Sureau C, Zoulim F, Dény P, Durantel D. HDV RNA replication is associated with HBV repression and interferon-stimulated genes induction in super-infected hepatocytes. Antiviral Res 2016; 136:19-31. [PMID: 27771387 DOI: 10.1016/j.antiviral.2016.10.006] [Citation(s) in RCA: 70] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2016] [Accepted: 10/18/2016] [Indexed: 12/14/2022]
Abstract
Hepatitis D virus (HDV) super-infection of Hepatitis B virus (HBV)-infected patients is the most aggressive form of viral hepatitis. HDV infection is not susceptible to direct anti-HBV drugs, and only suboptimal antiviral responses are obtained with interferon (IFN)-alpha-based therapy. To get insights on HDV replication and interplay with HBV in physiologically relevant hepatocytes, differentiated HepaRG (dHepaRG) cells, previously infected or not with HBV, were infected with HDV, and viral markers were extensively analyzed. Innate and IFN responses to HDV were monitored by measuring pro-inflammatory and interferon-stimulated gene (ISG) expression. Both mono- and super-infected dHepaRG cells supported a strong HDV intracellular replication, which was accompanied by a strong secretion of infectious HDV virions only in the super-infection setting and despite the low number of co-infected cells. Upon HDV super-infection, HBV replication markers including HBeAg, total HBV-DNA and pregenomic RNA were significantly decreased, confirming the interference of HDV on HBV. Yet, no decrease of circular covalently closed HBV DNA (cccDNA) and HBsAg levels was evidenced. At the peak of HDV-RNA accumulation and onset of interference on HBV replication, a strong type-I IFN response was observed, with interferon stimulated genes, RSAD2 (Viperin) and IFI78 (MxA) being highly induced. We established a cellular model to characterize in more detail the direct interference of HBV and HDV, and the indirect interplay between the two viruses via innate immune responses. This model will be instrumental to assess molecular and immunological mechanisms of this viral interference.
Collapse
Affiliation(s)
- Dulce Alfaiate
- INSERM U1052, CNRS UMR-5286, Cancer Research Center of Lyon (CRCL), 69008 Lyon, France; University of Lyon, Université Claude-Bernard (UCBL), 69008 Lyon, France
| | - Julie Lucifora
- INSERM U1052, CNRS UMR-5286, Cancer Research Center of Lyon (CRCL), 69008 Lyon, France; University of Lyon, Université Claude-Bernard (UCBL), 69008 Lyon, France.
| | - Natali Abeywickrama-Samarakoon
- INSERM U1052, CNRS UMR-5286, Cancer Research Center of Lyon (CRCL), 69008 Lyon, France; University of Lyon, Université Claude-Bernard (UCBL), 69008 Lyon, France
| | - Maud Michelet
- INSERM U1052, CNRS UMR-5286, Cancer Research Center of Lyon (CRCL), 69008 Lyon, France; University of Lyon, Université Claude-Bernard (UCBL), 69008 Lyon, France
| | - Barbara Testoni
- INSERM U1052, CNRS UMR-5286, Cancer Research Center of Lyon (CRCL), 69008 Lyon, France; University of Lyon, Université Claude-Bernard (UCBL), 69008 Lyon, France
| | - Jean-Claude Cortay
- INSERM U1052, CNRS UMR-5286, Cancer Research Center of Lyon (CRCL), 69008 Lyon, France; University of Lyon, Université Claude-Bernard (UCBL), 69008 Lyon, France
| | - Camille Sureau
- Institut National de Transfusion Sanguine, Laboratoire de Virologie Moléculaire, 75015 Paris, France
| | - Fabien Zoulim
- INSERM U1052, CNRS UMR-5286, Cancer Research Center of Lyon (CRCL), 69008 Lyon, France; University of Lyon, Université Claude-Bernard (UCBL), 69008 Lyon, France; Laboratoire d'excellence (LabEx), DEVweCAN, 69008 Lyon, France; Hospices Civils de Lyon (HCL), 69002 Lyon, France
| | - Paul Dény
- INSERM U1052, CNRS UMR-5286, Cancer Research Center of Lyon (CRCL), 69008 Lyon, France; Université Paris 13/SPC, UFR SMBH, Laboratoire de Bactériologie, Virologie - Hygiène, GHU Paris Seine Saint Denis, Assistance Publique - Hôpitaux de Paris, Bobigny, France.
| | - David Durantel
- INSERM U1052, CNRS UMR-5286, Cancer Research Center of Lyon (CRCL), 69008 Lyon, France; University of Lyon, Université Claude-Bernard (UCBL), 69008 Lyon, France; Laboratoire d'excellence (LabEx), DEVweCAN, 69008 Lyon, France.
| |
Collapse
|
10
|
The recombined cccDNA produced using minicircle technology mimicked HBV genome in structure and function closely. Sci Rep 2016; 6:25552. [PMID: 27174254 PMCID: PMC4865889 DOI: 10.1038/srep25552] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2016] [Accepted: 04/18/2016] [Indexed: 12/17/2022] Open
Abstract
HBV covalently closed circular DNA (cccDNA) is drug-resistant and responsible for viral persistence. To facilitate the development of anti-cccDNA drugs, we developed a minicircle DNA vector (MC)-based technology to produce large quantity of recombined cccDNA (rcccDNA) resembling closely to its wild-type counterpart both in structure and function. The rcccDNA differed to the wild-type cccDNA (wtcccDNA) only in that it carried an extra 36-bp DNA recombinant product attR upstream of the preC/C gene. Using a procedure similar to standard plasmid production, milligrams of rcccDNA can be generated in common laboratories conveniently. The rcccDNA demonstrated many essential biological features of wtcccDNA, including: (1) undergoing nucleation upon nucleus entry; (2) serving as template for production of all HBV RNAs and proteins; (3) deriving virions capable of infecting tree shrew, and subsequently producing viral mRNAs, proteins, rcccDNA and infectious virions. As an example to develop anti-cccDNA drugs, we used the Crispr/Cas9 system to provide clear-cut evidence that rcccDNA was cleaved by this DNA editing tool in vitro. In summary, we have developed a convenient technology to produce large quantity of rcccDNA as a surrogate of wtcccDNA for investigating HBV biology and developing treatment to eradicate this most wide-spreading virus.
Collapse
|
11
|
Experimental in vitro and in vivo models for the study of human hepatitis B virus infection. J Hepatol 2016; 64:S17-S31. [PMID: 27084033 DOI: 10.1016/j.jhep.2016.02.012] [Citation(s) in RCA: 101] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/07/2015] [Revised: 01/29/2016] [Accepted: 02/10/2016] [Indexed: 12/13/2022]
Abstract
Chronic infection with the hepatitis B virus (HBV) affects an estimate of 240 million people worldwide despite the availability of a preventive vaccine. Medication to repress viral replication is available but a cure is rarely achieved. The narrow species and tissue tropism of the virus and the lack of reliable in vitro models and laboratory animals susceptible to HBV infection, have limited research progress in the past. As a result, several aspects of the HBV life cycle as well as the network of virus host interactions occurring during the infection are not yet understood. Only recently, the identification of the functional cellular receptor enabling HBV entry has opened new possibilities to establish innovative infection systems. Regarding the in vivo models of HBV infection, the classical reference was the chimpanzee. However, because of the strongly restricted use of great apes for HBV research, major efforts have focused on the development of mouse models of HBV replication and infection such as the generation of humanized mice. This review summarizes the animal and cell culture based models currently available for the study of HBV biology. We will discuss the benefits and caveats of each model and present a selection of the most important findings that have been retrieved from the respective systems.
