1
|
Yu X, Gao Y, Zhang X, Ji L, Fang M, Li M, Gao Y. Hepatitis B: Model Systems and Therapeutic Approaches. J Immunol Res 2024; 2024:4722047. [PMID: 38745751 PMCID: PMC11093688 DOI: 10.1155/2024/4722047] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2023] [Revised: 03/29/2024] [Accepted: 04/05/2024] [Indexed: 05/16/2024] Open
Abstract
Hepatitis B virus (HBV) infection is a major global health issue and ranks among the top causes of liver cirrhosis and hepatocellular carcinoma. Although current antiviral medications, including nucleot(s)ide analogs and interferons, could inhibit the replication of HBV and alleviate the disease, HBV cannot be fully eradicated. The development of cellular and animal models for HBV infection plays an important role in exploring effective anti-HBV medicine. During the past decades, advancements in several cell culture systems, such as HepG2.2.15, HepAD38, HepaRG, hepatocyte-like cells, and primary human hepatocytes, have propelled the research in inhibiting HBV replication and expression and thus enriched our comprehension of the viral life cycle and enhancing antiviral drug evaluation efficacy. Mouse models, in particular, have emerged as the most extensively studied HBV animal models. Additionally, the present landscape of HBV therapeutics research now encompasses a comprehensive assessment of the virus's life cycle, targeting numerous facets and employing a variety of immunomodulatory approaches, including entry inhibitors, strategies aimed at cccDNA, RNA interference technologies, toll-like receptor agonists, and, notably, traditional Chinese medicine (TCM). This review describes the attributes and limitations of existing HBV model systems and surveys novel advancements in HBV treatment modalities, which will offer deeper insights toward discovering potentially efficacious pharmaceutical interventions.
Collapse
Affiliation(s)
- Xiaoxiao Yu
- Laboratory of Cellular Immunity, Shuguang Hospital, Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yating Gao
- Laboratory of Cellular Immunity, Shuguang Hospital, Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Xin Zhang
- Laboratory of Cellular Immunity, Shuguang Hospital, Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Longshan Ji
- Laboratory of Cellular Immunity, Shuguang Hospital, Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Miao Fang
- Laboratory of Cellular Immunity, Shuguang Hospital, Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Man Li
- Laboratory of Cellular Immunity, Shuguang Hospital, Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yueqiu Gao
- Laboratory of Cellular Immunity, Shuguang Hospital, Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Department of Hepatopathy, Shuguang Hospital, Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Institute of Infectious Diseases of Integrated Traditional Chinese and Western Medicine, Shanghai, China
| |
Collapse
|
2
|
Bhat S, Kazim SN. HBV cccDNA-A Culprit and Stumbling Block for the Hepatitis B Virus Infection: Its Presence in Hepatocytes Perplexed the Possible Mission for a Functional Cure. ACS OMEGA 2022; 7:24066-24081. [PMID: 35874215 PMCID: PMC9301636 DOI: 10.1021/acsomega.2c02216] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/17/2023]
Abstract
Hepatitis B virus infection (HBV) is still a big health problem across the globe. It has been linked to the development of liver cirrhosis and hepatocellular carcinoma and can trigger different types of liver damage. Existing medicines are unable to disable covalently closed circular DNA (cccDNA), which may result in HBV persistence and recurrence. The current therapeutic goal is to achieve a functional cure, which means HBV-DNA no longer exists when treatment stops and the absence of HBsAg seroclearance. However, due to the presence of integrated HBV DNA and cccDNA functional treatment is now regarded to be difficult. In order to uncover pathways for potential therapeutic targets and identify medicines that could result in large rates of functional cure, a thorough understanding of the virus' biology is required. The proteins of the virus and episomal cccDNA are thought to be critical for the management and support of the HBV replication cycle as they interact directly with the host proteome to establish the best atmosphere for the virus while evading immune detection. The breakthroughs of host dependence factors, cccDNA transcription, epigenetic regulation, and immune-mediated breakdown have all produced significant progress in our understanding of cccDNA biology during the past decade. There are some strategies where cccDNA can be targeted either in a direct or indirect way and are presently at the point of discovery or preclinical or early clinical advancement. Editing of genomes, techniques targeting host dependence factors or epigenetic gene maintenance, nucleocapsid modulators, miRNA, siRNA, virion secretory inhibitors, and immune-mediated degradation are only a few examples. Though cccDNA approaches for direct targeting are still in the early stages of development, the assembly of capsid modulators and immune-reliant treatments have made it to the clinic. Clinical trials are currently being conducted to determine their efficiency and safety in patients, as well as their effect on viral cccDNA. The influence of recent breakthroughs in the development of new treatment techniques on cccDNA biology is also summarized in this review.
Collapse
Affiliation(s)
- Sajad
Ahmad Bhat
- Jamia Millia Islamia Central University, Centre for Interdisciplinary Research in Basic Sciences, New Delhi 110025, India
| | - Syed Naqui Kazim
- Jamia Millia Islamia Central University, Centre for Interdisciplinary Research in Basic Sciences, New Delhi 110025, India
| |
Collapse
|
3
|
Proulx J, Ghaly M, Park IW, Borgmann K. HIV-1-Mediated Acceleration of Oncovirus-Related Non-AIDS-Defining Cancers. Biomedicines 2022; 10:biomedicines10040768. [PMID: 35453518 PMCID: PMC9024568 DOI: 10.3390/biomedicines10040768] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Revised: 03/22/2022] [Accepted: 03/22/2022] [Indexed: 11/25/2022] Open
Abstract
With the advent of combination antiretroviral therapy (cART), overall survival has been improved, and the incidence of acquired immunodeficiency syndrome (AIDS)-defining cancers has also been remarkably reduced. However, non-AIDS-defining cancers among human immunodeficiency virus-1 (HIV-1)-associated malignancies have increased significantly so that cancer is the leading cause of death in people living with HIV in certain highly developed countries, such as France. However, it is currently unknown how HIV-1 infection raises oncogenic virus-mediated cancer risks in the HIV-1 and oncogenic virus co-infected patients, and thus elucidation of the molecular mechanisms for how HIV-1 expedites the oncogenic viruses-triggered tumorigenesis in the co-infected hosts is imperative for developing therapeutics to cure or impede the carcinogenesis. Hence, this review is focused on HIV-1 and oncogenic virus co-infection-mediated molecular processes in the acceleration of non-AIDS-defining cancers.
Collapse
|
4
|
Li X, Xu Z, Mitra B, Wang M, Guo H, Feng Z. Elevated NTCP expression by an iPSC-derived human hepatocyte maintenance medium enhances HBV infection in NTCP-reconstituted HepG2 cells. Cell Biosci 2021; 11:123. [PMID: 34225786 PMCID: PMC8256212 DOI: 10.1186/s13578-021-00641-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2021] [Accepted: 06/28/2021] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND The sodium taurocholate cotransporting polypeptide (NTCP) is a functional receptor for hepatitis B virus (HBV). NTCP-reconstituted human hepatoma cells support HBV infection, but the infection is suboptimal and no apparent HBV spread has been observed in this system. RESULTS We found that NTCP-reconstituted HepG2 cells were highly susceptible to HBV infection after cells were cultured in a commercial human inducible pluripotent stem cell (iPSC)-derived hepatocyte maintenance medium (HMM). The enhanced HBV infection coincided with increased NTCP expression, and was observed in six different clones of HepG2-NTCP cells. Promoter assays indicated that HMM activated the cytomegalovirus immediate-early (IE) promoter that drives the NTCP expression in the HepG2-NTCP cells. RNA-Seq analysis revealed that HMM upregulated multiple metabolic pathways. Despite highly upregulated NTCP expression by HMM, no obvious HBV spread was observed even in the presence of PEG 8000. CONCLUSIONS Our data suggest that this particular medium could be used to enhance HBV infection in NTCP-reconstituted hepatocytes in vitro.
Collapse
Affiliation(s)
- Xinlei Li
- Center for Vaccines and Immunity, The Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, OH, 43205, USA
| | - Zhaohui Xu
- Center for Vaccines and Immunity, The Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, OH, 43205, USA
| | - Bidisha Mitra
- Department of Microbiology and Molecular Genetics and UPMC Hillman Cancer Center, University of Pittsburgh, Pittsburgh, PA, USA
| | - Minghang Wang
- Center for Vaccines and Immunity, The Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, OH, 43205, USA
| | - Haitao Guo
- Department of Microbiology and Molecular Genetics and UPMC Hillman Cancer Center, University of Pittsburgh, Pittsburgh, PA, USA
| | - Zongdi Feng
- Center for Vaccines and Immunity, The Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, OH, 43205, USA.
- Department of Pediatrics, The Ohio State University College of Medicine, Columbus, OH, USA.
| |
Collapse
|
5
|
Wettengel JM, Burwitz BJ. Innovative HBV Animal Models Based on the Entry Receptor NTCP. Viruses 2020; 12:E828. [PMID: 32751581 PMCID: PMC7472226 DOI: 10.3390/v12080828] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Revised: 07/24/2020] [Accepted: 07/27/2020] [Indexed: 02/07/2023] Open
Abstract
Hepatitis B is a major global health problem, with an estimated 257 million chronically infected patients and almost 1 million deaths per year. The causative agent is hepatitis B virus (HBV), a small, enveloped, partially double-stranded DNA virus. HBV has a strict species specificity, naturally infecting only humans and chimpanzees. Sodium taurocholate co-transporting polypeptide (NTCP), a bile acid transporter expressed on hepatocytes, has been shown to be one of the key factors in HBV infection, playing a crucial role in the HBV entry process in vitro and in vivo. Variations in the amino acid sequence of NTCP can inhibit HBV infection and, therefore, contributes, in part, to the species barrier. This discovery has revolutionized the search for novel animal models of HBV. Indeed, it was recently shown that variations in the amino acid sequence of NTCP represent the sole species barrier for HBV infection in macaques. Here, we review what is known about HBV entry through the NTCP receptor and highlight how this knowledge has been harnessed to build new animal models for the study of HBV pathogenesis and curative therapies.
Collapse
Affiliation(s)
- Jochen M. Wettengel
- Institute of Virology, Technische Universität München/Helmholtz Zentrum München, Trogerstr. 30, 81675 Munich, Germany;
- Vaccine & Gene Therapy Institute, Oregon Health & Science University, 505 N.W. 185th Avenue Beaverton, Tanasbourne, OR 97006, USA
| | - Benjamin J. Burwitz
- Vaccine & Gene Therapy Institute, Oregon Health & Science University, 505 N.W. 185th Avenue Beaverton, Tanasbourne, OR 97006, USA
| |
Collapse
|
6
|
Spyrou E, Smith CI, Ghany MG. Hepatitis B: Current Status of Therapy and Future Therapies. Gastroenterol Clin North Am 2020; 49:215-238. [PMID: 32389360 PMCID: PMC7444867 DOI: 10.1016/j.gtc.2020.01.003] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Despite the availability of a protective vaccine for over 3 decades, the number of persons with chronic hepatitis B virus (HBV) infection remains high. These persons are at risk for cirrhosis and hepatocellular carcinoma. Current treatment is effective at inhibiting viral replication and reducing complications of chronic HBV infection, but is not curative. There is a need for novel, finite therapy that can cure chronic HBV infection. Several agents are in early-phase development and can be broadly viewed as agents that target the virus directly or indirectly or the host immune response. This article highlights key developments in antiviral/immunomodulatory therapy, the rationale for these approaches, and possible therapeutic regimens.
Collapse
Affiliation(s)
- Elias Spyrou
- MedStar Georgetown Transplant Institute, MedStar Georgetown University Hospital, Washington, DC, USA,Nazih Zuhdi Transplant Institute, INTEGRIS Baptist Medical Center, Oklahoma City, OK, USA
| | - Coleman I. Smith
- MedStar Georgetown Transplant Institute, MedStar Georgetown University Hospital, Washington, DC, USA
| | - Marc G. Ghany
- Liver Diseases Branch, National Institutes of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
7
|
An P, Zeng Z, Winkler CA. The Loss-of-Function S267F Variant in HBV Receptor NTCP Reduces Human Risk for HBV Infection and Disease Progression. J Infect Dis 2019; 218:1404-1410. [PMID: 29905807 DOI: 10.1093/infdis/jiy355] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2018] [Accepted: 06/11/2018] [Indexed: 02/07/2023] Open
Abstract
Background Sodium taurocholate cotransporting polypeptide (NTCP, SLC10A1) is a hepatocyte receptor for hepatitis B virus (HBV) infection. The natural NTCP S267F variant causes loss of NTCP HBV receptor function. We assessed the association of S267F with HBV resistance, HBV infection clearance, and HBV-related cirrhosis and hepatocellular carcinoma (HCC). Methods We tested the effects of S267F in 1117 Han Chinese patients with various HBV infection outcomes using multivariate logistic regression analysis. Results The frequency of S267F (T allele) was higher in HBV-resistant healthy controls (n = 179, 4.0%) compared to HBV-infected patients (n = 648, 1.5%); odds ratio (OR) 0.32 (95% confidence interval [CI] 0.15-0.68; P = .003; dominant model). 267F variant genotypes were also associated with reduced risk for cirrhosis (n = 192, 0.5%) and HCC (n = 258, 1.0%) compared to those with chronic HBV infection (n = 202, 3.0%); OR 0.15 (95% CI, 0.03-0.70) and OR 0.21 (95% CI, 0.062-0.72), respectively. There was no association of the S267F variant with spontaneous HBV clearance. Conclusion The S267F variant for the HBV cell-entry receptor NTCP was associated with increased resistance to HBV infection and decreased risk for cirrhosis and liver cancer among those with chronic HBV infection.
