1
|
Wang J, Zheng X, Peng Q, Zhang X, Qin Z. Eph receptors: the bridge linking host and virus. Cell Mol Life Sci 2020; 77:2355-2365. [PMID: 31893311 PMCID: PMC7275029 DOI: 10.1007/s00018-019-03409-6] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2019] [Revised: 11/17/2019] [Accepted: 12/02/2019] [Indexed: 02/06/2023]
Abstract
Eph (erythropoietin-producing hepatoma) receptors and Ephrin ligands constitute the largest subfamily of receptor tyrosine kinase (RTK), which were first discovered in tumors. Heretofore, Eph protein has been shown to be involved in various tumor biological behaviors including proliferation and progression. The occurrence of specific types of tumor is closely related to the virus infection. Virus entry is a complex process characterized by a series of events. The entry into target cells is an essential step for virus to cause diseases, which requires the fusion of the viral envelope and host cellular membrane mediated by viral glycoproteins and cellular receptors. Integrin molecules are well known as entry receptors for most herpes viruses. However, in recent years, Eph receptors and their Ephrin ligands have been reported to be involved in virus infections. The main mechanism may be the interaction between Eph receptors and conserved viral surface glycoprotein, such as the gH/gL or gB protein of the herpesviridae. This review focuses on the relationship between Eph receptor family and virus infection that summarize the processes of viruses such as EBV, KSHV, HCV, RRV, etc., infecting target cells through Eph receptors and activating its downstream signaling pathways resulting in malignancies. Finally, we discussed the perspectives to block virus infection, prevention, and treatment of viral-related tumors via Eph receptor family.
Collapse
Affiliation(s)
- Jia Wang
- Department of Immunology, Changzhi Medical College, Changzhi, 046000, Shanxi, China
| | - Xiang Zheng
- Department of Pathology, Affiliated Hospital of Guilin Medical University, Guilin, 541001, Guangxi, China
| | - Qiu Peng
- School of Basic Medical Science, Cancer Research Institute, Central South University, Changsha, 410008, Hunan, China
| | - Xuemei Zhang
- Department of Pathology, Affiliated Hospital of Guilin Medical University, Guilin, 541001, Guangxi, China.
| | - Zailong Qin
- Laboratory of Genetics and Metabolism, Maternal and Child Health Hospital of Guangxi Zhuang Autonomous Region, Guangxi Birth Defects Research And Prevention Institute, Nanning, 530003, Guangxi, China.
| |
Collapse
|
2
|
Hepatitis C Virus Entry into Macrophages/Monocytes Mainly Depends on the Phagocytosis of Macrophages. Dig Dis Sci 2019; 64:1226-1237. [PMID: 30535782 DOI: 10.1007/s10620-018-5401-0] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/13/2018] [Accepted: 11/27/2018] [Indexed: 12/11/2022]
Abstract
BACKGROUND Hepatitis C virus (HCV) has been classified as a strictly hepatotropic pathogen for a long time, and hepatocytes are target cells for HCV infection. More and more studies showed non-liver cells supported HCV entry and replication, such as macrophages. The mechanisms of HCV entry into macrophages are still not clear. AIMS This study aims to determine the way of HCV entry into macrophages. METHODS Cell culture-derived infectious HCV particles (HCVcc) were prepared using Huh7 cells transfected with HCV RNA. CD81-knockdown cells were obtained through siRNA transfection. HCV RNA levels were determined by RT-qPCR. Flow cytometry analyses were used to determine cell surface levels of CD11b, CD68, and CD81. ELISA and western blotting were performed to quantify the protein levels of IL-1β, IL-6, and TNF-α. Phagocytic ability was determined by neutral red uptake assay. RESULTS CD81 knockdown could not inhibit HCVcc entry into macrophages. The entry of HCV into macrophages could not be blocked by pooled IgG from chronic hepatitis C patient's sera. Macrophages derived from THP-1 cells displayed stronger phagocytic capacity, which also swallowed more HCV RNA. Treatment of macrophages with endocytic inhibitor, methyl-β-cyclodextrin, decreased the internalization of HCV. HCV uptake by macrophages was related to the reorganization of F-actin cytoskeleton and PI3Ks activation. HCV infection significantly increased the expression of IL1β and IL6 in macrophages and promoted apoptosis of macrophages. CONCLUSIONS HCV entry into macrophages mainly depends on phagocytosis of macrophages.