Collapse
|
12
|
Witt-Kehati D, Bitton Alaluf M, Shlomai A. Advances and Challenges in Studying Hepatitis B Virus In Vitro. Viruses 2016; 8:v8010021. [PMID: 26784218 PMCID: PMC4728581 DOI: 10.3390/v8010021] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2015] [Revised: 11/30/2015] [Accepted: 01/06/2016] [Indexed: 12/17/2022] Open
Abstract
Hepatitis B virus (HBV) is a small DNA virus that infects the liver. Current anti-HBV drugs efficiently suppress viral replication but do not eradicate the virus due to the persistence of its episomal DNA. Efforts to develop reliable in vitro systems to model HBV infection, an imperative tool for studying HBV biology and its interactions with the host, have been hampered by major limitations at the level of the virus, the host and infection readouts. This review summarizes major milestones in the development of in vitro systems to study HBV. Recent advances in our understanding of HBV biology, such as the discovery of the bile-acid pump sodium-taurocholate cotransporting polypeptide (NTCP) as a receptor for HBV, enabled the establishment of NTCP expressing hepatoma cell lines permissive for HBV infection. Furthermore, advanced tissue engineering techniques facilitate now the establishment of HBV infection systems based on primary human hepatocytes that maintain their phenotype and permissiveness for infection over time. The ability to differentiate inducible pluripotent stem cells into hepatocyte-like cells opens the door for studying HBV in a more isogenic background, as well. Thus, the recent advances in in vitro models for HBV infection holds promise for a better understanding of virus-host interactions and for future development of more definitive anti-viral drugs.
Collapse
Affiliation(s)
- Dvora Witt-Kehati
- The Liver Institute, Rabin Medical Center Beilinson Hospital, Petah-Tikva, Israel.
- Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel.
| | - Maya Bitton Alaluf
- Department of Medicine D, Rabin Medical Center Beilinson Hospital, Petah-Tikva, Israel.
| | - Amir Shlomai
- The Liver Institute, Rabin Medical Center Beilinson Hospital, Petah-Tikva, Israel.
- Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel.
- Department of Medicine D, Rabin Medical Center Beilinson Hospital, Petah-Tikva, Israel.
| |
Collapse
|
13
|
Cheng X, Guan W, Sun S, Li B, Li H, Kang F, Kang J, Yang D, Nassal M, Sun D. Stable Human Hepatoma Cell Lines for Efficient Regulated Expression of Nucleoside/Nucleotide Analog Resistant and Vaccine Escape Hepatitis B Virus Variants and Woolly Monkey Hepatitis B Virus. PLoS One 2015; 10:e0145746. [PMID: 26699621 PMCID: PMC4689378 DOI: 10.1371/journal.pone.0145746] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2015] [Accepted: 12/08/2015] [Indexed: 12/17/2022] Open
Abstract
Hepatitis B virus (HBV) causes acute and chronic hepatitis B (CHB). Due to its error-prone replication via reverse transcription, HBV can rapidly evolve variants that escape vaccination and/or become resistant to CHB treatment with nucleoside/nucleotide analogs (NAs). This is particularly problematic for the first generation NAs lamivudine and adefovir. Though now superseded by more potent NAs, both are still widely used. Furthermore, resistance against the older NAs can contribute to cross-resistance against more advanced NAs. For lack of feasible HBV infection systems, the biology of such variants is not well understood. From the recent discovery of Na+-taurocholate cotransporting polypeptide (NTCP) as an HBV receptor new in vitro infection systems are emerging, yet access to the required large amounts of virions, in particular variants, remains a limiting factor. Stably HBV producing cell lines address both issues by allowing to study intracellular viral replication and as a permanent source of defined virions. Accordingly, we generated a panel of new tetracycline regulated TetOFF HepG2 hepatoma cell lines which produce six lamivudine and adefovir resistance-associated and two vaccine escape variants of HBV as well as the model virus woolly monkey HBV (WMHBV). The cell line-borne viruses reproduced the expected NA resistance profiles and all were equally sensitive against a non-NA drug. The new cell lines should be valuable to investigate under standardized conditions HBV resistance and cross-resistance. With titers of secreted virions reaching >3x107 viral genome equivalents per ml they should also facilitate exploitation of the new in vitro infection systems.
Collapse
Affiliation(s)
- Xin Cheng
- The Liver Disease Diagnosis and Treatment Center of PLA, Bethune International Peace Hospital, Shijiazhuang, PR China
| | - Weiwei Guan
- The Liver Disease Diagnosis and Treatment Center of PLA, Bethune International Peace Hospital, Shijiazhuang, PR China
| | - Shuo Sun
- The Liver Disease Diagnosis and Treatment Center of PLA, Bethune International Peace Hospital, Shijiazhuang, PR China
- Troop 66220 of PLA, Xingtai of Hebei Province, PR China
| | - Baosheng Li
- The Liver Disease Diagnosis and Treatment Center of PLA, Bethune International Peace Hospital, Shijiazhuang, PR China
| | - Haijun Li
- The Liver Disease Diagnosis and Treatment Center of PLA, Bethune International Peace Hospital, Shijiazhuang, PR China
| | - Fubiao Kang
- The Liver Disease Diagnosis and Treatment Center of PLA, Bethune International Peace Hospital, Shijiazhuang, PR China
| | - Jiwen Kang
- The Liver Disease Diagnosis and Treatment Center of PLA, Bethune International Peace Hospital, Shijiazhuang, PR China
| | - Dongliang Yang
- Department of Infectious Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, PR China
| | - Michael Nassal
- Department of Internal Medicine II / Molecular Biology, University Hospital Freiburg, Freiburg, Germany
- * E-mail: (MN); (SD)
| | - Dianxing Sun
- The Liver Disease Diagnosis and Treatment Center of PLA, Bethune International Peace Hospital, Shijiazhuang, PR China
- * E-mail: (MN); (SD)
| |
Collapse
|
14
|
Luangsay S, Gruffaz M, Isorce N, Testoni B, Michelet M, Faure-Dupuy S, Maadadi S, Ait-Goughoulte M, Parent R, Rivoire M, Javanbakht H, Lucifora J, Durantel D, Zoulim F. Early inhibition of hepatocyte innate responses by hepatitis B virus. J Hepatol 2015. [PMID: 26216533 DOI: 10.1016/j.jhep.2015.07.014] [Citation(s) in RCA: 106] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
BACKGROUND & AIMS The outcome of hepatitis B virus (HBV) infection may be influenced by early interactions between the virus and hepatocyte innate immune responses. To date, the study of such interactions during the very early step of infection has not been adequately investigated. METHODS We used the HepaRG cell line, as well as primary human hepatocytes to analyze, within 24h of exposure to HBV, either delivered by a physiologic route or baculovirus vector (Bac-HBV), the early modulation of the expression of selected antiviral/pro-inflammatory cytokines and interferon stimulated genes. Experiments were also performed in the presence or absence of innate receptor agonists to investigate early HBV-induced blockade of innate responses. RESULTS We show that hepatocytes themselves could detect HBV, and express innate genes when exposed to either HBV virions or Bac-HBV. Whereas Bac-HBV triggered a strong antiviral cytokine secretion followed by the clearance of replicative intermediates, a physiologic HBV exposure led to an abortive response. The early inhibition of innate response by HBV was mainly evidenced on Toll-like receptor 3 and RIG-I/MDA5 signaling pathways upon engagement with exogenous agonist, leading to a decreased expression of several pro-inflammatory and antiviral cytokine genes. Finally, we demonstrate that this early inhibition of dsRNA-mediated response is due to factor(s) present in the HBV inoculum, but not being HBsAg or HBeAg themselves, and does not require de novo viral protein synthesis and replication. CONCLUSIONS Our data provide strong evidence that HBV viral particles themselves can readily inhibit host innate immune responses upon virion/cell interactions, and may explain, at least partially, the "stealthy" character of HBV.