Collapse
Affiliation(s)
- Ping An
- Basic Research Laboratory, Center for Cancer Research, National Cancer Institute.,Basic Science Program, Leidos Biomedical Research Inc., Frederick National Laboratory for Cancer Research, Frederick, Maryland
| | - Zheng Zeng
- Department of Infectious Diseases, Peking University First Hospital, Beijing, China
| | - Cheryl A Winkler
- Basic Research Laboratory, Center for Cancer Research, National Cancer Institute.,Basic Science Program, Leidos Biomedical Research Inc., Frederick National Laboratory for Cancer Research, Frederick, Maryland
| |
Collapse
|
8
|
A Single Adaptive Mutation in Sodium Taurocholate Cotransporting Polypeptide Induced by Hepadnaviruses Determines Virus Species Specificity. J Virol 2019; 93:JVI.01432-18. [PMID: 30541857 DOI: 10.1128/jvi.01432-18] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2018] [Accepted: 12/05/2018] [Indexed: 12/14/2022] Open
Abstract
Hepatitis B virus (HBV) and its hepadnavirus relatives infect a wide range of vertebrates, from fish to human. Hepadnaviruses and their hosts have a long history of acquiring adaptive mutations. However, there are no reports providing direct molecular evidence for such a coevolutionary "arms race" between hepadnaviruses and their hosts. Here, we present evidence suggesting that the adaptive evolution of the sodium taurocholate cotransporting polypeptide (NTCP), an HBV receptor, has been influenced by virus infection. Evolutionary analysis of the NTCP-encoding genes from 20 mammals showed that most NTCP residues are highly conserved among species, exhibiting evolution under negative selection (dN/dS ratio [ratio of nonsynonymous to synonymous evolutionary changes] of <1); this observation implies that the evolution of NTCP is restricted by maintaining its original protein function. However, 0.7% of NTCP amino acid residues exhibit rapid evolution under positive selection (dN/dS ratio of >1). Notably, a substitution at amino acid (aa) 158, a positively selected residue, converting the human NTCP to a monkey-type sequence abrogated the capacity to support HBV infection; conversely, a substitution at this residue converting the monkey Ntcp to the human sequence was sufficient to confer HBV susceptibility. Together, these observations suggested a close association of the aa 158 positive selection with the pressure by virus infection. Moreover, the aa 158 sequence determined attachment of the HBV envelope protein to the host cell, demonstrating the mechanism whereby HBV infection would create positive selection at this NTCP residue. In summary, we provide the first evidence in agreement with the function of hepadnavirus as a driver for inducing adaptive mutation in host receptor.IMPORTANCE HBV and its hepadnavirus relatives infect a wide range of vertebrates, with a long infectious history (hundreds of millions of years). Such a long history generally allows adaptive mutations in hosts to escape from infection while simultaneously allowing adaptive mutations in viruses to overcome host barriers. However, there is no published molecular evidence for such a coevolutionary arms race between hepadnaviruses and hosts. In the present study, we performed coevolutionary phylogenetic analysis between hepadnaviruses and the sodium taurocholate cotransporting polypeptide (NTCP), an HBV receptor, combined with virological experimental assays for investigating the biological significance of NTCP sequence variation. Our data provide the first molecular evidence supporting that HBV-related hepadnaviruses drive adaptive evolution in the NTCP sequence, including a mechanistic explanation of how NTCP mutations determine host viral susceptibility. Our novel insights enhance our understanding of how hepadnaviruses evolved with their hosts, permitting the acquisition of strong species specificity.
Collapse
|
9
|
Zhao K, Liu S, Chen Y, Yao Y, Zhou M, Yuan Y, Wang Y, Pei R, Chen J, Hu X, Zhou Y, Zhao H, Lu M, Wu C, Chen X. Upregulation of HBV transcription by sodium taurocholate cotransporting polypeptide at the postentry step is inhibited by the entry inhibitor Myrcludex B. Emerg Microbes Infect 2018; 7:186. [PMID: 30459339 PMCID: PMC6246608 DOI: 10.1038/s41426-018-0189-8] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2018] [Revised: 09/30/2018] [Accepted: 10/26/2018] [Indexed: 02/07/2023]
Abstract
Sodium taurocholate cotransporting polypeptide (NTCP) is a functional receptor for hepatitis B virus (HBV) entry. However, little is known regarding whether NTCP is involved in regulating the postentry steps of the HBV life cycle. Here, we found that NTCP expression upregulated HBV transcription at the postentry step and that the NTCP-targeting entry inhibitor Myrcludex B (MyrB) effectively suppressed HBV transcription both in an HBV in vitro infection system and in mice hydrodynamically injected with an HBV expression plasmid. Mechanistically, NTCP upregulated HBV transcription via farnesoid X receptor α (FxRα)-mediated activation of the HBV EN2/core promoter at the postentry step in a manner that was dependent on the bile acid (BA)-transport function of NTCP, which was blocked by MyrB. Our findings uncover a novel role for NTCP in the HBV life cycle and provide a reference for the use of novel NTCP-targeting entry inhibitors to suppress HBV infection and replication.
Collapse
Affiliation(s)
- Kaitao Zhao
- State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, 430071, Wuhan, China.,University of Chinese Academy of Sciences, 100049, Beijing, China
| | - Shuhui Liu
- State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, 430071, Wuhan, China.,University of Chinese Academy of Sciences, 100049, Beijing, China
| | - Yingshan Chen
- State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, 430071, Wuhan, China.,University of Chinese Academy of Sciences, 100049, Beijing, China
| | - Yongxuan Yao
- State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, 430071, Wuhan, China.,University of Chinese Academy of Sciences, 100049, Beijing, China
| | - Ming Zhou
- Shenzhen Xenotransplantation Research and Development Center, State and Local Joint Cancer Genome Clinical Application of Key Technology Laboratory, Shenzhen Second People's Hospital, First Affiliated Hospital of Shenzhen University, 518035, Shenzhen, China
| | - Yifei Yuan
- State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, 430071, Wuhan, China.,University of Chinese Academy of Sciences, 100049, Beijing, China
| | - Yun Wang
- State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, 430071, Wuhan, China
| | - Rongjuan Pei
- State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, 430071, Wuhan, China
| | - Jizheng Chen
- State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, 430071, Wuhan, China
| | - Xue Hu
- State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, 430071, Wuhan, China
| | - Yuan Zhou
- State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, 430071, Wuhan, China
| | - He Zhao
- State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, 430071, Wuhan, China
| | - Mengji Lu
- Institute of Virology, University Hospital of Essen, 45147, Essen, Germany
| | - Chunchen Wu
- State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, 430071, Wuhan, China.
| | - Xinwen Chen
- State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, 430071, Wuhan, China. .,University of Chinese Academy of Sciences, 100049, Beijing, China.
| |
Collapse
|
10
|
Eller C, Heydmann L, Colpitts CC, Verrier ER, Schuster C, Baumert TF. The functional role of sodium taurocholate cotransporting polypeptide NTCP in the life cycle of hepatitis B, C and D viruses. Cell Mol Life Sci 2018; 75:3895-3905. [PMID: 30097692 PMCID: PMC7613421 DOI: 10.1007/s00018-018-2892-y] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2018] [Revised: 07/02/2018] [Accepted: 07/25/2018] [Indexed: 12/11/2022]
Abstract
Chronic hepatitis B, C and D virus (HBV, HCV and HDV) infections are a major cause of liver disease and cancer worldwide. Despite employing distinct replication strategies, the three viruses are exclusively hepatotropic, and therefore depend on hepatocyte-specific host factors. The sodium taurocholate co-transporting polypeptide (NTCP), a transmembrane protein highly expressed in human hepatocytes that mediates the transport of bile acids, plays a key role in HBV and HDV entry into hepatocytes. Recently, NTCP has been shown to modulate HCV infection of hepatocytes by regulating innate antiviral immune responses in the liver. Here, we review the current knowledge of the functional role and the molecular and cellular biology of NTCP in the life cycle of the three major hepatotropic viruses, highlight the impact of NTCP as an antiviral target and discuss future avenues of research.
Collapse
Affiliation(s)
- Carla Eller
- Inserm, U1110, Institut de Recherche sur les Maladies Virales et Hépatiques, 3 Rue Koeberlé, 67000, Strasbourg, France
- Université de Strasbourg, 67000, Strasbourg, France
| | - Laura Heydmann
- Inserm, U1110, Institut de Recherche sur les Maladies Virales et Hépatiques, 3 Rue Koeberlé, 67000, Strasbourg, France
- Université de Strasbourg, 67000, Strasbourg, France
| | - Che C Colpitts
- Division of Infection and Immunity, University College London, London, UK
| | - Eloi R Verrier
- Inserm, U1110, Institut de Recherche sur les Maladies Virales et Hépatiques, 3 Rue Koeberlé, 67000, Strasbourg, France
- Université de Strasbourg, 67000, Strasbourg, France
| | - Catherine Schuster
- Inserm, U1110, Institut de Recherche sur les Maladies Virales et Hépatiques, 3 Rue Koeberlé, 67000, Strasbourg, France
- Université de Strasbourg, 67000, Strasbourg, France
| | - Thomas F Baumert
- Inserm, U1110, Institut de Recherche sur les Maladies Virales et Hépatiques, 3 Rue Koeberlé, 67000, Strasbourg, France.
- Université de Strasbourg, 67000, Strasbourg, France.
- Institut Hospitalo-Universitaire, Pôle Hépato-digestif, Nouvel Hôpital Civil, 67000, Strasbourg, France.
| |
Collapse
|
11
|
Ghany MG, Block TM. Disease Pathways and Mechanisms of Potential Drug Targets. Clin Liver Dis (Hoboken) 2018; 12:12-18. [PMID: 30988903 PMCID: PMC6385908 DOI: 10.1002/cld.735] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/30/2018] [Revised: 07/11/2018] [Accepted: 06/20/2018] [Indexed: 02/04/2023] Open
Affiliation(s)
- Marc G. Ghany
- Liver Diseases Branch, National Institute of Diabetes and Digestive and Kidney DiseasesNational Institutes of HealthBethesdaMD
| | - Timothy M. Block
- Hepatitis B Foundation and Baruch S. Blumberg InstituteDoylestownPA
| |
Collapse
|
12
|
Liu C, Xu G, Gao Z, Zhou Z, Guo G, Li D, Jing Z, Sui J, Li W. The p.Ser267Phe variant of sodium taurocholate cotransporting polypeptide (NTCP) supports HBV infection with a low efficiency. Virology 2018; 522:168-176. [PMID: 30032030 DOI: 10.1016/j.virol.2018.07.006] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2018] [Revised: 06/17/2018] [Accepted: 07/06/2018] [Indexed: 12/20/2022]
Abstract
Sodium taurocholate cotransporting polypeptide (NTCP) is a functional receptor for human hepatitis B virus (HBV) and its satellite virus Hepatitis D virus (HDV). Physiologically, NTCP is responsible for the majority of sodium-dependent bile acids uptake by hepatocytes. The p.Ser267Phe (S267F) variant of NTCP is a single nucleotide polymorphism (SNP) previously found to cause substantial loss of ability to support HBV and HDV infection and its taurocholic acid uptake function in vitro. Intriguingly, ten individuals were identified as S267F homozygotes in population studies of chronic hepatitis B (CHB) patients. In this study, we identified new HBV isolates from one homozygous S267F mutation carrier and confirmed new isolates also use wildtype-NTCP as a cellular receptor. Furthermore, we demonstrated S267F variant of NTCP, though inefficient, is still a functional receptor for HBV entry. This study advances our understanding of NTCP-mediated HBV infection.