Collapse
|
3
|
Mulot M, Monsion B, Boissinot S, Rastegar M, Meyer S, Bochet N, Brault V. Transmission of Turnip yellows virus by Myzus persicae Is Reduced by Feeding Aphids on Double-Stranded RNA Targeting the Ephrin Receptor Protein. Front Microbiol 2018; 9:457. [PMID: 29593696 PMCID: PMC5859162 DOI: 10.3389/fmicb.2018.00457] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2017] [Accepted: 02/27/2018] [Indexed: 11/13/2022] Open
Abstract
Aphid-transmitted plant viruses are a threat for major crops causing massive economic loss worldwide. Members in the Luteoviridae family are transmitted by aphids in a circulative and non-replicative mode. Virions are acquired by aphids when ingesting sap from infected plants and are transported through the gut and the accessory salivary gland (ASG) cells by a transcytosis mechanism relying on virus-specific receptors largely unknown. Once released into the salivary canal, virions are inoculated to plants, together with saliva, during a subsequent feeding. In this paper, we bring in vivo evidence that the membrane-bound Ephrin receptor (Eph) is a novel aphid protein involved in the transmission of the Turnip yellows virus (TuYV, Polerovirus genus, Luteoviridae family) by Myzus persicae. The minor capsid protein of TuYV, essential for aphid transmission, was able to bind the external domain of Eph in yeast. Feeding M. persicae on in planta- or in vitro-synthesized dsRNA targeting Eph-mRNA (dsRNAEph) did not affect aphid feeding behavior but reduced accumulation of TuYV genomes in the aphid's body. Consequently, TuYV transmission efficiency by the dsRNAEph-treated aphids was reproducibly inhibited and we brought evidence that Eph is likely involved in intestinal uptake of the virion. The inhibition of virus uptake after dsRNAEph acquisition was also observed for two other poleroviruses transmitted by M. persicae, suggesting a broader role of Eph in polerovirus transmission. Finally, dsRNAEph acquisition by aphids did not affect nymph production. These results pave the way toward an ecologically safe alternative of insecticide treatments that are used to lower aphid populations and reduce polerovirus damages.
Collapse
Affiliation(s)
- Michaël Mulot
- SVQV, Université de Strasbourg, Institut National de la Recherche Agronomique, Colmar, France
| | - Baptiste Monsion
- SVQV, Université de Strasbourg, Institut National de la Recherche Agronomique, Colmar, France
| | - Sylvaine Boissinot
- SVQV, Université de Strasbourg, Institut National de la Recherche Agronomique, Colmar, France
| | - Maryam Rastegar
- SVQV, Université de Strasbourg, Institut National de la Recherche Agronomique, Colmar, France.,Department of Plant Protection, Shiraz University, Shiraz, Iran
| | - Sophie Meyer
- SVQV, Université de Strasbourg, Institut National de la Recherche Agronomique, Colmar, France
| | - Nicole Bochet
- SVQV, Université de Strasbourg, Institut National de la Recherche Agronomique, Colmar, France
| | - Véronique Brault
- SVQV, Université de Strasbourg, Institut National de la Recherche Agronomique, Colmar, France
| |
Collapse
|
4
|
Morozov VA, Lagaye S. Hepatitis C virus: Morphogenesis, infection and therapy. World J Hepatol 2018; 10:186-212. [PMID: 29527256 PMCID: PMC5838439 DOI: 10.4254/wjh.v10.i2.186] [Citation(s) in RCA: 87] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/04/2017] [Revised: 01/11/2018] [Accepted: 02/07/2018] [Indexed: 02/06/2023] Open
Abstract
Hepatitis C virus (HCV) is a major cause of liver diseases including liver cirrhosis and hepatocellular carcinoma. Approximately 3% of the world population is infected with HCV. Thus, HCV infection is considered a public healthy challenge. It is worth mentioning, that the HCV prevalence is dependent on the countries with infection rates around 20% in high endemic countries. The review summarizes recent data on HCV molecular biology, the physiopathology of infection (immune-mediated liver damage, liver fibrosis and lipid metabolism), virus diagnostic and treatment. In addition, currently available in vitro, ex vivo and animal models to study the virus life cycle, virus pathogenesis and therapy are described. Understanding of both host and viral factors may in the future lead to creation of new approaches in generation of an efficient therapeutic vaccine.