Collapse
Affiliation(s)
- Souphalone Luangsay
- INSERM U1052, Cancer Research Center of Lyon (CRCL), Lyon 69008, France; University of Lyon, UMR_S1052, UCBL, 69008 Lyon, France
| | - Marion Gruffaz
- INSERM U1052, Cancer Research Center of Lyon (CRCL), Lyon 69008, France; University of Lyon, UMR_S1052, UCBL, 69008 Lyon, France
| | - Nathalie Isorce
- INSERM U1052, Cancer Research Center of Lyon (CRCL), Lyon 69008, France; University of Lyon, UMR_S1052, UCBL, 69008 Lyon, France
| | - Barbara Testoni
- INSERM U1052, Cancer Research Center of Lyon (CRCL), Lyon 69008, France; University of Lyon, UMR_S1052, UCBL, 69008 Lyon, France
| | - Maud Michelet
- INSERM U1052, Cancer Research Center of Lyon (CRCL), Lyon 69008, France; University of Lyon, UMR_S1052, UCBL, 69008 Lyon, France
| | - Suzanne Faure-Dupuy
- INSERM U1052, Cancer Research Center of Lyon (CRCL), Lyon 69008, France; University of Lyon, UMR_S1052, UCBL, 69008 Lyon, France
| | - Sarah Maadadi
- INSERM U1052, Cancer Research Center of Lyon (CRCL), Lyon 69008, France; University of Lyon, UMR_S1052, UCBL, 69008 Lyon, France
| | - Malika Ait-Goughoulte
- INSERM U1052, Cancer Research Center of Lyon (CRCL), Lyon 69008, France; University of Lyon, UMR_S1052, UCBL, 69008 Lyon, France
| | - Romain Parent
- INSERM U1052, Cancer Research Center of Lyon (CRCL), Lyon 69008, France; University of Lyon, UMR_S1052, UCBL, 69008 Lyon, France
| | - Michel Rivoire
- Centre Léon Bérard (CLB), Lyon 69008, France; INSERM U1032, 69003 Lyon, France
| | - Hassan Javanbakht
- Hoffmann-La Roche Ltd, Roche Pharmaceutical Research and Early Development, 4070 Basel, Switzerland
| | - Julie Lucifora
- INSERM U1052, Cancer Research Center of Lyon (CRCL), Lyon 69008, France; University of Lyon, UMR_S1052, UCBL, 69008 Lyon, France
| | - David Durantel
- INSERM U1052, Cancer Research Center of Lyon (CRCL), Lyon 69008, France; University of Lyon, UMR_S1052, UCBL, 69008 Lyon, France.
| | - Fabien Zoulim
- INSERM U1052, Cancer Research Center of Lyon (CRCL), Lyon 69008, France; University of Lyon, UMR_S1052, UCBL, 69008 Lyon, France; Hospices Civils de Lyon (HCL), 69002 Lyon, France; Institut Universitaire de France (IUF), 75005 Paris, France.
| |
Collapse
|
15
|
Hernández S, Jiménez G, Alarcón V, Prieto C, Muñoz F, Riquelme C, Venegas M, Brahm J, Loyola A, Villanueva RA. Replication of a chronic hepatitis B virus genotype F1b construct. Arch Virol 2015; 161:583-94. [PMID: 26620585 DOI: 10.1007/s00705-015-2702-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2015] [Accepted: 11/23/2015] [Indexed: 12/18/2022]
Abstract
Genotype F is one of the less-studied genotypes of human hepatitis B virus, although it is widely distributed in regions of Central and South American. Our previous studies have shown that HBV genotype F is prevalent in Chile, and phylogenetic analysis of its full-length sequence amplified from the sera of chronically infected patients identified it as HBV subgenotype F1b. We have previously reported the full-length sequence of a HBV molecular clone obtained from a patient chronically infected with genotype F1b. In this report, we established a system to study HBV replication based on hepatoma cell lines transfected with full-length monomers of the HBV genome. Culture supernatants were analyzed after transfection and found to contain both HBsAg and HBeAg viral antigens. Consistently, fractionated cell extracts revealed the presence of viral replication, with both cytoplasmic and nuclear DNA intermediates. Analysis of HBV-transfected cells by indirect immunofluorescence or immunoelectron microscopy revealed the expression of viral antigens and cytoplasmic viral particles, respectively. To test the functionality of the ongoing viral replication further at the level of chromatinized cccDNA, transfected cells were treated with a histone deacetylase inhibitor, and this resulted in increased viral replication. This correlated with changes posttranslational modifications of histones at viral promoters. Thus, the development of this viral replication system for HBV genotype F will facilitate studies on the regulation of viral replication and the identification of new antiviral drugs.
Collapse
Affiliation(s)
- Sergio Hernández
- Laboratorio de Virus Hepatitis, Facultad de Ciencias Biológicas, Universidad Andrés Bello, Avda República 217, 2do piso, 8370146, Santiago, Chile
| | - Gustavo Jiménez
- Laboratorio de Virus Hepatitis, Facultad de Ciencias Biológicas, Universidad Andrés Bello, Avda República 217, 2do piso, 8370146, Santiago, Chile
| | - Valentina Alarcón
- Laboratorio de Epigenética y Cromatina, Fundación Ciencia and Vida, 7780272, Santiago, Chile
| | - Cristian Prieto
- Laboratorio de Virus Hepatitis, Facultad de Ciencias Biológicas, Universidad Andrés Bello, Avda República 217, 2do piso, 8370146, Santiago, Chile
| | - Francisca Muñoz
- Laboratorio de Epigenética y Cromatina, Fundación Ciencia and Vida, 7780272, Santiago, Chile
| | - Constanza Riquelme
- Laboratorio de Virus Hepatitis, Facultad de Ciencias Biológicas, Universidad Andrés Bello, Avda República 217, 2do piso, 8370146, Santiago, Chile
| | - Mauricio Venegas
- Sección de Gastroenterología, Hospital Clínico Universidad de Chile, 8380456, Santiago, Chile
| | - Javier Brahm
- Sección de Gastroenterología, Hospital Clínico Universidad de Chile, 8380456, Santiago, Chile
| | - Alejandra Loyola
- Laboratorio de Epigenética y Cromatina, Fundación Ciencia and Vida, 7780272, Santiago, Chile.,Universidad San Sebastián, 7510157, Santiago, Chile
| | - Rodrigo A Villanueva
- Laboratorio de Virus Hepatitis, Facultad de Ciencias Biológicas, Universidad Andrés Bello, Avda República 217, 2do piso, 8370146, Santiago, Chile.