Collapse
Affiliation(s)
- Chenxuan Liu
- College of Life Sciences, Beijing Normal University, No. 19, XinJieKouWai St., HaiDian District, Beijing 100875, China; National Institute of Biological Sciences, Beijing, 7 Science Park Road ZGC Life Science Park, Beijing 102206, China
| | - Guangwei Xu
- National Institute of Biological Sciences, Beijing, 7 Science Park Road ZGC Life Science Park, Beijing 102206, China
| | - Zhenchao Gao
- National Institute of Biological Sciences, Beijing, 7 Science Park Road ZGC Life Science Park, Beijing 102206, China
| | - Zhongmin Zhou
- College of Life Sciences, Beijing Normal University, No. 19, XinJieKouWai St., HaiDian District, Beijing 100875, China; National Institute of Biological Sciences, Beijing, 7 Science Park Road ZGC Life Science Park, Beijing 102206, China
| | - Guilan Guo
- College of Life Sciences, Beijing Normal University, No. 19, XinJieKouWai St., HaiDian District, Beijing 100875, China; National Institute of Biological Sciences, Beijing, 7 Science Park Road ZGC Life Science Park, Beijing 102206, China
| | - Dan Li
- National Institute of Biological Sciences, Beijing, 7 Science Park Road ZGC Life Science Park, Beijing 102206, China
| | - Zhiyi Jing
- National Institute of Biological Sciences, Beijing, 7 Science Park Road ZGC Life Science Park, Beijing 102206, China
| | - Jianhua Sui
- National Institute of Biological Sciences, Beijing, 7 Science Park Road ZGC Life Science Park, Beijing 102206, China
| | - Wenhui Li
- National Institute of Biological Sciences, Beijing, 7 Science Park Road ZGC Life Science Park, Beijing 102206, China.
| |
Collapse
|
13
|
Abstract
Hepatitis delta virus (HDV) is a defective RNA virus that depends on the presence of hepatitis B virus (HBV) for the creation of new virions and propagation of the infection to hepatocytes. Chronic infection with HDV is usually associated with a worsening of HBV infection, leading more frequently to cirrhosis, increased risk of liver decompensation and hepatocellular carcinoma (HCC) occurrence. In spite of a progressive declining prevalence of both acute and chronic HDV infection observed over several years, mainly due to increased global health policies and mass vaccination against HBV, several European countries have more recently observed stable HDV prevalence mainly due to migrants from non-European countries. Persistent HDV replication has been widely demonstrated as associated with cirrhosis development and, as a consequence, development of liver decompensation and occurrence of HCC. Several treatment options have been attempted with poor results in terms of HDV eradication and improvement of long-term prognosis. A global effort is deemed urgent to enhance the models already existing as well as to learn more about HDV infection and correlated tumourigenesis mechanisms.
Collapse
|
14
|
Müller SF, König A, Döring B, Glebe D, Geyer J. Characterisation of the hepatitis B virus cross-species transmission pattern via Na+/taurocholate co-transporting polypeptides from 11 New World and Old World primate species. PLoS One 2018; 13:e0199200. [PMID: 29912972 PMCID: PMC6005513 DOI: 10.1371/journal.pone.0199200] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2018] [Accepted: 06/04/2018] [Indexed: 12/18/2022] Open
Abstract
The hepatic Na+/taurocholate co-transporting polypeptide (NTCP in man, Ntcp in animals) is the high-affinity receptor for the hepatitis B (HBV) and hepatitis D (HDV) viruses. Species barriers for human HBV/HDV within the order Primates were previously attributed to Ntcp sequence variations that disable virus-receptor interaction. However, only a limited number of primate Ntcps have been analysed so far. In the present study, a total of 11 Ntcps from apes, Old and New World monkeys were cloned and expressed in vitro to characterise their interaction with HBV and HDV. All Ntcps showed intact bile salt transport. Human NTCP as well as the Ntcps from the great apes chimpanzee and orangutan showed transport-competing binding of HBV derived myr-preS1-peptides. In contrast, all six Ntcps from the group of Old World monkeys were insensitive to HBV myr-preS1-peptide binding and HBV/HDV infection. This is basically predetermined by the amino acid arginine at position 158 of all studied Old World monkey Ntcps. An exchange from arginine to glycine (as present in humans and great apes) at this position (R158G) alone was sufficient to achieve full transport-competing HBV myr-preS1-peptide binding and susceptibility for HBV/HDV infection. New World monkey Ntcps showed higher sequence heterogeneity, but in two cases with 158G showed transport-competing HBV myr-preS1-peptide binding, and in one case (Saimiri sciureus) even susceptibility for HBV/HDV infection. In conclusion, amino acid position 158 of NTCP/Ntcp is sufficient to discriminate between the HBV/HDV susceptible group of humans and great apes (158G) and the non-susceptible group of Old World monkeys (158R). In the case of the phylogenetically more distant New World monkey Ntcps amino acid 158 plays a significant, but not exclusive role.
Collapse
Affiliation(s)
- Simon F. Müller
- Institute of Pharmacology and Toxicology, Biomedical Research Center Seltersberg, Justus Liebig University Giessen, Giessen, Germany
| | - Alexander König
- Institute of Medical Virology, Biomedical Research Center Seltersberg, Justus Liebig University Giessen, Giessen, Germany
| | - Barbara Döring
- Institute of Pharmacology and Toxicology, Biomedical Research Center Seltersberg, Justus Liebig University Giessen, Giessen, Germany
| | - Dieter Glebe
- Institute of Medical Virology, Biomedical Research Center Seltersberg, Justus Liebig University Giessen, Giessen, Germany
| | - Joachim Geyer
- Institute of Pharmacology and Toxicology, Biomedical Research Center Seltersberg, Justus Liebig University Giessen, Giessen, Germany
| |
Collapse
|
15
|
Virus entry and its inhibition to prevent and treat hepatitis B and hepatitis D virus infections. Curr Opin Virol 2018; 30:68-79. [DOI: 10.1016/j.coviro.2018.04.004] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2018] [Revised: 04/19/2018] [Accepted: 04/20/2018] [Indexed: 12/13/2022]
|
16
|
Abstract
The tree shrew (Tupaia belangeri) is a promising laboratory animal that possesses a closer genetic relationship to primates than to rodents. In addition, advantages such as small size, easy breeding, and rapid reproduction make the tree shrew an ideal subject for the study of human disease. Numerous tree shrew disease models have been generated in biological and medical studies in recent years. Here we summarize current tree shrew disease models, including models of infectious diseases, cancers, depressive disorders, drug addiction, myopia, metabolic diseases, and immune-related diseases. With the success of tree shrew transgenic technology, this species will be increasingly used in biological and medical studies in the future.
Collapse
Affiliation(s)
- Ji Xiao
- Medical Faculty of Kunming University of Science and Technology, Kunming Yunnan 650500, China; Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences and Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming Yunnan 650223, China
| | - Rong Liu
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences and Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming Yunnan 650223, China
| | - Ce-Shi Chen
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences and Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming Yunnan 650223, China.
| |
Collapse
|
17
|
Sargiacomo C, El-Kehdy H, Dallmeier K, de Kock J, Hernandez-Kelly C, Rogiers V, Ortega A, Neyts J, Sokal E, Najimi M. Upregulation of sodium taurocholate cotransporter polypeptide during hepatogenic differentiation of umbilical cord matrix mesenchymal stem cells facilitates hepatitis B entry. Stem Cell Res Ther 2017; 8:204. [PMID: 28962642 PMCID: PMC5622580 DOI: 10.1186/s13287-017-0656-5] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2017] [Revised: 08/17/2017] [Accepted: 09/01/2017] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND Hepatitis B virus (HBV) carriers worldwide number approximately 240 million people and around 780,000 people die every year from HBV infection. HBV entry and uptake are functionally linked to the presence of the human sodium-taurocholate cotransporting peptide (hNTCP) receptor. Recently, our group demonstrated that human umbilical cord matrix stem cells (UCMSCs) become susceptible to HBV after in-vitro hepatogenic differentiation (D-UCMSCs). METHODS In the present study, we examined the involvement of hNTCP in governing D-UCMSC susceptibility to HBV infection by characterizing the modulation of this transporter expression during hepatogenic differentiation and by appreciating the inhibition of its activity on infection efficacy. RESULTS We show here that in-vitro hepatogenic differentiation upregulated hNTCP mRNA and protein expression as well as its activity in D-UCMSCs. Pre-treatment of D-UCMSCs with taurocholate, a specific NTCP substrate, blocked their infection by HBV which supports the crucial involvement of this transporter in the early steps of the virus entry. CONCLUSION Altogether, our data support the usefulness of D-UCMSCs as a unique human and non-transformed in-vitro model to study the early stages of HBV infection thanks to its ability to endogenously regulate the expression of hNTCP.
Collapse
Affiliation(s)
- Camillo Sargiacomo
- Institute of Experimental and Clinical Research (IREC), Laboratory of Pediatric Hepatology & Cell Therapy, Université Catholique de Louvain, Avenue Mounier, 52, 1200 Brussels, Belgium
| | - Hoda El-Kehdy
- Institute of Experimental and Clinical Research (IREC), Laboratory of Pediatric Hepatology & Cell Therapy, Université Catholique de Louvain, Avenue Mounier, 52, 1200 Brussels, Belgium
| | - Kai Dallmeier
- Rega Institute for Medical Research, Department of Microbiology & Immunology, University of Leuven (KU Leuven), Leuven, Belgium
| | - Joery de Kock
- Faculty of Medicine and Pharmacy, Department of Toxicology, Dermato-Cosmetology and Pharmacognosy, Vrije Universiteit Brussel, Brussels, Belgium
| | - Clara Hernandez-Kelly
- Centro de Investigación y Estudios Avanzados del Instituto Politécnico Nacional (CINVESTAV-IPN),Departamento de Genética y Biología Molecular, México D.F, Mexico
| | - Vera Rogiers
- Faculty of Medicine and Pharmacy, Department of Toxicology, Dermato-Cosmetology and Pharmacognosy, Vrije Universiteit Brussel, Brussels, Belgium
| | - Arturo Ortega
- Centro de Investigación y Estudios Avanzados del Instituto Politécnico Nacional (CINVESTAV-IPN),Departamento de Genética y Biología Molecular, México D.F, Mexico
| | - Johan Neyts
- Rega Institute for Medical Research, Department of Microbiology & Immunology, University of Leuven (KU Leuven), Leuven, Belgium
| | - Etienne Sokal
- Institute of Experimental and Clinical Research (IREC), Laboratory of Pediatric Hepatology & Cell Therapy, Université Catholique de Louvain, Avenue Mounier, 52, 1200 Brussels, Belgium
| | - Mustapha Najimi
- Institute of Experimental and Clinical Research (IREC), Laboratory of Pediatric Hepatology & Cell Therapy, Université Catholique de Louvain, Avenue Mounier, 52, 1200 Brussels, Belgium
| |
Collapse
|
18
|
Rendon JC, Cortes-Mancera F, Restrepo-Gutierrez JC, Hoyos S, Navas MC. Molecular characterization of occult hepatitis B virus infection in patients with end-stage liver disease in Colombia. PLoS One 2017; 12:e0180447. [PMID: 28686707 PMCID: PMC5501523 DOI: 10.1371/journal.pone.0180447] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2017] [Accepted: 06/15/2017] [Indexed: 12/11/2022] Open
Abstract
Background Hepatitis B virus (HBV) occult infection (OBI) is a risk factor to be taken into account in transfusion, hemodialysis and organ transplantation. The aim of this study was to identify and characterize at the molecular level OBI cases in patients with end-stage liver disease. Methods Sixty-six liver samples were obtained from patients with diagnosis of end-stage liver disease submitted to liver transplantation in Medellin (North West, Colombia). Samples obtained from patients who were negative for the surface antigen of HBV (n = 50) were tested for viral DNA detection by nested PCR for ORFs S, C, and X and confirmed by Southern-Blot. OBI cases were analyzed by sequencing the viral genome to determine the genotype and mutations; additionally, viral genome integration events were examined by the Alu-PCR technique. Results In five cases out of 50 patients (10%) the criteria for OBI was confirmed. HBV genotype F (subgenotypes F1 and F3), genotype A and genotype D were characterized in liver samples. Three integration events in chromosomes 5q14.1, 16p13 and 20q12 affecting Receptor-type tyrosine-protein phosphatase T, Ras Protein Specific Guanine Nucleotide Releasing Factor 2, and the zinc finger 263 genes were identified in two OBI cases. Sequence analysis of the viral genome of the 5 OBI cases showed several punctual missense and nonsense mutations affecting ORFs S, P, Core and X. Conclusions This is the first characterization of OBI in patients with end-stage liver disease in Colombia. The OBI cases were identified in patients with HCV infection or cryptogenic cirrhosis. The integration events (5q14.1, 16p13 and 20q12) described in this study have not been previously reported. Further studies are required to validate the role of mutations and integration events in OBI pathogenesis.
Collapse
Affiliation(s)
- Julio Cesar Rendon
- Grupo de Gastrohepatologia, Facultad de Medicina, Universidad de Antioquia, UdeA, Medellin, Colombia
| | - Fabian Cortes-Mancera
- Grupo de Investigación e Innovacion Biomédica GIB, Facultad de Ciencias Exactas y Aplicadas, Instituto Tecnologico Metropolitano (ITM), Medellin, Colombia
| | - Juan Carlos Restrepo-Gutierrez
- Grupo de Gastrohepatologia, Facultad de Medicina, Universidad de Antioquia, UdeA, Medellin, Colombia
- Unidad de Hepatologia y Trasplante Hepatico, Hospital Pablo Tobon Uribe, Medellin, Colombia
| | - Sergio Hoyos
- Grupo de Gastrohepatologia, Facultad de Medicina, Universidad de Antioquia, UdeA, Medellin, Colombia
- Unidad de Hepatologia y Trasplante Hepatico, Hospital Pablo Tobon Uribe, Medellin, Colombia
| | - Maria-Cristina Navas
- Grupo de Gastrohepatologia, Facultad de Medicina, Universidad de Antioquia, UdeA, Medellin, Colombia
- * E-mail:
| |
Collapse
|
19
|
Woodchuck sodium taurocholate cotransporting polypeptide supports low-level hepatitis B and D virus entry. Virology 2017; 505:1-11. [PMID: 28213271 DOI: 10.1016/j.virol.2017.02.006] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2016] [Revised: 02/07/2017] [Accepted: 02/08/2017] [Indexed: 12/15/2022]
Abstract
Sodium taurocholate cotransporting polypeptide (NTCP) is the functional receptor for human hepatitis B virus (HBV) and its satellite hepatitis D virus (HDV). Species barriers to HBV/HDV infection are mainly determined at entry level by variations in the sequences of particular NTCP orthologs. In this study, we sought to determine whether the NTCP ortholog in woodchuck (Marmota monax), woodchuck NTCP (wNTCP) supports viral infection. We found that wNTCP is capable of supporting HBV/HDV infection in HepG2 cells, but to much lower extent than human NTCP (hNTCP), which is about 90% reduction of hNTCP. Comprehensive site-directed mutagenesis mapping of hNTCP and wNTCP revealed that the residue at position 263 is a novel site crucial for viral entry. The important role of site 263 in infection is conserved among NTCP orthologs and may therefore be a potential target for blocking the viral entry.
Collapse
|
20
|
Abstract
Sodium taurocholate cotransporting polypeptide (NTCP) has been identified as a functional receptor for hepatitis B virus (HBV). Expressing human NTCP in human hepatoma HepG2 cells (HepG2-NTCP) renders these cells susceptible for HBV infection. The HepG2-NTCP stably transfected cell line provides a much-needed and easily accessible platform for studying the virus. HepG2-NTCP cells could also be used to identify chemicals targeting key steps of the virus life cycle including HBV covalent closed circular (ccc) DNA, and enable the development of novel antivirals against the infection.Many factors may contribute to the efficiency of HBV infection on HepG2-NTCP cells, with clonal differences among cell line isolates, the source of viral inoculum, and infection medium among the most critical ones. Here, we provide detailed protocols for efficient HBV infection of HepG2-NTCP cells in culture; generation and selection of single cell clones of HepG2-NTCP; production of infectious HBV virion stock through DNA transfection of recombinant plasmid that enables studying primary clinical HBV isolates; and assessing the infection with immunostaining of HBV antigens and Southern blot analysis of HBV cccDNA.
Collapse
Affiliation(s)
- Yinyan Sun
- National Institute of Biological Sciences, Beijing, 102206, China
| | - Yonghe Qi
- National Institute of Biological Sciences, Beijing, 102206, China
| | - Bo Peng
- National Institute of Biological Sciences, Beijing, 102206, China.,Graduate Program in School of Life Sciences, Peking University, Beijing, 100871, China
| | - Wenhui Li
- National Institute of Biological Sciences, Beijing, 102206, China.
| |
Collapse
|
21
|
Qi Y, Gao Z, Xu G, Peng B, Liu C, Yan H, Yao Q, Sun G, Liu Y, Tang D, Song Z, He W, Sun Y, Guo JT, Li W. DNA Polymerase κ Is a Key Cellular Factor for the Formation of Covalently Closed Circular DNA of Hepatitis B Virus. PLoS Pathog 2016; 12:e1005893. [PMID: 27783675 PMCID: PMC5081172 DOI: 10.1371/journal.ppat.1005893] [Citation(s) in RCA: 147] [Impact Index Per Article: 18.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2016] [Accepted: 08/24/2016] [Indexed: 12/11/2022] Open
Abstract
Hepatitis B virus (HBV) infection of hepatocytes begins by binding to its cellular receptor sodium taurocholate cotransporting polypeptide (NTCP), followed by the internalization of viral nucleocapsid into the cytoplasm. The viral relaxed circular (rc) DNA genome in nucleocapsid is transported into the nucleus and converted into covalently closed circular (ccc) DNA to serve as a viral persistence reservoir that is refractory to current antiviral therapies. Host DNA repair enzymes have been speculated to catalyze the conversion of rcDNA to cccDNA, however, the DNA polymerase(s) that fills the gap in the plus strand of rcDNA remains to be determined. Here we conducted targeted genetic screening in combination with chemical inhibition to identify the cellular DNA polymerase(s) responsible for cccDNA formation, and exploited recombinant HBV with capsid coding deficiency which infects HepG2-NTCP cells with similar efficiency of wild-type HBV to assure cccDNA synthesis is exclusively from de novo HBV infection. We found that DNA polymerase κ (POLK), a Y-family DNA polymerase with maximum activity in non-dividing cells, substantially contributes to cccDNA formation during de novo HBV infection. Depleting gene expression of POLK in HepG2-NTCP cells by either siRNA knockdown or CRISPR/Cas9 knockout inhibited the conversion of rcDNA into cccDNA, while the diminished cccDNA formation in, and hence the viral infection of, the knockout cells could be effectively rescued by ectopic expression of POLK. These studies revealed that POLK is a crucial host factor required for cccDNA formation during a de novo HBV infection and suggest that POLK may be a potential target for developing antivirals against HBV. HBV chronically infects 240 million people worldwide. Persistent HBV infection relies on stable maintenance of the nuclear form of viral genome, the covalently closed circular (ccc) DNA. However, the molecular mechanism underlying the conversion of HBV genomic relaxed circular (rc) DNA into cccDNA remains elusive. Our studies reported herein provide unambiguous evidence suggesting that host DNA polymerase κ (POLK) is required for repairing the gap of rcDNA and formation of cccDNA in a de novo HBV infection. POLK is thus a potential therapeutic target for treatment of chronic hepatitis B.
Collapse
Affiliation(s)
- Yonghe Qi
- National Institute of Biological Sciences, Beijing, China
| | - Zhenchao Gao
- National Institute of Biological Sciences, Beijing, China
- Graduate program in School of Life Sciences, Peking University, Beijing, China
| | - Guangwei Xu
- National Institute of Biological Sciences, Beijing, China
| | - Bo Peng
- National Institute of Biological Sciences, Beijing, China
- Graduate program in School of Life Sciences, Peking University, Beijing, China
| | - Chenxuan Liu
- National Institute of Biological Sciences, Beijing, China
- College of Life Sciences Beijing Normal University, Beijing, China
| | - Huan Yan
- National Institute of Biological Sciences, Beijing, China
| | - Qiyan Yao
- National Institute of Biological Sciences, Beijing, China
| | - Guoliang Sun
- National Institute of Biological Sciences, Beijing, China
| | - Yang Liu
- National Institute of Biological Sciences, Beijing, China
- School of Life Science, Tsinghua University, Beijing, China
| | - Dingbin Tang
- National Institute of Biological Sciences, Beijing, China
- Graduate program in School of Life Sciences, Peking University, Beijing, China
| | - Zilin Song
- National Institute of Biological Sciences, Beijing, China
| | - Wenhui He
- National Institute of Biological Sciences, Beijing, China
| | - Yinyan Sun
- National Institute of Biological Sciences, Beijing, China
| | - Ju-Tao Guo
- Baruch S. Blumberg Institute, Doylestown, Pennsylvania, United States of America
| | - Wenhui Li
- National Institute of Biological Sciences, Beijing, China
- * E-mail: (WL)
| |
Collapse
|
22
|
Li W, Urban S. Entry of hepatitis B and hepatitis D virus into hepatocytes: Basic insights and clinical implications. J Hepatol 2016; 64:S32-S40. [PMID: 27084034 PMCID: PMC7114860 DOI: 10.1016/j.jhep.2016.02.011] [Citation(s) in RCA: 84] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/24/2015] [Revised: 02/05/2016] [Accepted: 02/07/2016] [Indexed: 12/30/2022]
Abstract
For almost three decades following the discovery of the human Hepatitis B Virus (HBV) the early events of virus infection (attachment to hepatocytes, specific binding to a receptor on hepatocytes) remained enigmatic. The gradual improvement of tissue culture systems for HBV has enabled the identification of viral determinants for viral infectivity and facilitated the discovery of the human sodium taurocholate co-transporting polypeptide (hNTCP) as a liver specific receptor of HBV and its satellite, the human Hepatitis Delta Virus (HDV). These findings are currently leading basic and clinical research activities in new directions. (1) Stable hNTCP-expressing cell lines have become a valuable platform to study the full HBV replication cycle from its native template, the cccDNA. (2) The suitability of NTCP complemented cell culture systems for high throughput screening approaches will facilitate identification of novel host factors involved in HBV replication (including those that determine the peculiar host specificity of HBV infection) and will enable identification and development of novel drug candidates for improved therapeutics. (3) Since NTCP is a major host-specific restriction factor for HBV and HDV, hNTCP-expressing animals provide the basis for future susceptible in vivo models. (4) The concept obtained with the entry inhibitor Myrcludex B demonstrates that NTCP is a suitable target for clinical interference with viral entry. This will foster further clinical approaches aiming at curative combination therapies.
Collapse
Affiliation(s)
- Wenhui Li
- National Institute of Biological Sciences, Beijing 102206, China.
| | - Stephan Urban
- Department of Infectious Diseases, Molecular Virology, University Hospital Heidelberg, Im Neuenheimer Feld 345, D-69120 Heidelberg, Germany; German Center of Infectious Diseases (DZIF), Heidelberg, Germany.
| |
Collapse
|
23
|
Yang J, Yang Y, Xia M, Wang L, Zhou W, Yang Y, Jiang Y, Wang H, Qian J, Jin L, Wang X. A genetic variant of the NTCP gene is associated with HBV infection status in a Chinese population. BMC Cancer 2016; 16:211. [PMID: 26968990 PMCID: PMC4788942 DOI: 10.1186/s12885-016-2257-6] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2015] [Accepted: 03/08/2016] [Indexed: 12/23/2022] Open
Abstract
Background To investigate whether genetic variants of the HBV receptor gene NTCP are associated with HBV infection in the Han Chinese population. Methods We sequenced the entire 23 kb NTCP gene from 111 HBeAg-positive HBsAg carriers (PSE group), 110 HBeAg-negative HBsAg carriers (PS group), and 110 control subjects. Then, we performed association analyses of suggestively significant SNPs with HBV infection in 1075 controls, 1936 PSs and 639 PSEs. Results In total, 109 rare variants (74 novel) and 38 single nucleotide polymorphisms (SNPs, one novel) were screened. Of the seven non-synonymous rare variants, six were singletons and one was a double hit. All three damaging rare singletons presented exclusively in the PSE group. Of the five SNPs validated in all 3650 subjects, the T allele of rs4646287 was significantly decreased (p = 0.002) in the PS group (10.1 %) and PSE group (8.1 %) compared to the controls (10.9 %) and was decreased to 7.4 % in the PSE hepatocellular carcinoma (HCC) subgroup. Additionally, rs4646287-T was associated with a 0.68-fold (95 % CI = 0.51–0.89, p = 0.006) decreased risk of PSE compared with the controls. The NTCP mRNA level was lower in HCC tissues in “CT + TT” carriers than in “CC” carriers. Conclusions We found a genetic variant (rs4646287) located in intron 1 of NTCP that may be associated with increased risk of HBV infection in Han Chinese. Electronic supplementary material The online version of this article (doi:10.1186/s12885-016-2257-6) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Jingmin Yang
- Epidemiology unit of MOE Key Laboratory of Contemporary Anthropology and State Key Laboratory of Genetic Engineering, School of Life Sciences and Institutes of Biomedical Sciences, Fudan University, 220 Handan Rd., Shanghai, 200433, China.,China Medical City Institute of Health Sciences, 1 Yaocheng Road, Taizhou, Jiangsu, 225300, China
| | - Yuan Yang
- Eastern Hepatobiliary Surgery Hospital, Second Military Medical University, Shanghai, 200438, China.,Department of Health Statistics, Second Military Medical University, Shanghai, 200433, China
| | - Mingying Xia
- Epidemiology unit of MOE Key Laboratory of Contemporary Anthropology and State Key Laboratory of Genetic Engineering, School of Life Sciences and Institutes of Biomedical Sciences, Fudan University, 220 Handan Rd., Shanghai, 200433, China.,China Medical City Institute of Health Sciences, 1 Yaocheng Road, Taizhou, Jiangsu, 225300, China
| | - Lianghui Wang
- Epidemiology unit of MOE Key Laboratory of Contemporary Anthropology and State Key Laboratory of Genetic Engineering, School of Life Sciences and Institutes of Biomedical Sciences, Fudan University, 220 Handan Rd., Shanghai, 200433, China.,China Medical City Institute of Health Sciences, 1 Yaocheng Road, Taizhou, Jiangsu, 225300, China
| | - Weiping Zhou
- Eastern Hepatobiliary Surgery Hospital, Second Military Medical University, Shanghai, 200438, China.,National Innovation Alliance for Hepatitis & Liver Cancer, Shanghai, 200438, China
| | - Yajun Yang
- Epidemiology unit of MOE Key Laboratory of Contemporary Anthropology and State Key Laboratory of Genetic Engineering, School of Life Sciences and Institutes of Biomedical Sciences, Fudan University, 220 Handan Rd., Shanghai, 200433, China.,China Medical City Institute of Health Sciences, 1 Yaocheng Road, Taizhou, Jiangsu, 225300, China
| | - Yueming Jiang
- Epidemiology unit of MOE Key Laboratory of Contemporary Anthropology and State Key Laboratory of Genetic Engineering, School of Life Sciences and Institutes of Biomedical Sciences, Fudan University, 220 Handan Rd., Shanghai, 200433, China.,China Medical City Institute of Health Sciences, 1 Yaocheng Road, Taizhou, Jiangsu, 225300, China
| | - Hongyang Wang
- Eastern Hepatobiliary Surgery Hospital, Second Military Medical University, Shanghai, 200438, China.,Department of Health Statistics, Second Military Medical University, Shanghai, 200433, China.,National Innovation Alliance for Hepatitis & Liver Cancer, Shanghai, 200438, China
| | - Ji Qian
- Epidemiology unit of MOE Key Laboratory of Contemporary Anthropology and State Key Laboratory of Genetic Engineering, School of Life Sciences and Institutes of Biomedical Sciences, Fudan University, 220 Handan Rd., Shanghai, 200433, China. .,China Medical City Institute of Health Sciences, 1 Yaocheng Road, Taizhou, Jiangsu, 225300, China. .,Cancer Institute, Fudan University Shanghai Cancer Center, Shanghai, 200032, China. .,Department of Oncology, Fudan University Shanghai Medical College, Shanghai, 200032, China.