| |
Collapse
|
16
|
Glebe D, Geipel A. Selected phenotypic assays used to evaluate antiviral resistance and viral fitness of hepatitis B virus and its variants. Intervirology 2014; 57:225-31. [PMID: 25034492 DOI: 10.1159/000360950] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Currently available antiviral therapies specifically target viral replication by blocking reverse transcription with orally given nucleos(t)ide analogues and are able to specifically suppress viral replication. The unique replication strategy of hepatitis B virus (HBV), however, allows long-term persistence of the viral genome within infected hepatocytes in spite of successful therapy. Thus, antiviral therapy needs to be continued for years. Therapy can result either in the emergence and selection of antiviral-resistant variants or the relapse of viral replication after the termination of antiviral therapy. Resistance is a major problem for 4 of the 5 approved HBV nucleos(t)ide analogues, but it is not the only reason for therapy failure. An accurate phenotypic in vitro assay for resistance allows the identification of a viral variant selected in vivo during antiviral therapy and helps to find therapeutic alternatives. Furthermore, these assays can be used to measure viral fitness and pathogenicity in vitro. With the help of these assays, the prediction of emerging viral variants with drug resistance or increased pathogenic potential can be realized. Phenotypic resistance tests for HBV are not trivial because the virus cannot be readily grown in cell culture. This review focuses on currently available phenotypic assays to evaluate antiviral resistance of HBV and fitness of viral variants in general.
Collapse
Affiliation(s)
- Dieter Glebe
- Institute of Medical Virology, Justus Liebig University Giessen, National Reference Center for Hepatitis B and D Viruses, German Center for Infection Research (DZIF), Biomedical Research Center Seltersberg, Giessen, Germany
| | | |
Collapse
|
17
|
Salipalli S, Singh PK, Borlak J. Recent advances in live cell imaging of hepatoma cells. BMC Cell Biol 2014; 15:26. [PMID: 25005127 PMCID: PMC4108253 DOI: 10.1186/1471-2121-15-26] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2013] [Accepted: 06/25/2014] [Indexed: 11/10/2022] Open
Abstract
Live cell imaging enables the study of dynamic processes of living cells in real time by use of suitable reporter proteins and the staining of specific cellular structures and/or organelles. With the availability of advanced optical devices and improved cell culture protocols it has become a rapidly growing research methodology. The success of this technique relies mainly on the selection of suitable reporter proteins, construction of recombinant plasmids possessing cell type specific promoters as well as reliable methods of gene transfer. This review aims to provide an overview of the recent developments in the field of marker proteins (bioluminescence and fluorescent) and methodologies (fluorescent resonance energy transfer, fluorescent recovery after photobleaching and proximity ligation assay) employed as to achieve an improved imaging of biological processes in hepatoma cells. Moreover, different expression systems of marker proteins and the modes of gene transfer are discussed with emphasis on the study of lipid droplet formation in hepatocytes as an example.
Collapse
Affiliation(s)
| | | | - Jürgen Borlak
- Centre for Pharmacology and Toxicology, Hannover Medical School, Carl-Neuberg-Str, 1, 30625 Hannover, Germany.
| |
Collapse
|
18
|
Hu JL, Cui J, Guo JJ, Zhang WL, Cai XF, Yuan ZW, Li QL, Deng XY, Zeng AZ, Hu Y, Tang N, Huang AL. Phenotypic assay of a hepatitis B virus strain carrying an rtS246T variant using a new strategy. J Med Virol 2011; 84:34-43. [PMID: 22052677 DOI: 10.1002/jmv.22260] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/21/2011] [Indexed: 12/13/2022]
Abstract
Phenotypic assays of hepatitis B virus (HBV) play an important role in research related to the problem of drug resistance that emerges during long-term nucleot(s)ide therapy in patients with chronic hepatitis B. Most of the phenotypic assay systems that are available currently rely on the transfection of recombinant replication-competent HBV DNA into hepatoma cell lines. Cloning clinical HBV isolates using conventional digestion-and-ligation techniques to generate replication-competent recombinants can be very difficult because of the sequence heterogeneity and unique structure of the HBV genome. In this study, a new strategy for constructing an HBV 1.1× recombinant was developed. The core of this strategy is the "fragment substitution reaction" (FSR). FSR allows PCR fragments to be cloned without digestion or ligation, providing a new tool for cloning fragments or genomes amplified from serum HBV DNA, and therefore making the assay of HBV phenotypes more convenient. Using this strategy, a phenotypic assay was performed on an HBV strain carrying an rtS246T variant isolated from a patient with chronic hepatitis B that was only responsive partially to entecavir therapy. The results indicated that this strain is sensitive to entecavir in vitro.
Collapse
Affiliation(s)
- Jie-li Hu
- Institute for Viral Hepatitis, Key Laboratory of Molecular Biology on Infectious Diseases, Ministry of Education, Chongqing Medical University, Chongqing, China
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
19
|
Lucifora J, Arzberger S, Durantel D, Belloni L, Strubin M, Levrero M, Zoulim F, Hantz O, Protzer U. Hepatitis B virus X protein is essential to initiate and maintain virus replication after infection. J Hepatol 2011; 55:996-1003. [PMID: 21376091 DOI: 10.1016/j.jhep.2011.02.015] [Citation(s) in RCA: 348] [Impact Index Per Article: 24.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/08/2010] [Revised: 02/01/2011] [Accepted: 02/03/2011] [Indexed: 02/07/2023]
Abstract
BACKGROUND & AIMS The molecular biology of hepatitis B virus (HBV) has been extensively studied but the exact role of the hepatitis B X protein (HBx) in the context of natural HBV infections remains unknown. METHODS Primary human hepatocytes and differentiated HepaRG cells allowing conditional trans complementation of HBx were infected with wild type (HBV(wt)) or HBx deficient (HBV(x-)) HBV particles and establishment of HBV replication was followed. RESULTS We observed that cells inoculated with HBx-deficient HBV particles (HBV(x-)) did not lead to productive HBV infection contrary to cells inoculated with wild type HBV particles (HBV(wt)). Although equal amounts of nuclear covalently closed circular HBV-DNA (cccDNA) demonstrated comparable uptake and nuclear import, active transcription was only observed from HBV(wt) genomes. Trans-complementation of HBx was able to rescue transcription from the HBV(x-) genome and led to antigen and virion secretion, even weeks after infection. Constant expression of HBx was necessary to maintain HBV antigen expression and replication. Finally, we demonstrated that HBx is not packaged into virions during assembly but is expressed after infection within the new host cell to allow epigenetic control of HBV transcription from cccDNA. CONCLUSIONS Our results demonstrate that HBx is required to initiate and maintain HBV replication and highlight HBx as the key regulator during the natural infection process.
Collapse
Affiliation(s)
- Julie Lucifora
- Institute of Virology, Technische Universität München/Helmholtz Zentrum München, Trogerstrasse, 30, 81675 Munich, Germany
| | | | | | | | | | | | | | | | | |
Collapse
|
20
|
Chen CY, Lin CY, Chen GY, Hu YC. Baculovirus as a gene delivery vector: recent understandings of molecular alterations in transduced cells and latest applications. Biotechnol Adv 2011; 29:618-31. [PMID: 21550393 PMCID: PMC7126054 DOI: 10.1016/j.biotechadv.2011.04.004] [Citation(s) in RCA: 107] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2011] [Revised: 04/15/2011] [Accepted: 04/15/2011] [Indexed: 12/13/2022]
Abstract
Baculovirus infects insects in nature and is non-pathogenic to humans, but can transduce a broad range of mammalian and avian cells. Thanks to the biosafety, large cloning capacity, low cytotoxicity and non-replication nature in the transduced cells as well as the ease of manipulation and production, baculovirus has gained explosive popularity as a gene delivery vector for a wide variety of applications. This article extensively reviews the recent understandings of the molecular mechanisms pertinent to baculovirus entry and cellular responses, and covers the latest advances in the vector improvements and applications, with special emphasis on antiviral therapy, cancer therapy, regenerative medicine and vaccine.
Collapse
Affiliation(s)
- Chi-Yuan Chen
- Department of Chemical Engineering, National Tsing Hua University, Hsinchu 30013, Taiwan
| | | | | | | |
Collapse
|
21
|
Schildgen O. Host factors may influence response to antiviral therapy in chronic hepatitis B virus infections. Med Hypotheses 2011; 76:417-20. [DOI: 10.1016/j.mehy.2010.11.007] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2010] [Revised: 11/02/2010] [Accepted: 11/08/2010] [Indexed: 10/18/2022]
|
22
|
Abstract
Worldwide, it is estimated that more than 350 million people are chronically infected with hepatitis B virus (HBV), approximately 15 million of whom are coinfected with hepatitis D virus (HDV), a satellite of HBV that uses the envelope proteins of the latter to assemble its infectious particles. For a long time after HBV discovery, research on the viral life cycle, viral entry in particular, has been hampered by the lack of practical tissue culture systems. To date, in vitro isolation and serial propagation of HBV are still problematic, but the examination of the entire HBV life cycle is possible using two separate systems: (i) permissive human hepatoma cell lines to study HBV DNA replication, viral transcription, translation, assembly, and release of viral particles and (ii) primary cultures of human or chimpanzee hepatocytes or the susceptible HepaRG cell line for viral entry examination. The experimental model described here for analyzing the function of HBV envelope proteins at viral entry is based on this dual tissue culture system, in which HDV is substituted to HBV for practical reasons.
Collapse
Affiliation(s)
- Camille Sureau
- Laboratoire de Virologie Moléculaire, Institut National de la Transfusion Sanguine, Paris, France
| |
Collapse
|
23
|
Durantel D. Fitness and infectivity of drug-resistant and cross-resistant hepatitis B virus mutants: why and how is it studied? Antivir Ther 2010; 15:521-7. [PMID: 20516574 DOI: 10.3851/imp1551] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
The emergence of hepatitis B virus (HBV) drug-resistant (and multidrug-resistant) strains during long-term therapy with nucleoside/nucleotide analogues is associated with treatment failure and, therefore, represents a clinical challenge. For clinicians, the close monitoring and management of resistance has become a key issue in clinical practice. For HBV virologists, the understanding of the mechanism of emergence of specific mutant strains in the viral quasispecies during treatment is also an important issue. If a particular viral strain can emerge in the quasispecies within a particular environment, it is probably because its fitness is superior to other strains. The present review focuses on viral fitness as well as viral infectivity, and in particular on technical means that are available to study this viral fitness in vitro and in animal models.
Collapse
|
24
|
Zheng H, Liu C, Zhuang J, Yuan S. Baculovirus expression of cloned porcine arterivirus generates infectious particles in both insect and mammalian cells. J Biotechnol 2010; 150:251-8. [PMID: 20728481 PMCID: PMC7114269 DOI: 10.1016/j.jbiotec.2010.08.009] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2010] [Revised: 07/29/2010] [Accepted: 08/02/2010] [Indexed: 12/28/2022]
Abstract
Studies on several viral pathogens have been hampered by the lack of appropriate in vitro systems for their propagation and amplification. Porcine reproductive and respiratory syndrome virus (PRRSV), an arterivirus containing a single-stranded positive-sense RNA genome (∼15kb), was served as a model virus and its genomic cDNA was recombinated into baculovirus. We investigated whether infectious virus particles could be generated by expression of the full-length cloned genome from the modified baculovirus vector. The recombinant baculovirus, AcAPRRS, was used to infect sf9 cells. Immunofluorescence assay demonstrated the presence of PRRSV nonstructural protein (nsp) 2 and nucleocapsid (N) protein and electron microscopy revealed PRRSV particles in the culture supernatant. Infectious PRRSV particles were also produced in susceptible MARC-145 cells inoculated with AcAPRRS, and the growth characteristics of the PRRSV generated were similar to those of the parental PRRSV strain. Infectious PRRSV particles were also generated following AcAPRRS transduction of BHK-21 cells and Vero cells that are not sensitive to PRRSV. Titers of PRRSV obtained from BHK-21 and Vero cells were up to 10(4.05)TCID(50)/ml. These findings open a new route to the propagation of the virus in vitro and will be of utility in vaccine development.
Collapse
Affiliation(s)
- Hao Zheng
- Department of Swine Infectious Diseases, Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, No. 518 Ziyue Road, The Key Laboratory of Animal Parasitology, Chinese Ministry of Agriculture, Shanghai 200241, People's Republic of China
| | | | | | | |
Collapse
|
25
|
Ait-goughoulte M, Lucifora J, Zoulim F, Durantel D. Innate antiviral immune responses to hepatitis B virus. Viruses 2010; 2:1394-1410. [PMID: 21994686 PMCID: PMC3185716 DOI: 10.3390/v2071394] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2010] [Revised: 06/22/2010] [Accepted: 07/01/2010] [Indexed: 02/06/2023] Open
Abstract
Hepatitis B virus (HBV) is a major cause of acute and chronic hepatitis in humans. As HBV itself is currently viewed as a non-cytopathic virus, the liver pathology associated with hepatitis B is mainly thought to be due to immune responses directed against HBV antigens. The outcome of HBV infection is the result of complex interactions between replicating HBV and the immune system. While the role of the adaptive immune response in the resolution of HBV infection is well understood, the contribution of innate immune mechanisms remains to be clearly defined. The innate immune system represents the first line of defense against viral infection, but its role has been difficult to analyze in humans due to late diagnosis of HBV infection. In this review, we discuss recent advances in the field of innate immunity to HBV infection.
Collapse
Affiliation(s)
- Malika Ait-goughoulte
- INSERM, U871, Molecular Physiopathology and New Treatment of Viral Hepatitis, 151 Cours Albert Thomas, 69003 Lyon, France; E-Mails: (M.A.-g.); (J.L.); (F.Z.)