| | - Li Jin
- Epidemiology unit of MOE Key Laboratory of Contemporary Anthropology and State Key Laboratory of Genetic Engineering, School of Life Sciences and Institutes of Biomedical Sciences, Fudan University, 220 Handan Rd., Shanghai, 200433, China. .,China Medical City Institute of Health Sciences, 1 Yaocheng Road, Taizhou, Jiangsu, 225300, China.
| | - Xiaofeng Wang
- Epidemiology unit of MOE Key Laboratory of Contemporary Anthropology and State Key Laboratory of Genetic Engineering, School of Life Sciences and Institutes of Biomedical Sciences, Fudan University, 220 Handan Rd., Shanghai, 200433, China. .,China Medical City Institute of Health Sciences, 1 Yaocheng Road, Taizhou, Jiangsu, 225300, China.
| |
Collapse
|
24
|
Slagle BL, Bouchard MJ. Hepatitis B Virus X and Regulation of Viral Gene Expression. Cold Spring Harb Perspect Med 2016; 6:a021402. [PMID: 26747833 DOI: 10.1101/cshperspect.a021402] [Citation(s) in RCA: 93] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The efficient replication of hepatitis B virus (HBV) requires the HBV regulatory hepatitis B virus X (HBx) protein. The exact contributions of HBx are not fully understood, in part because of the limitations of the assays used for its study. When HBV replication is driven from a plasmid DNA, the contribution of HBx is modest. However, there is an absolute requirement for HBx in assays that recapitulate the infectious virus life cycle. There is much evidence that HBx can contribute directly to HBV replication by acting on viral promoters embedded within protein coding sequences. In addition, HBx may also contribute indirectly by modulating cellular pathways to benefit virus replication. Understanding the mechanism(s) of HBx action during virus replication may provide insight into novel ways to disrupt chronic HBV replication.
Collapse
Affiliation(s)
- Betty L Slagle
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, Texas 77030
| | - Michael J Bouchard
- Department of Biochemistry and Molecular Biology, Drexel University College of Medicine, Philadelphia, Pennsylvania 19102
| |
Collapse
|
25
|
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
|
26
|
Abstract
Hepatitis B virus (HBV) infection affects 240 million people worldwide. A liver-specific bile acid transporter named the sodium taurocholate cotransporting polypeptide (NTCP) has been identified as the cellular receptor for HBV and its satellite, the hepatitis D virus (HDV). NTCP likely acts as a major determinant for the liver tropism and species specificity of HBV and HDV at the entry level. NTCP-mediated HBV entry interferes with bile acid transport in cell cultures and has been linked with alterations in bile acid and cholesterol metabolism in vivo. The human liver carcinoma cell line HepG2, complemented with NTCP, now provides a valuable platform for studying the basic biology of the viruses and developing treatments for HBV infection. This review summarizes critical findings regarding NTCP's role as a viral receptor for HBV and HDV and discusses important questions that remain unanswered.
Collapse
Affiliation(s)
- Wenhui Li
- National Institute of Biological Sciences, Zhongguancun Life Science Park, Beijing 102206, China;
| |
Collapse
|
27
|
Li J, Tong S. From DCPD to NTCP: the long journey towards identifying a functional hepatitis B virus receptor. Clin Mol Hepatol 2015; 21:193-9. [PMID: 26523264 PMCID: PMC4612279 DOI: 10.3350/cmh.2015.21.3.193] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/05/2015] [Accepted: 08/15/2015] [Indexed: 12/13/2022] Open
Abstract
Hepatitis B virus (HBV) is the prototype of hepatotropic DNA viruses (hepadnaviruses) infecting a wide range of human and non-human hosts. Previous studies with duck hepatitis B virus (DHBV) identified duck carboxypeptidase D (dCPD) as a host specific binding partner for full-length large envelope protein, and p120 as a binding partner for several truncated versions of the large envelope protein. p120 is the P protein of duck glycine decarboxylase (dGLDC) with restricted expression in DHBV infectible tissues. Several lines of evidence suggest the importance of dCPD, and especially p120, in productive DHBV infection, although neither dCPD nor p120 cDNA could confer susceptibility to DHBV infection in any cell line. Recently, sodium taurocholate cotransporting polypeptide (NTCP) has been identified as a binding partner for the N-terminus of HBV large envelope protein. Importantly, knock down and reconstitution experiments unequivocally demonstrated that NTCP is both necessary and sufficient for in vitro infection by HBV and hepatitis delta virus (HDV), an RNA virus using HBV envelope proteins for its transmission. What remains unclear is whether NTCP is the major HBV receptor in vivo. The fact that some HBV patients are homozygous with an NTCP mutation known to abolish its receptor function suggests the existence of NTCP-independent pathways of HBV entry. Also, NTCP very likely mediates just one step of the HBV entry process, with additional co-factors for productive HBV infection still to be discovered. NTCP offers a novel therapeutic target for the control of chronic HBV infection.
Collapse
Affiliation(s)
- Jisu Li
- Liver Research Center, Rhode Island Hospital, The Warren Alpert Medical School of Brown University, Providence, USA
| | - Shuping Tong
- Liver Research Center, Rhode Island Hospital, The Warren Alpert Medical School of Brown University, Providence, USA. ; Key lab of Medical Molecular Virology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| |
Collapse
|
28
|
Colpitts CC, Verrier ER, Baumert TF. Targeting Viral Entry for Treatment of Hepatitis B and C Virus Infections. ACS Infect Dis 2015; 1:420-7. [PMID: 27617925 DOI: 10.1021/acsinfecdis.5b00039] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Hepatitis B virus (HBV) and hepatitis C virus (HCV) infections remain major health problems worldwide, with 400-500 million chronically infected people worldwide. Chronic infection results in liver cirrhosis and hepatocellular carcinoma, the second leading cause of cancer death. Current treatments for HBV limit viral replication without efficiently curing infection. HCV treatment has markedly progressed with the licensing of direct-acting antivirals (DAAs) for HCV cure, yet limited access for the majority of patients is a major challenge. Preventative and curative treatment strategies, aimed at novel targets, are needed for both viruses. Viral entry represents one such target, although detailed knowledge of the entry mechanisms is a prerequisite. For HBV, the recent discovery of the NTCP cell entry factor enabled the establishment of an HBV cell culture model and showed that cyclosporin A and Myrcludex B are NTCP-targeting entry inhibitors. Advances in the understanding of HCV entry revealed it to be a complex process involving many factors, offering several antiviral targets. These include viral envelope proteins E1 and E2, virion-associated lipoprotein ApoE, and cellular factors CD81, SRBI, EGFR, claudin-1, occludin, and the cholesterol transporter NPC1L1. Small molecules targeting SR-BI, EGFR, and NPC1L1 have entered clinical trials, whereas other viral- and host-targeted small molecules, peptides, and antibodies show promise in preclinical models. This review summarizes the current understanding of HBV and HCV entry and describes novel antiviral targets and compounds in different stages of clinical development. Overall, proof-of-concept studies indicate that entry inhibitors are a promising class of antivirals to prevent and treat HBV and HCV infections.
Collapse
Affiliation(s)
- Che C. Colpitts
- Inserm, U1110, Institut de Recherche sur les Maladies
Virales et Hépatiques, 67000 Strasbourg, France
- Université de Strasbourg, 67000 Strasbourg, France
| | - Eloi R. Verrier
- Inserm, U1110, Institut de Recherche sur les Maladies
Virales et Hépatiques, 67000 Strasbourg, France
- Université de Strasbourg, 67000 Strasbourg, France
| | - Thomas F. Baumert
- Inserm, U1110, Institut de Recherche sur les Maladies
Virales et Hépatiques, 67000 Strasbourg, France
- Université de Strasbourg, 67000 Strasbourg, France
- Institut Hospitalo-Universitaire,
Pôle Hépato-digestif, Hopitaux Universitaires de Strasbourg, 67000 Strasbourg, France
| |
Collapse
|
29
|
NTCP opens the door for hepatitis B virus infection. Antiviral Res 2015; 121:24-30. [DOI: 10.1016/j.antiviral.2015.06.002] [Citation(s) in RCA: 57] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2015] [Revised: 06/03/2015] [Accepted: 06/08/2015] [Indexed: 02/07/2023]
|
30
|
Baumert TF, Verrier ER, Nassal M, Chung RT, Zeisel MB. Host-targeting agents for treatment of hepatitis B virus infection. Curr Opin Virol 2015; 14:41-6. [PMID: 26262886 DOI: 10.1016/j.coviro.2015.07.009] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2015] [Revised: 07/16/2015] [Accepted: 07/20/2015] [Indexed: 02/07/2023]
Abstract
Hepatitis B virus (HBV) infection is a major cause of chronic liver disease, including liver cirrhosis, liver failure and hepatocellular carcinoma (HCC)-the second leading and fastest rising cause of cancer death world-wide. While de novo infection can be efficiently prevented by vaccination and chronic infection can be controlled using antivirals targeting the viral polymerase, the development of efficient antiviral strategies to eliminate the virus and thus to cure infection remains a key unmet medical need. The recent progress in the development of robust infectious HBV cell culture models now enables the investigation of the full viral life cycle, including a more detailed study of the molecular mechanisms of virus-host interactions responsible for viral persistence. The understanding of these virus-host interactions will be instrumental for the development of curative treatments. Host-dependency factors have recently emerged as promising candidates to treat and prevent infection by various pathogens. This review focuses on the potential of host-targeting agents (HTAs) as novel antivirals to treat and cure HBV infection. These include HTAs that inhibit de novo and re-infection, synthesis and spread of cccDNA as well as development of immune-based approaches eliminating or curing infected hepatocytes, including the eradication of viral cccDNA.