- Université de Lyon, UCBL, and IFR62 Lyon Est, 69008 Lyon, France
| | - Julie Lucifora
- INSERM, U871, Molecular Physiopathology and New Treatment of Viral Hepatitis, 151 Cours Albert Thomas, 69003 Lyon, France; E-Mails: (M.A.-g.); (J.L.); (F.Z.)
- Université de Lyon, UCBL, and IFR62 Lyon Est, 69008 Lyon, France
| | - Fabien Zoulim
- INSERM, U871, Molecular Physiopathology and New Treatment of Viral Hepatitis, 151 Cours Albert Thomas, 69003 Lyon, France; E-Mails: (M.A.-g.); (J.L.); (F.Z.)
- Université de Lyon, UCBL, and IFR62 Lyon Est, 69008 Lyon, France
- Hospices Civils de Lyon (HCL), Hôtel Dieu Hospital, 69002 Lyon, France
| | - David Durantel
- INSERM, U871, Molecular Physiopathology and New Treatment of Viral Hepatitis, 151 Cours Albert Thomas, 69003 Lyon, France; E-Mails: (M.A.-g.); (J.L.); (F.Z.)
- Université de Lyon, UCBL, and IFR62 Lyon Est, 69008 Lyon, France
- Hospices Civils de Lyon (HCL), Hôtel Dieu Hospital, 69002 Lyon, France
- Author to whom correspondence should be addressed; E-Mail: ; Tel.: +33-472-681-970; Fax: +33-472-681-971
| |
Collapse
|
26
|
Zhao Y, Ben H, Qu S, Zhou X, Yan L, Xu B, Zhou S, Lou Q, Ye R, Zhou T, Yang P, Qu D. Proteomic analysis of primary duck hepatocytes infected with duck hepatitis B virus. Proteome Sci 2010; 8:28. [PMID: 20529248 PMCID: PMC2904733 DOI: 10.1186/1477-5956-8-28] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2010] [Accepted: 06/07/2010] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Hepatitis B virus (HBV) is a major cause of liver infection in human. Because of the lack of an appropriate cell culture system for supporting HBV infection efficiently, the cellular and molecular mechanisms of hepadnavirus infection remain incompletely understood. Duck heptatitis B virus (DHBV) can naturally infect primary duck hepatocytes (PDHs) that provide valuable model systems for studying hepadnavirus infection in vitro. In this report, we explored global changes in cellular protein expression in DHBV infected PDHs by two-dimension gel electrophoresis (2-DE) combined with MALDI-TOF/TOF tandem mass spectrometry (MS/MS). RESULTS The effects of hepadnavirus infection on hepatocytes were investigated in DHBV infected PDHs by the 2-DE analysis. Proteomic profile of PDHs infected with DHBV were analyzed at 24, 72 and 120 h post-infection by comparing with uninfected PDHs, and 75 differentially expressed protein spots were revealed by 2-DE analysis. Among the selected protein spots, 51 spots were identified corresponding to 42 proteins by MS/MS analysis; most of them were matched to orthologous proteins of Gallus gallus, Anas platyrhynchos or other avian species, including alpha-enolase, lamin A, aconitase 2, cofilin-2 and annexin A2, etc. The down-regulated expression of beta-actin and annexin A2 was confirmed by Western blot analysis, and potential roles of some differentially expressed proteins in the virus-infected cells have been discussed. CONCLUSIONS Differentially expressed proteins of DHBV infected PDHs revealed by 2-DE, are involved in carbohydrate metabolism, amino acid metabolism, stress responses and cytoskeleton processes etc, providing the insight to understanding of interactions between hepadnavirus and hepatocytes and molecular mechanisms of hepadnavirus pathogenesis.
Collapse
Affiliation(s)
- Yanfeng Zhao
- Key Laboratory of Medical Molecular Virology of Ministries of Education and Health, Institute of Medical Microbiology and Institutes of Biomedical Sciences, Shanghai Medical College of Fudan University, Shanghai, China
| | - Haijing Ben
- Key Laboratory of Medical Molecular Virology of Ministries of Education and Health, Institute of Medical Microbiology and Institutes of Biomedical Sciences, Shanghai Medical College of Fudan University, Shanghai, China
| | - Su Qu
- Key Laboratory of Medical Molecular Virology of Ministries of Education and Health, Institute of Medical Microbiology and Institutes of Biomedical Sciences, Shanghai Medical College of Fudan University, Shanghai, China
| | - Xinwen Zhou
- Department of Chemistry, Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| | - Liang Yan
- Key Laboratory of Medical Molecular Virology of Ministries of Education and Health, Institute of Medical Microbiology and Institutes of Biomedical Sciences, Shanghai Medical College of Fudan University, Shanghai, China
| | - Bin Xu
- Key Laboratory of Medical Molecular Virology of Ministries of Education and Health, Institute of Medical Microbiology and Institutes of Biomedical Sciences, Shanghai Medical College of Fudan University, Shanghai, China
| | - Shuangcheng Zhou
- Key Laboratory of Medical Molecular Virology of Ministries of Education and Health, Institute of Medical Microbiology and Institutes of Biomedical Sciences, Shanghai Medical College of Fudan University, Shanghai, China
| | - Qiang Lou
- Key Laboratory of Medical Molecular Virology of Ministries of Education and Health, Institute of Medical Microbiology and Institutes of Biomedical Sciences, Shanghai Medical College of Fudan University, Shanghai, China
| | - Rong Ye
- Key Laboratory of Medical Molecular Virology of Ministries of Education and Health, Institute of Medical Microbiology and Institutes of Biomedical Sciences, Shanghai Medical College of Fudan University, Shanghai, China
| | - Tianlun Zhou
- Key Laboratory of Medical Molecular Virology of Ministries of Education and Health, Institute of Medical Microbiology and Institutes of Biomedical Sciences, Shanghai Medical College of Fudan University, Shanghai, China
| | - Pengyuan Yang
- Department of Chemistry, Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| | - Di Qu
- Key Laboratory of Medical Molecular Virology of Ministries of Education and Health, Institute of Medical Microbiology and Institutes of Biomedical Sciences, Shanghai Medical College of Fudan University, Shanghai, China
| |
Collapse
|
27
|
Lucifora J, Vincent IE, Berthillon P, Dupinay T, Michelet M, Protzer U, Zoulim F, Durantel D, Trepo C, Chemin I. Hepatitis B virus replication in primary macaque hepatocytes: crossing the species barrier toward a new small primate model. Hepatology 2010; 51:1954-1960. [PMID: 20301206 DOI: 10.1002/hep.23602] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
UNLABELLED The development of new anti-hepatitis B virus (HBV) therapies, especially immunotherapeutic approaches, has been limited by the lack of a primate model more accessible than chimpanzees. We have previously demonstrated that sylvanus and cynomolgus macaques are susceptible to in vivo HBV infection after intrahepatic HBV DNA inoculation. In this study, we evaluated the susceptibility of primary macaque hepatocytes (PMHs) to HBV infection with a highly efficient HBV genome-mediated transfer system via a recombinant baculovirus (Bac-HBV). Freshly prepared PMHs, isolated from macaque liver tissue by collagenase perfusion, were transduced with Bac-HBV, and intermediates of replication were followed for 9 days post-transduction. Evidence of HBV replication (hepatitis B surface antigen secretion, viral DNA, RNA, and covalently closed circular DNA) was detected from day 1 to day 9 post-transduction. HBV markers were dose-dependent and still detectable at a multiplicity of infection of 10. Importantly, transduced PMHs secreted all typical forms of HBV particles, as evidenced by a cesium chloride gradient as well as transmission electron microscopy. Furthermore, the Toll-like receptor 9 (TLR9) ligand was used to stimulate freshly prepared macaque peripheral blood mononuclear cells to generate TLR9-induced cytokines. We then demonstrated the antiviral effects of both TLR9-induced cytokines and nucleoside analogue (lamivudine) on HBV replication in transduced PMHs. CONCLUSION Baculovirus-mediated genome transfer initiated a full HBV replication cycle in PMHs; thus highlighted both the baculovirus efficiency in crossing the species barrier and macaque susceptibility to HBV infection. Moreover, our results demonstrate the relevance of thus system for antiviral compound evaluations with either nucleoside analogues or inhibitory cytokines. Cynomolgus macaques are readily available, are immunologically closely related to humans, and may therefore represent a promising model for the development of new immunotherapeutic strategies.