Collapse
Affiliation(s)
- Thomas F Baumert
- Inserm, U1110, Institut de Recherche sur les Maladies Virales et Hépatiques, 67000 Strasbourg, France; Université de Strasbourg, 67000 Strasbourg, France; Institut Hospitalo-Universitaire, Pôle Hépato-digestif, Nouvel Hôpital Civil, 67000 Strasbourg, France; Liver Center and Gastrointestinal Unit, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts 02114, United States.
| | - Eloi R Verrier
- Inserm, U1110, Institut de Recherche sur les Maladies Virales et Hépatiques, 67000 Strasbourg, France; Université de Strasbourg, 67000 Strasbourg, France
| | - Michael Nassal
- Department of Internal Medicine 2/Molecular Biology, University Hospital Freiburg, D-79106 Freiburg, Germany
| | - Raymond T Chung
- Liver Center and Gastrointestinal Unit, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts 02114, United States
| | - Mirjam B Zeisel
- Inserm, U1110, Institut de Recherche sur les Maladies Virales et Hépatiques, 67000 Strasbourg, France; Université de Strasbourg, 67000 Strasbourg, France
| |
Collapse
|
31
|
Progress and Prospects of Anti-HBV Gene Therapy Development. Int J Mol Sci 2015; 16:17589-610. [PMID: 26263978 PMCID: PMC4581210 DOI: 10.3390/ijms160817589] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2015] [Revised: 07/20/2015] [Accepted: 07/22/2015] [Indexed: 12/11/2022] Open
Abstract
Despite the availability of an effective vaccine against hepatitis B virus (HBV), chronic infection with the virus remains a major global health concern. Current drugs against HBV infection are limited by emergence of resistance and rarely achieve complete viral clearance. This has prompted vigorous research on developing better drugs against chronic HBV infection. Advances in understanding the life cycle of HBV and improvements in gene-disabling technologies have been impressive. This has led to development of better HBV infection models and discovery of new drug candidates. Ideally, a regimen against chronic HBV infection should completely eliminate all viral replicative intermediates, especially covalently closed circular DNA (cccDNA). For the past few decades, nucleic acid-based therapy has emerged as an attractive alternative that may result in complete clearance of HBV in infected patients. Several genetic anti-HBV strategies have been developed. The most studied approaches include the use of antisense oligonucleotides, ribozymes, RNA interference effectors and gene editing tools. This review will summarize recent developments and progress made in the use of gene therapy against HBV.
Collapse
|
32
|
Ye L, Yu H, Li C, Hirsch ML, Zhang L, Samulski RJ, Li W, Liu Z. Adeno-Associated Virus Vector Mediated Delivery of the HBV Genome Induces Chronic Hepatitis B Virus Infection and Liver Fibrosis in Mice. PLoS One 2015; 10:e0130052. [PMID: 26075890 PMCID: PMC4468063 DOI: 10.1371/journal.pone.0130052] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2015] [Accepted: 05/15/2015] [Indexed: 01/04/2023] Open
Abstract
Liver cirrhosis and hepatocellular carcinomas are major health problems of chronic hepatitis B virus (HBV) infection. To date, rare model has reproduced liver fibrosis associated with long-term HBV infection which in turn has hindered both the understanding of HBV biology and the development of new treatment options. Here, using adeno-associated virus serotype 8 (AAV8) mediated delivery of a 1.2-kb HBV genome, we successfully generated a chronic HBV infectious mouse model that presents the associated liver fibrosis observed following human infection. After AAV8/HBV1.2 vector administration, mice demonstrated effective HBV replication and transcription which resulted in HBV antigen expression and viremia over 6 months. Although no obvious acute inflammatory response was noted, these mice still developed chronic liver disease and hepatic fibrogenesis as demonstrated by increased ground glass-like hepatocytes, an increasing trend of collagen deposition and upregulated fibrosis markers, including type I collagen, type III collagen, tissue inhibitor of metalloproteinase (TIMP), and transforming growth factor-β1(TGF-β1). Taken together, AAV-mediated HBV gene delivery to the mouse liver, induced HBV persistent infection accompanied by liver fibrosis which can serve as a model for investigating the precise mechanisms underlying liver fibrosis following chronic HBV infection as well as for the potential development of novel therapeutics.
Collapse
MESH Headings
- Animals
- Blotting, Northern
- Blotting, Southern
- Carcinoma, Hepatocellular/genetics
- Carcinoma, Hepatocellular/virology
- Cells, Cultured
- Dependovirus/genetics
- Disease Models, Animal
- Drug Delivery Systems
- Enzyme-Linked Immunosorbent Assay
- Genetic Vectors/administration & dosage
- Genome, Viral
- HEK293 Cells
- Hepatitis B virus/genetics
- Hepatitis B, Chronic/genetics
- Hepatitis B, Chronic/virology
- Humans
- Liver Cirrhosis/genetics
- Liver Cirrhosis/virology
- Liver Neoplasms/genetics
- Liver Neoplasms/virology
- Mice
- Mice, Inbred C57BL
- RNA, Messenger/genetics
- Real-Time Polymerase Chain Reaction
- Reverse Transcriptase Polymerase Chain Reaction
- Viremia/genetics
- Viremia/virology
- Virus Replication
Collapse
Affiliation(s)
- Lei Ye
- Institute of Blood Transfusion, Chinese Academy of Medical Sciences and Peking Union Medical College, Chengdu, China
| | - Haisheng Yu
- Key Laboratory of Immunity and Infection, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
| | - Chengwen Li
- Gene Therapy Center, Department of Pharmacology, University of North Carolina, Chapel Hill, North Carolina, United States of America
| | - Matthew L. Hirsch
- Gene Therapy Center, Department of Pharmacology, University of North Carolina, Chapel Hill, North Carolina, United States of America
| | - Liguo Zhang
- Key Laboratory of Immunity and Infection, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
| | - R. Jude Samulski
- Gene Therapy Center, Department of Pharmacology, University of North Carolina, Chapel Hill, North Carolina, United States of America
| | - Wuping Li
- Institute of Blood Transfusion, Chinese Academy of Medical Sciences and Peking Union Medical College, Chengdu, China
- MOH Key Laboratory of Systems Biology of Pathogens, Institute of Pathogen Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
- * E-mail:
| | - Zhong Liu
- Institute of Blood Transfusion, Chinese Academy of Medical Sciences and Peking Union Medical College, Chengdu, China
| |
Collapse
|
33
|
Yan H, Li W. Sodium taurocholate cotransporting polypeptide acts as a receptor for hepatitis B and D virus. Dig Dis 2015; 33:388-96. [PMID: 26045274 DOI: 10.1159/000371692] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Infection of hepatitis B virus (HBV) remains a major public health problem worldwide. Understanding the viral infection and developing antivirals against HBV have been hampered by the lack of convenient culture systems and animal models for the infection. Sodium taurocholate cotransporting polypeptide (NTCP), a key bile acid transporter expressed in liver, was recently identified as a critical receptor for viral entry of HBV and its satellite virus hepatitis D virus (HDV). This finding enabled a reliable cell culture system for the viruses. Detailed studies have shown that NTCP is the major determinant for the species specificity of HBV and HDV at entry level. NTCP is responsible for most sodium-dependent bile salt uptake in liver. The molecular determinant critical for HBV/HDV infection overlaps with that for bile acids transporting on NTCP. We evaluated bile acids as potential antivirals for HBV and HDV infection, and developed bile acid derivatives that effectively block taurocholate transporting as well as viral infections. The discovery that NTCP acts as a receptor for HBV has opens a new door for future studies towards the ultimate goal of curative treatment of HBV infection.
Collapse
Affiliation(s)
- Huan Yan
- National Institute of Biological Sciences, Beijing, China
| | | |
Collapse
|
34
|
Interaction of human tumor viruses with host cell surface receptors and cell entry. Viruses 2015; 7:2592-617. [PMID: 26008702 PMCID: PMC4452921 DOI: 10.3390/v7052592] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2015] [Accepted: 05/12/2015] [Indexed: 02/06/2023] Open
Abstract
Currently, seven viruses, namely Epstein-Barr virus (EBV), Kaposi's sarcoma-associated herpes virus (KSHV), high-risk human papillomaviruses (HPVs), Merkel cell polyomavirus (MCPyV), hepatitis B virus (HBV), hepatitis C virus (HCV) and human T cell lymphotropic virus type 1 (HTLV-1), have been described to be consistently associated with different types of human cancer. These oncogenic viruses belong to distinct viral families, display diverse cell tropism and cause different malignancies. A key to their pathogenicity is attachment to the host cell and entry in order to replicate and complete their life cycle. Interaction with the host cell during viral entry is characterized by a sequence of events, involving viral envelope and/or capsid molecules as well as cellular entry factors that are critical in target cell recognition, thereby determining cell tropism. Most oncogenic viruses initially attach to cell surface heparan sulfate proteoglycans, followed by conformational change and transfer of the viral particle to secondary high-affinity cell- and virus-specific receptors. This review summarizes the current knowledge of the host cell surface factors and molecular mechanisms underlying oncogenic virus binding and uptake by their cognate host cell(s) with the aim to provide a concise overview of potential target molecules for prevention and/or treatment of oncogenic virus infection.
Collapse
|
35
|
Seeger C, Mason WS. Molecular biology of hepatitis B virus infection. Virology 2015; 479-480:672-86. [PMID: 25759099 PMCID: PMC4424072 DOI: 10.1016/j.virol.2015.02.031] [Citation(s) in RCA: 566] [Impact Index Per Article: 62.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2015] [Revised: 02/09/2015] [Accepted: 02/16/2015] [Indexed: 02/06/2023]
Abstract
Human hepatitis B virus (HBV) is the prototype of a family of small DNA viruses that productively infect hepatocytes, the major cell of the liver, and replicate by reverse transcription of a terminally redundant viral RNA, the pregenome. Upon infection, the circular, partially double-stranded virion DNA is converted in the nucleus to a covalently closed circular DNA (cccDNA) that assembles into a minichromosome, the template for viral mRNA synthesis. Infection of hepatocytes is non-cytopathic. Infection of the liver may be either transient (<6 months) or chronic and lifelong, depending on the ability of the host immune response to clear the infection. Chronic infections can cause immune-mediated liver damage progressing to cirrhosis and hepatocellular carcinoma (HCC). The mechanisms of carcinogenesis are unclear. Antiviral therapies with nucleoside analog inhibitors of viral DNA synthesis delay sequelae, but cannot cure HBV infections due to the persistence of cccDNA in hepatocytes.
Collapse
|
36
|
Yang C, Ruan P, Ou C, Su J, Cao J, Luo C, Tang Y, Wang Q, Qin H, Sun W, Li Y. Chronic hepatitis B virus infection and occurrence of hepatocellular carcinoma in tree shrews (Tupaia belangeri chinensis). Virol J 2015; 12:26. [PMID: 25889678 PMCID: PMC4369070 DOI: 10.1186/s12985-015-0256-x] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2014] [Accepted: 01/31/2015] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Hepatitis B virus (HBV) infection has been believed as a major cause of hepatocellular carcinoma (HCC) for a long time, however, the evidences of which are mostly from clinical and epidemiological investigations while there is no evidence from animal experiments. Tree shrew (Tupaia) is a small animal closely related to primates evolutionarily, with about 8 years of lifespan. Our previous study proved that tree shrews can be chronically HBV-infected after being inoculated neonatally with HBV. The present study reports the further results from the longer-term observation of these animals. METHODS Neonatal tree shrews were inoculated with sera from HBV-infected patient or tree shrew. Their serum samples and liver biopsies were collected periodically for detection of HBV markers as well as for histopathological and immunohistochemical examinations. Group A consisted of six tree shrews with chronic HBV-infection, and group B consisted of nine tree shrews without chronic HBV infection. RESULTS Periodical examinations on serum and liver biopsies of the animals in group A showed the progress of HBV infection, and two cases of HCC occurred at their late stage of life. The courses of HBV infection and the hepatic histopathological and immunohistochemical changes in the tree shrews were similar to those in humans. In contrast, neither HCC nor obvious hepatitis histopathological change was found among the tree shrews in group B. CONCLUSIONS The course of HBV infection and the features of HCC discovered in tree shrews are similar to those of chronically HBV-infected humans. The tree shrew model might be used to investigate the underlying mechanisms favoring susceptibility for chronic HBV infection and disease progression.