Collapse
Affiliation(s)
- Julie Lucifora
- INSERM Unité 871, Institut National de la Santé et de la Recherche Médicale, Lyon, France
| | | | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Marion MJ, Hantz O, Durantel D. The HepaRG cell line: biological properties and relevance as a tool for cell biology, drug metabolism, and virology studies. Methods Mol Biol 2010; 640:261-72. [PMID: 20645056 DOI: 10.1007/978-1-60761-688-7_13] [Citation(s) in RCA: 98] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/23/2023]
Abstract
Liver progenitor cells may play an important role in carcinogenesis in vivo and represent therefore useful cellular materials for in vitro studies. The HepaRG cell line, which is a human bipotent progenitor cell line capable to differentiate toward two different cell phenotypes (i.e., biliary-like and hepatocyte-like cells), has been established from a liver tumor associated with chronic hepatitis C. This cell line represents a valuable alternative to ex vivo cultivated primary human hepatocytes (PHH), as HepaRG cells share some features and properties with adult hepatocytes. The cell line is particularly useful to evaluate drugs and perform drug metabolism studies, as many detoxifying enzymes are expressed and functional. It is also an interesting tool to study some aspect of progenitor biology (e.g., differentiation process), carcinogenesis, and the infection by some pathogens for which the cell line is permissive (e.g., HBV infection). Overall, this chapter gives a concise overview of the biological properties and potential applications of this cell line.
Collapse
Affiliation(s)
- Marie-Jeanne Marion
- INSERM U871, Molecular physiopathology and new treatments of viral hepatitis, Lyon, France
| | | | | |
Collapse
|
29
|
Lucifora J, Durantel D, Testoni B, Hantz O, Levrero M, Zoulim F. Control of hepatitis B virus replication by innate response of HepaRG cells. Hepatology 2010; 51:63-72. [PMID: 19877170 DOI: 10.1002/hep.23230] [Citation(s) in RCA: 108] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
UNLABELLED Hepatitis B virus (HBV) is currently viewed as a stealth virus that does not elicit innate immunity in vivo. This assumption has not yet been challenged in vitro because of the lack of a relevant cell culture system. The HepaRG cell line, which is physiologically closer to differentiated hepatocytes and permissive to HBV infection, has opened new perspectives in this respect.HBV baculoviruses were used to initiate an HBV replication in both HepG2 and HepaRG cells. To monitor HBV replication, the synthesis of encapsidated DNA, and secretion of hepatitis B surface antigen (HBsAg), was respectively analyzed by southern blot and enzyme-linked immunosorbent assay. The induction of a type I interferon (IFN) response was monitored by targeted quantitative reverse transcription polymerase chain reaction (qRT-PCR), low-density arrays, and functional assays. The invalidation of type I IFN response was obtained by either antibody neutralization or RNA interference. We demonstrate that HBV elicits a strong and specific innate antiviral response that results in a noncytopathic clearance of HBV DNA in HepaRG cells. Challenge experiment showed that transduction with Bac-HBV-WT, but not with control baculoviruses, leads to this antiviral response in HepaRG cells, whereas no antiviral response is observed in HepG2 cells. Cellular gene expression analyses showed that IFN-beta and other IFN-stimulated genes were up-regulated in HepG2 and HepaRG cells, but not in cells transduced by control baculoviruses. Interestingly, a rescue of viral replication was observed when IFN-beta action was neutralized by antibodies or RNA interference of type I IFN receptor. CONCLUSION Our data suggest that a strong HBV replication is able to elicit a type I IFN response in HepaRG-transduced cells.
Collapse
Affiliation(s)
- Julie Lucifora
- INSERM (Institut National de la Santé et de la Recherche Médicale), U871, Lyon, France
| | | | | | | | | | | |
Collapse
|
30
|
Selection and counterselection of the rtI233V adefovir resistance mutation during antiviral therapy. J Clin Microbiol 2009; 48:631-4. [PMID: 20007398 DOI: 10.1128/jcm.01073-09] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Recently, we reported on three patients with chronic hepatitis B virus (HBV) infection for whom adefovir (ADF) therapy virologically failed, most likely due to a preexisting rtI233V HBV polymerase mutation. Here, we describe two further patients with chronic HBV infection who were found to develop the rtI233V mutation after initiation of ADF therapy. These patients represent the first cases known so far in which the rtI233V ADF resistance mutation evolved under persistent HBV replication during HBV therapy with ADF. Interestingly, one of the previously described patients, who was initially successfully switched from ADF to tenofovir (TDF) and became virologically suppressed subsequently, experienced a moderate but remarkable rebound of HBV viremia after switching from TDF to entecavir, due to the emergence of renal toxicity. Thus, we provide evidence for the selection and counterselection of the rtI233V ADF resistance mutation during antiviral therapy.
Collapse
|
31
|
Durantel D, Zoulim F. Innate response to hepatitis B virus infection: observations challenging the concept of a stealth virus. Hepatology 2009; 50:1692-5. [PMID: 19937686 DOI: 10.1002/hep.23361] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
|
32
|
Levrero M, Pollicino T, Petersen J, Belloni L, Raimondo G, Dandri M. Control of cccDNA function in hepatitis B virus infection. J Hepatol 2009; 51:581-92. [PMID: 19616338 DOI: 10.1016/j.jhep.2009.05.022] [Citation(s) in RCA: 420] [Impact Index Per Article: 26.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
The template of hepatitis B virus (HBV) transcription, the covalently closed circular DNA (cccDNA), plays a key role in the life cycle of the virus and permits the persistence of infection. Novel molecular techniques have opened new possibilities to investigate the organization and the activity of the cccDNA minichromosome in vivo, and recent advances have started to shed light on the complexity of the mechanisms controlling cccDNA function. Nuclear cccDNA accumulates in hepatocyte nuclei as a stable minichromosome organized by histone and non-histone viral and cellular proteins. Identification of the molecular mechanisms regulating cccDNA stability and its transcriptional activity at the RNA, DNA and epigenetic levels in the course of chronic hepatitis B (CH-B) infection may reveal new potential therapeutic targets for anti-HBV drugs and hence assist in the design of strategies aimed at silencing and eventually depleting the cccDNA reservoir.