Collapse
Affiliation(s)
- Chun Yang
- Department of Experimental Pathology, Guangxi Cancer Institute (Guangxi Tumor Hospital), Nanning, 530021, China.
| | - Ping Ruan
- Department of Pathology, Guangxi Ruikang Hospital, Guangxi University of Chinese Medicine, Nanning, 530011, China.
| | - Chao Ou
- Department of Experimental Pathology, Guangxi Cancer Institute (Guangxi Tumor Hospital), Nanning, 530021, China.
| | - Jianjia Su
- Department of Experimental Pathology, Guangxi Cancer Institute (Guangxi Tumor Hospital), Nanning, 530021, China.
| | - Ji Cao
- Department of Experimental Pathology, Guangxi Cancer Institute (Guangxi Tumor Hospital), Nanning, 530021, China.
| | - Chengpiao Luo
- Department of Experimental Pathology, Guangxi Cancer Institute (Guangxi Tumor Hospital), Nanning, 530021, China.
| | - Yanping Tang
- Department of Experimental Pathology, Guangxi Cancer Institute (Guangxi Tumor Hospital), Nanning, 530021, China.
| | - Qi Wang
- Department of Experimental Pathology, Guangxi Cancer Institute (Guangxi Tumor Hospital), Nanning, 530021, China.
| | - Hong Qin
- Department of Experimental Pathology, Guangxi Cancer Institute (Guangxi Tumor Hospital), Nanning, 530021, China.
| | - Wen Sun
- Department of Experimental Pathology, Guangxi Cancer Institute (Guangxi Tumor Hospital), Nanning, 530021, China.
| | - Yuan Li
- Department of Experimental Pathology, Guangxi Cancer Institute (Guangxi Tumor Hospital), Nanning, 530021, China.
| |
Collapse
|
37
|
Aldabe R, Suárez-Amarán L, Usai C, González-Aseguinolaza G. Animal models of chronic hepatitis delta virus infection host-virus immunologic interactions. Pathogens 2015; 4:46-65. [PMID: 25686091 PMCID: PMC4384072 DOI: 10.3390/pathogens4010046] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2014] [Accepted: 02/05/2015] [Indexed: 02/08/2023] Open
Abstract
Hepatitis delta virus (HDV) is a defective RNA virus that has an absolute requirement for a virus belonging to the hepadnaviridae family like hepatitis B virus (HBV) for its replication and formation of new virions. HDV infection is usually associated with a worsening of HBV-induced liver pathogenesis, which leads to more frequent cirrhosis, increased risk of hepatocellular carcinoma (HCC), and fulminant hepatitis. Importantly, no selective therapies are available for HDV infection. The mainstay of treatment for HDV infection is pegylated interferon alpha; however, response rates to this therapy are poor. A better knowledge of HDV–host cell interaction will help with the identification of novel therapeutic targets, which are urgently needed. Animal models like hepadnavirus-infected chimpanzees or the eastern woodchuck have been of great value for the characterization of HDV chronic infection. Recently, more practical animal models in which to perform a deeper study of host virus interactions and to evaluate new therapeutic strategies have been developed. Therefore, the main focus of this review is to discuss the current knowledge about HDV host interactions obtained from cell culture and animal models.
Collapse
Affiliation(s)
- Rafael Aldabe
- Gene Therapy and Regulation of Gene Expression Program, Centro de Investigación Médica Aplicada (CIMA), Universidad de Navarra (UNAV), Pamplona 31008, Spain.
| | - Lester Suárez-Amarán
- Gene Therapy and Regulation of Gene Expression Program, Centro de Investigación Médica Aplicada (CIMA), Universidad de Navarra (UNAV), Pamplona 31008, Spain
| | - Carla Usai
- Gene Therapy and Regulation of Gene Expression Program, Centro de Investigación Médica Aplicada (CIMA), Universidad de Navarra (UNAV), Pamplona 31008, Spain.
| | - Gloria González-Aseguinolaza
- Gene Therapy and Regulation of Gene Expression Program, Centro de Investigación Médica Aplicada (CIMA), Universidad de Navarra (UNAV), Pamplona 31008, Spain.
| |
Collapse
|
38
|
Therapeutic vaccines in HBV: lessons from HCV. Med Microbiol Immunol 2015; 204:79-86. [PMID: 25573348 PMCID: PMC4305103 DOI: 10.1007/s00430-014-0376-8] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2014] [Accepted: 10/07/2014] [Indexed: 12/15/2022]
Abstract
Currently, millions of people infected with hepatitis B virus (HBV) are committed to decades of treatment with anti-viral therapy to control viral replication. However, new tools for immunotherapy that include both viral vectors and molecular checkpoint inhibitors are now available. This has led to a resurgence of interest in new strategies to develop immunotherapeutic strategies with the aim of inducing HBeAg seroconversion—an end-point that has been associated with a decrease in the rates of disease progression. Ultimately, a true cure will involve the elimination of covalently closed circular DNA which presents a greater challenge for immunotherapy. In this manuscript, I describe the development of immunotherapeutic strategies for HBV that are approaching or currently in clinical studies, and draw on observations of T cell function in natural infection supported by recent animal studies that may lead to additional rational vaccine strategies using checkpoint inhibitors. I also draw on our recent experience in developing potent vaccines for HCV prophylaxis based on simian adenoviral and MVA vectors used in prime–boost strategies in both healthy volunteers and HCV infected patients. I have shown that the induction of T cell immune responses is markedly attenuated when administered to people with persistent HCV viremia. These studies and recently published animal studies using the woodchuck model suggest that potent vaccines based on DNA or adenoviral vectored vaccination represent a rational way forward. However, combining these with drugs to suppress viral replication, alongside checkpoint inhibitors may be required to induce long-term immune control.
Collapse
|
39
|
Hepatitis B virus HBx protein interactions with the ubiquitin proteasome system. Viruses 2014; 6:4683-702. [PMID: 25421893 PMCID: PMC4246244 DOI: 10.3390/v6114683] [Citation(s) in RCA: 54] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2014] [Revised: 11/16/2014] [Accepted: 11/20/2014] [Indexed: 01/04/2023] Open
Abstract
The hepatitis B virus (HBV) causes acute and chronic hepatitis, and the latter is a major risk factor for the development of hepatocellular carcinoma (HCC). HBV encodes a 17-kDa regulatory protein, HBx, which is required for virus replication. Although the precise contribution(s) of HBx to virus replication is unknown, many viruses target cellular pathways to create an environment favorable for virus replication. The ubiquitin proteasome system (UPS) is a major conserved cellular pathway that controls several critical processes in the cell by regulating the levels of proteins involved in cell cycle, DNA repair, innate immunity, and other processes. We summarize here the interactions of HBx with components of the UPS, including the CUL4 adaptor DDB1, the cullin regulatory complex CSN, and the 26S proteasome. Understanding how these protein interactions benefit virus replication remains a challenge due to limited models in which to study HBV replication. However, studies from other viral systems that similarly target the UPS provide insight into possible strategies used by HBV.
Collapse
|
40
|
Su Z, Li Y, Liao Y, Cai B, Chen J, Zhang J, Li L, Ying B, Tao C, Wang L. Association of the gene polymorphisms in sodium taurocholate cotransporting polypeptide with the outcomes of hepatitis B infection in Chinese Han population. INFECTION GENETICS AND EVOLUTION 2014; 27:77-82. [PMID: 25010264 DOI: 10.1016/j.meegid.2014.07.001] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/27/2014] [Revised: 06/24/2014] [Accepted: 07/01/2014] [Indexed: 02/06/2023]
Abstract
OBJECTIVE In recent years, sodium taurocholate cotransporting polypeptide (NTCP) was newly identified as a hepatitis B virus (HBV) receptor, which partly shed light on the reason for HBV hepatotropism and its host specificity. However, the related researches were limited to in vitro or animal experiments. Therefore, this study aimed to investigate the association of NTCP polymorphisms with HBV natural course in humans. METHODS According to their serological and clinical characteristics, 933 Chinese Han individuals were divided into two major groups, 352 viral clearance controls and 581 persistently infected patients. The latter one included 186 hepatocellular carcinoma (HCC) and 395 non-HCC subjects. A total of five single nucleotide polymorphisms (SNPs) were selected from HapMap dataset and genotyped by high resolution melting (HRM) curve method. RESULTS The rs7154439 AA genotype was observed slightly more common in viral clearance group than in persistently infected group [16 (4.5%) subjects vs. 10 (1.7%) subjects. p=0.008, adjusted odds ratio (AOR)=0.33, 95% confidence interval (CI)=0.15-0.75 in a codominant model; and p=0.006, AOR=0.32, 95% CI=0.14-0.72 in a recessive model]. While the rs4646287 AA genotype was observed slightly more frequent in HCC group than in non-HCC group [6 (3.2%) subjects vs. 1 (0.3%) subject. p=0.018, AOR=15.74, 95% CI=1.59-155.54 in a codominant model; and p=0.018, AOR=15.91, 95% CI=1.61-157.01 in a recessive model]. There were no statistically significant differences of allele or haplotype distribution between any two groups. CONCLUSIONS This study suggests that polymorphisms in the NTCP region may be associated with the natural course of HBV infection. The rs7154439 AA genotype was associated with HBV clearance, while the rs4646287 AA genotype was associated with HCC occurrence. However, considering the sample size is relatively small, larger studies, especially through multicenter collaboration will be needed to fully validate the significance of these findings.
Collapse
Affiliation(s)
- Zhenzhen Su
- Department of Laboratory Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan, PR China
| | - Yi Li
- Department of Laboratory Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan, PR China
| | - Yun Liao
- Department of Laboratory Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan, PR China
| | - Bei Cai
- Department of Laboratory Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan, PR China
| | - Jie Chen
- Department of Laboratory Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan, PR China
| | - Junlong Zhang
- Department of Laboratory Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan, PR China
| | - Lixin Li
- Department of Laboratory Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan, PR China
| | - Binwu Ying
- Department of Laboratory Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan, PR China
| | - Chuanmin Tao
- Department of Laboratory Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan, PR China
| | - Lanlan Wang
- Department of Laboratory Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan, PR China.
| |
Collapse
|
41
|
Marimani M, Hean J, Bloom K, Ely A, Arbuthnot P. Recent advances in developing nucleic acid-based HBV therapy. Future Microbiol 2014; 8:1489-504. [PMID: 24199806 DOI: 10.2217/fmb.13.87] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
Chronic HBV infection remains an important public health problem and currently licensed therapies rarely prevent complications of viral persistence. Silencing HBV gene expression using gene therapy, particularly with exogenous activators of RNAi, holds promise for developing an HBV gene therapy. However, immune stimulation, off-targeting effects and inefficient delivery of RNAi activators remain problematic. Several new approaches have recently been employed to address these issues. Chemical modifications to anti-HBV synthetic siRNAs have been investigated and a variety of vectors are being developed for delivery of RNAi effectors. In this article, we review the potential utility of gene therapy for treating HBV infection.
Collapse
Affiliation(s)
- Musa Marimani
- Antiviral Gene Therapy Research Unit, School of Pathology, Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | | | | | | | | |
Collapse
|
42
|
TONG SHUPING, LI JISU. Identification of NTCP as an HBV receptor: the beginning of the end or the end of the beginning? Gastroenterology 2014; 146:902-5. [PMID: 24576732 PMCID: PMC6342000 DOI: 10.1053/j.gastro.2014.02.024] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
See “Hepatitis B and D viruses exploit sodium taurocholate co-transporting polypeptide for species-specific entry into hepatocytes,” by Ni Y, Lempp FA, Mehrle S, et al, on page 1070.
Collapse
Affiliation(s)
- SHUPING TONG
- Liver Research Center, Rhode Island Hospital, Brown University, Providence, Rhode Island,Medical Molecular Virology Laboratory Shanghai, Medical College, Fudan University, Shanghai, China
| | - JISU LI
- Liver Research Center, Rhode Island Hospital, Brown University, Providence, Rhode Island
| |
Collapse
|
43
|
Ni Y, Lempp FA, Mehrle S, Nkongolo S, Kaufman C, Fälth M, Stindt J, Königer C, Nassal M, Kubitz R, Sültmann H, Urban S. Hepatitis B and D viruses exploit sodium taurocholate co-transporting polypeptide for species-specific entry into hepatocytes. Gastroenterology 2014; 146:1070-83. [PMID: 24361467 DOI: 10.1053/j.gastro.2013.12.024] [Citation(s) in RCA: 586] [Impact Index Per Article: 58.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/24/2013] [Revised: 12/04/2013] [Accepted: 12/07/2013] [Indexed: 12/11/2022]
Abstract
BACKGROUND & AIMS Hepatitis B and D viruses (HBV and HDV) are human pathogens with restricted host ranges and high selectivity for hepatocytes; the HBV L-envelope protein interacts specifically with a receptor on these cells. We aimed to identify this receptor and analyze whether it is the recently described sodium-taurocholate co-transporter polypeptide (NTCP), encoded by the SLC10A1 gene. METHODS To identify receptor candidates, we compared gene expression patterns between differentiated HepaRG cells, which express the receptor, and naïve cells, which do not. Receptor candidates were evaluated by small hairpin RNA silencing in HepaRG cells; the ability of receptor expression to confer binding and infection were tested in transduced hepatoma cell lines. We used interspecies domain swapping to identify motifs for receptor-mediated host discrimination of HBV and HDV binding and infection. RESULTS Bioinformatic analyses of comparative expression arrays confirmed that NTCP, which was previously identified through a biochemical approach is a bona fide receptor for HBV and HDV. NTCPs from rat, mouse, and human bound Myrcludex B, a peptide ligand derived from the HBV L-protein. Myrcludex B blocked NTCP transport of bile salts; small hairpin RNA-mediated knockdown of NTCP in HepaRG cells prevented their infection by HBV or HDV. Expression of human but not mouse NTCP in HepG2 and HuH7 cells conferred a limited cell-type-related and virus-dependent susceptibility to infection; these limitations were overcome when cells were cultured with dimethyl sulfoxide. We identified 2 short-sequence motifs in human NTCP that were required for species-specific binding and infection by HBV and HDV. CONCLUSIONS Human NTCP is a specific receptor for HBV and HDV. NTCP-expressing cell lines can be efficiently infected with these viruses, and might be used in basic research and high-throughput screening studies. Mapping of motifs in NTCPs have increased our understanding of the species specificities of HBV and HDV, and could lead to small animal models for studies of viral infection and replication.