Collapse
Affiliation(s)
- Massimo Levrero
- Department of Internal Medicine, Sapienza University of Rome, Policlinico Umberto I, 0061 Rome, Italy.
| | | | | | | | | | | |
Collapse
|
33
|
HBV life cycle: entry and morphogenesis. Viruses 2009; 1:185-209. [PMID: 21994545 PMCID: PMC3185491 DOI: 10.3390/v1020185] [Citation(s) in RCA: 84] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2009] [Revised: 07/31/2009] [Accepted: 08/13/2009] [Indexed: 02/07/2023] Open
Abstract
Hepatitis B virus (HBV) is a major cause of liver disease. HBV primarily infects hepatocytes by a still poorly understood mechanism. After an endocytotic process, the nucleocapsids are released into the cytoplasm and the relaxed circular rcDNA genome is transported towards the nucleus where it is converted into covalently closed circular cccDNA. Replication of the viral genome occurs via an RNA pregenome (pgRNA) that binds to HBV polymerase (P). P initiates pgRNA encapsidation and reverse transcription inside the capsid. Matured, rcDNA containing nucleocapsids can re-deliver the RC-DNA to the nucleus, or be secreted via interaction with the envelope proteins as progeny virions.
Collapse
|
34
|
Vincent IE, Lucifora J, Durantel D, Hantz O, Chemin I, Zoulim F, Trepo C. Inhibitory effect of the combination of CpG-induced cytokines with lamivudine against hepatitis B virus replication in vitro. Antivir Ther 2009. [DOI: 10.1177/135965350901400115] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Background Currently approved antiviral monotherapies against chronic hepatitis B fail to eradicate hepatitis B virus (HBV), to overcome the defects in HBV-specific immune responses and to prevent HBV relapse after cessation of therapy. CpG oligodesoxynucleotides (CpG ODN) are synthetic agonists of Toll-like receptor 9 and potent inducers of innate and acquired immunity. Our aim was to establish the proof of concept of the antiviral benefit of combining a nucleoside analogue with CpG-induced cytokines on HBV replication in vitro. Methods Peripheral blood mononuclear cells from HBV-negative individuals were stimulated with CpG ODN to generate CpG-induced cytokine supernatants. Proliferating HepaRG and HepG2 cells were transduced with recombinant HBV baculovirus and differentiated HepaRG cells were inoculated with HBV virions. Antiviral effects of CpG-induced cytokine with or without lamivudine were evaluated by analysing HBV DNA, HBV RNA and antigen secretion (hepatitis B surface antigen [HBsAg] and hepatitis B e antigen [HBeAg]). Results Following transduction or HBV inoculation, CpG-induced cytokines strongly inhibited HBV viral intermediates of replication, as well as HBsAg and HBeAg secretion from infected cells. Strikingly, in transduced HepaRG cells, the combination of CpG-induced cytokines with lamivudine reduced the 50% effective concentration of lamivudine by 100-fold. Importantly, the treatment of CpG-induced cytokines prior to HBV inoculation conferred a partial protection against infection to hepatocytes. Conclusions CpG-induced cytokines associated with polymerase inhibitors represent a promising combination to suppress HBV replication. Such an immunotherapeutic strategy should be evaluated in vivo to assess restoration and duration of anti-HBV-specific immune responses.
Collapse
Affiliation(s)
| | - Julie Lucifora
- INSERM U871, Lyon, France
- Université Lyon 1, Lyon, France
| | - David Durantel
- INSERM U871, Lyon, France
- Université Lyon 1, Lyon, France
- Hospices Civils de Lyon, Hôtel Dieu Hospital, Lyon, France
| | - Olivier Hantz
- INSERM U871, Lyon, France
- Université Lyon 1, Lyon, France
| | | | - Fabien Zoulim
- INSERM U871, Lyon, France
- Université Lyon 1, Lyon, France
- Hospices Civils de Lyon, Hôtel Dieu Hospital, Lyon, France
| | - Christian Trepo
- INSERM U871, Lyon, France
- Université Lyon 1, Lyon, France
- Hospices Civils de Lyon, Hôtel Dieu Hospital, Lyon, France
| |
Collapse
|
35
|
Margeridon S, Carrouée-Durantel S, Chemin I, Barraud L, Zoulim F, Trépo C, Kay A. Rolling circle amplification, a powerful tool for genetic and functional studies of complete hepatitis B virus genomes from low-level infections and for directly probing covalently closed circular DNA. Antimicrob Agents Chemother 2008; 52:3068-3073. [PMID: 18606836 PMCID: PMC2533481 DOI: 10.1128/aac.01318-07] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2007] [Revised: 11/21/2007] [Accepted: 07/01/2008] [Indexed: 02/07/2023] Open
Abstract
Complete characterization of the biological properties of hepatitis B virus (HBV) variants requires the generation of full-length genomes. The aim of this study was to develop new tools for the efficient full-length genome amplification of virus from samples with low viral loads. Rolling circle amplification (RCA) was used to amplify full-length HBV genomes from both sera and liver biopsy samples from chronic HBV carriers. Serum-derived relaxed circular HBV DNA could be amplified only after completion and ligation of plus-strand DNA. Covalently closed circular DNA (cccDNA) from liver biopsies could be amplified directly from as few as 13 copies, using RCA, followed by a full-length HBV PCR. Three serial liver biopsy samples were obtained from a lamivudine-resistant patient who cleared detectable serum HBV after adefovir dipivoxil was added to the lamivudine therapy and then seroconverted to anti-HBs. Only the genomes from the last biopsy specimen obtained after the emergence of lamivudine resistance contained the lamivudine resistance-associated mutations rtL180M and rtM204V ("rt" indicates reverse transcriptase domain). Defective genomes were also found in this biopsy sample. Genomes cloned from the liver biopsy specimens were transfected into HuH7 cells to study their replication competence and their susceptibility to lamivudine. RCA is a powerful tool for amplifying full-length HBV genomes and will be especially useful for the study of occult or inactive HBV infections and patients undergoing antiviral treatment. It can also be used to probe HBV cccDNA, the crucial intermediate in viral persistence and the archive of resistance mutations.
Collapse
MESH Headings
- Antiviral Agents/pharmacology
- Antiviral Agents/therapeutic use
- Carrier State/drug therapy
- Carrier State/virology
- Cell Line, Tumor
- DNA, Circular/analysis
- DNA, Circular/biosynthesis
- DNA, Viral/analysis
- DNA, Viral/biosynthesis
- Drug Resistance, Viral
- Genome, Viral
- Hepatitis B virus/genetics
- Hepatitis B virus/metabolism
- Hepatitis B, Chronic/drug therapy
- Hepatitis B, Chronic/virology
- Humans
- Lamivudine/pharmacology
- Lamivudine/therapeutic use
- Molecular Sequence Data
- Nucleic Acid Amplification Techniques/methods
- Sequence Analysis, DNA
- Transfection
- Virus Replication
Collapse
|