Collapse
Affiliation(s)
- Yi Ni
- Department of Infectious Diseases, Molecular Virology, University Hospital Heidelberg, Heidelberg, Germany
| | - Florian A Lempp
- Department of Infectious Diseases, Molecular Virology, University Hospital Heidelberg, Heidelberg, Germany
| | - Stefan Mehrle
- Department of Infectious Diseases, Molecular Virology, University Hospital Heidelberg, Heidelberg, Germany
| | - Shirin Nkongolo
- Department of Infectious Diseases, Molecular Virology, University Hospital Heidelberg, Heidelberg, Germany
| | - Christina Kaufman
- Department of Infectious Diseases, Molecular Virology, University Hospital Heidelberg, Heidelberg, Germany
| | - Maria Fälth
- German Cancer Research Center and National Center for Tumor Diseases, Unit Cancer Genome Research, Heidelberg, Germany
| | - Jan Stindt
- Clinic for Gastroenterology, Hepatology and Infectiology, University Hospital Düsseldorf, Düsseldorf, Germany
| | - Christian Königer
- Department of Internal Medicine II, University Hospital Freiburg, Freiburg, Germany
| | - Michael Nassal
- Department of Internal Medicine II, University Hospital Freiburg, Freiburg, Germany
| | - Ralf Kubitz
- Clinic for Gastroenterology, Hepatology and Infectiology, University Hospital Düsseldorf, Düsseldorf, Germany
| | - Holger Sültmann
- German Cancer Research Center and National Center for Tumor Diseases, Unit Cancer Genome Research, Heidelberg, Germany
| | - Stephan Urban
- Department of Infectious Diseases, Molecular Virology, University Hospital Heidelberg, Heidelberg, Germany.
| |
Collapse
|
44
|
Watashi K, Urban S, Li W, Wakita T. NTCP and beyond: opening the door to unveil hepatitis B virus entry. Int J Mol Sci 2014; 15:2892-905. [PMID: 24557582 PMCID: PMC3958888 DOI: 10.3390/ijms15022892] [Citation(s) in RCA: 102] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2014] [Revised: 02/13/2014] [Accepted: 02/14/2014] [Indexed: 12/19/2022] Open
Abstract
Chronic hepatitis B virus (HBV) infection, affecting approximately 240 million people worldwide, is a major public health problem that elevates the risk of developing liver cirrhosis and hepatocellular carcinoma. Given that current anti-HBV drugs are limited to interferon-based regimens and nucleos(t)ide analogs, the development of new anti-HBV agents is urgently needed. The viral entry process is generally an attractive target implicated in antiviral strategies. Using primary cells from humans and Tupaia belangeri, as well as HepaRG cells, important determinants of viral entry have been achieved. Recently, sodium taurocholate cotransporting polypeptide (NTCP) was identified as an HBV entry receptor and enabled the establishment of a susceptible cell line that can efficiently support HBV infection. This finding will allow a deeper understanding of the requirements for efficient HBV infection, including the elucidation of the molecular entry mechanism. In addition, pharmacological studies suggest that NTCP is able to serve as a therapeutic target. This article summarizes our current knowledge on the mechanisms of HBV entry and the role of NTCP in this process.
Collapse
Affiliation(s)
- Koichi Watashi
- Department of Virology II, National Institute of Infectious Diseases, 1-23-1 Toyama, Shinjuku-ku, 162-8640 Tokyo, Japan.
| | - Stephan Urban
- Department of Infectious Diseases, Molecular Virology, University Hospital Heidelberg, Im Neuenheimer Feld 345, D-69120 Heidelberg, Germany.
| | - Wenhui Li
- National Institute of Biological Sciences, No.7 Science Park Road, ZGC Life Science Park, Changping, 102206 Beijing, China.
| | - Takaji Wakita
- Department of Virology II, National Institute of Infectious Diseases, 1-23-1 Toyama, Shinjuku-ku, 162-8640 Tokyo, Japan.
| |
Collapse
|
45
|
Baumert TF, Meredith L, Ni Y, Felmlee DJ, McKeating JA, Urban S. Entry of hepatitis B and C viruses - recent progress and future impact. Curr Opin Virol 2014; 4:58-65. [PMID: 24418809 DOI: 10.1016/j.coviro.2013.12.002] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2013] [Revised: 11/28/2013] [Accepted: 12/02/2013] [Indexed: 02/07/2023]
Abstract
Chronic hepatitis B and C virus infections are major causes of liver disease and hepatocellular carcinoma worldwide. Although both viruses infect hepatocytes, the molecular virology and cellular biology of their respective replication cycles differ. Viral entry is the first step of the life cycle and recent developments in functional genomic and proteomic methodologies have increased our understanding of the entry pathways for these two important human pathogens. In this review we provide a comparative analysis of the internalization routes for these viruses and highlight differences and how they impact the viral life cycle, immune responses and development of antivirals.
Collapse
Affiliation(s)
- Thomas F Baumert
- Inserm U1110, University of Strasbourg and Center for Liver and Digestive Diseases, Strasbourg University Hospitals, 3 Rue Koeberlé, F-67000 Strasbourg, France
| | - Luke Meredith
- Centre for Human Virology, NIHR Centre for Liver Disease, University of Birmingham, Vincent Drive, Birmingham B15 2TT, UK
| | - Yi Ni
- Department of Infectious Diseases, Molecular Virology, University Hospital Heidelberg, ImNeuenheimer Feld 345, D-69120 Heidelberg, Germany
| | - Daniel J Felmlee
- Inserm U1110, University of Strasbourg and Center for Liver and Digestive Diseases, Strasbourg University Hospitals, 3 Rue Koeberlé, F-67000 Strasbourg, France
| | - Jane A McKeating
- Centre for Human Virology, NIHR Centre for Liver Disease, University of Birmingham, Vincent Drive, Birmingham B15 2TT, UK
| | - Stephan Urban
- Department of Infectious Diseases, Molecular Virology, University Hospital Heidelberg, ImNeuenheimer Feld 345, D-69120 Heidelberg, Germany.
| |
Collapse
|
46
|
Viral entry of hepatitis B and D viruses and bile salts transportation share common molecular determinants on sodium taurocholate cotransporting polypeptide. J Virol 2014; 88:3273-84. [PMID: 24390325 DOI: 10.1128/jvi.03478-13] [Citation(s) in RCA: 182] [Impact Index Per Article: 18.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
UNLABELLED The liver bile acids transporter sodium taurocholate cotransporting polypeptide (NTCP) is responsible for the majority of sodium-dependent bile salts uptake by hepatocytes. NTCP also functions as a cellular receptor for viral entry of hepatitis B virus (HBV) and hepatitis D virus (HDV) through a specific interaction between NTCP and the pre-S1 domain of HBV large envelope protein. However, it remains unknown if these two functions of NTCP are independent or if they interfere with each other. Here we show that binding of the pre-S1 domain to human NTCP blocks taurocholate uptake by the receptor; conversely, some bile acid substrates of NTCP inhibit HBV and HDV entry. Mutations of NTCP residues critical for bile salts binding severely impair viral infection by HDV and HBV; to a lesser extent, the residues important for sodium binding also inhibit viral infection. The mutation S267F, corresponding to a single nucleotide polymorphism (SNP) found in about 9% of the East Asian population, renders NTCP without either taurocholate transporting activity or the ability to support HBV or HDV infection in cell culture. These results demonstrate that molecular determinants critical for HBV and HDV entry overlap with that for bile salts uptake by NTCP, indicating that viral infection may interfere with the normal function of NTCP, and bile acids and their derivatives hold the potential for further development into antiviral drugs. IMPORTANCE Human hepatitis B virus (HBV) and its satellite virus, hepatitis D virus (HDV), are important human pathogens. Available therapeutics against HBV are limited, and there is no drug that is clinically available for HDV infection. A liver bile acids transporter (sodium taurocholate cotransporting polypeptide [NTCP]) critical for maintaining homeostasis of bile acids serves as a functional receptor for HBV and HDV. We report here that the NTCP-binding lipopeptide that originates from the first 47 amino acids of the pre-S1 domain of the HBV L protein blocks taurocholate transport. Some bile salts dose dependently inhibit HBV and HDV infection mediated by NTCP; molecular determinants of NTCP critical for HBV and HDV entry overlap with that for bile acids transport. This work advances our understanding of NTCP-mediated HBV and HDV infection in relation to NTCP's physiological function. Our results also suggest that bile acids or their derivatives hold potential for development into novel drugs against HBV and HDV infection.
Collapse
|
47
|
Lin S, Yan H, Li L, Yang M, Peng B, Chen S, Li W, Chen PR. Site-Specific Engineering of Chemical Functionalities on the Surface of Live Hepatitis D Virus. Angew Chem Int Ed Engl 2013; 52:13970-4. [DOI: 10.1002/anie.201305787] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2013] [Revised: 09/25/2013] [Indexed: 12/11/2022]
|
48
|
Lin S, Yan H, Li L, Yang M, Peng B, Chen S, Li W, Chen PR. Site-Specific Engineering of Chemical Functionalities on the Surface of Live Hepatitis D Virus. Angew Chem Int Ed Engl 2013. [DOI: 10.1002/ange.201305787] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
49
|
Xu G, Gao Z, He W, Ma Y, Feng X, Cai T, Lu F, Liu L, Li W. microRNA expression in hepatitis B virus infected primary treeshrew hepatocytes and the independence of intracellular miR-122 level for de novo HBV infection in culture. Virology 2013; 448:247-54. [PMID: 24314655 DOI: 10.1016/j.virol.2013.10.016] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2013] [Revised: 08/02/2013] [Accepted: 10/08/2013] [Indexed: 12/17/2022]
Abstract
Infection of Hepatitis B virus (HBV) in hepatocytes has been known to be controlled by multiple cellular factors, while the relationship of the infection and liver microRNAs remains obscure. In this study, a miRNA database, containing 168 unique mature miRNA members from primary hepatocytes of a primate-like animal, northern treeshrew (Tupaia belangeri) that is the only species susceptible for HBV infection other than human and chimpanzee, was established. The relative level of a liver predominant microRNA, miR-122, was markedly increased upon HBV infection of the primary tupaia hepatocyte (PTH). However, introducing neither miR-122 nor its antagonist anti-miR-122 into PTHs, or, HepG2-NTCP that is HepG2 cells with the newly identified receptor sodium taurocholate cotransporting polypeptide (NTCP) did not alter the viral infection on these cells. These data suggest that de novo HBV infection of cultured hepatocytes does not depend on the expression level of intracellular miR-122 of the target cells.
Collapse
Affiliation(s)
- Guangwei Xu
- School of Basic Medical Sciences, Peking Union Medical College & Chinese Academy of Medical Sciences, 5 Dongdan Santiao, Beijing 100005, China; National Institute of Biological Sciences, Beijing, 7 Science Park Road, Zhongguancun Life Science Park, Changping, Beijing 102206, China
| | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Molecular determinants of hepatitis B and D virus entry restriction in mouse sodium taurocholate cotransporting polypeptide. J Virol 2013; 87:7977-91. [PMID: 23678176 DOI: 10.1128/jvi.03540-12] [Citation(s) in RCA: 149] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Human hepatitis B virus (HBV) and its satellite virus, hepatitis D virus (HDV), primarily infect humans, chimpanzees, or tree shrews (Tupaia belangeri). Viral infections in other species are known to be mainly restricted at the entry level since viral replication can be achieved in the cells by transfection of the viral genome. Sodium taurocholate cotransporting polypeptide (NTCP) is a functional receptor for HBV and HDV, and amino acids 157 to 165 of NTCP are critical for viral entry and likely limit viral infection of macaques. However, the molecular determinants for viral entry restriction in mouse NTCP (mNTCP) remain unclear. In this study, mNTCP was found to be unable to support either HBV or HDV infection, although it can bind to pre-S1 of HBV L protein and is functional in transporting substrate taurocholate; comprehensive swapping and point mutations of human NTCP (hNTCP) and mNTCP revealed molecular determinants restricting mNTCP for viral entry of HBV and HDV. Remarkably, when mNTCP residues 84 to 87 were substituted by human counterparts, mNTCP can effectively support viral infections. In addition, a number of cell lines, regardless of their species or tissue origin, supported HDV infection when transfected with hNTCP or mNTCP with residues 84 to 87 replaced by human counterparts, highlighting the central role of NTCP for viral infections mediated by HBV envelope proteins. These studies advance our understanding of NTCP-mediated viral entry of HBV and HDV and have important implications for developing the mouse model for their infections.
Collapse
|