Collapse
Affiliation(s)
- Vladimir Alexei Morozov
- Center for HIV and Retrovirology, Department of Infectious Diseases, Robert Koch Institute, Berlin 13353, Germany
| | - Sylvie Lagaye
- Department of Immunology, Institut Pasteur, INSERM U1223, Paris 75015, France
| |
Collapse
|
5
|
Chang CC, Hsu HJ, Yen JH, Lo SY, Liou JW. A Sequence in the loop domain of hepatitis C virus E2 protein identified in silico as crucial for the selective binding to human CD81. PLoS One 2017; 12:e0177383. [PMID: 28481946 PMCID: PMC5421814 DOI: 10.1371/journal.pone.0177383] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2017] [Accepted: 04/26/2017] [Indexed: 12/15/2022] Open
Abstract
Hepatitis C virus (HCV) is a species-specific pathogenic virus that infects only humans and chimpanzees. Previous studies have indicated that interactions between the HCV E2 protein and CD81 on host cells are required for HCV infection. To determine the crucial factors for species-specific interactions at the molecular level, this study employed in silico molecular docking involving molecular dynamic simulations of the binding of HCV E2 onto human and rat CD81s. In vitro experiments including surface plasmon resonance measurements and cellular binding assays were applied for simple validations of the in silico results. The in silico studies identified two binding regions on the HCV E2 loop domain, namely E2-site1 and E2-site2, as being crucial for the interactions with CD81s, with the E2-site2 as the determinant factor for human-specific binding. Free energy calculations indicated that the E2/CD81 binding process might follow a two-step model involving (i) the electrostatic interaction-driven initial binding of human-specific E2-site2, followed by (ii) changes in the E2 orientation to facilitate the hydrophobic and van der Waals interaction-driven binding of E2-site1. The sequence of the human-specific, stronger-binding E2-site2 could serve as a candidate template for the future development of HCV-inhibiting peptide drugs.
Collapse
Affiliation(s)
- Chun-Chun Chang
- Institute of Medical Sciences, Tzu Chi University, Hualien, Taiwan.,Department of Laboratory Medicine, Tzu Chi Medical Center, Hualien, Taiwan
| | - Hao-Jen Hsu
- Department of Life Sciences, Tzu Chi University, Hualien, Taiwan
| | - Jui-Hung Yen
- Department of Molecular Biology and Human Genetics, Tzu Chi University, Hualien, Taiwan
| | - Shih-Yen Lo
- Department of Laboratory Medicine and Biotechnology, Tzu Chi University, Hualien, Taiwan
| | - Je-Wen Liou
- Institute of Medical Sciences, Tzu Chi University, Hualien, Taiwan.,Department of Biochemistry, School of Medicine, Tzu Chi University, Hualien, Taiwan
| |
Collapse
|
6
|
Functional Analysis of Hepatitis C Virus (HCV) Envelope Protein E1 Using a trans-Complementation System Reveals a Dual Role of a Putative Fusion Peptide of E1 in both HCV Entry and Morphogenesis. J Virol 2017; 91:JVI.02468-16. [PMID: 28100619 DOI: 10.1128/jvi.02468-16] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2016] [Accepted: 01/12/2017] [Indexed: 02/07/2023] Open
Abstract
Hepatitis C virus (HCV) is an enveloped RNA virus belonging to the Flaviviridae family. It infects mainly human hepatocytes and causes chronic liver diseases, including cirrhosis and cancer. HCV encodes two envelope proteins, E1 and E2, that form a heterodimer and mediate virus entry. While E2 has been extensively studied, less has been done so for E1, and its role in the HCV life cycle still needs to be elucidated. Here we developed a new cell culture model for HCV infection based on the trans-complementation of E1. Virus production of the HCV genome lacking the E1-encoding sequence can be efficiently rescued by the ectopic expression of E1 in trans The resulting virus, designated HCVΔE1, can propagate in packaging cells expressing E1 but results in only single-cycle infection in naive cells. By using the HCVΔE1 system, we explored the role of a putative fusion peptide (FP) of E1 in HCV infection. Interestingly, we found that the FP not only contributes to HCV entry, as previously reported, but also may be involved in virus morphogenesis. Finally, we identified amino acid residues in FP that are critical for biological functions of E1. In summary, our work not only provides a new cell culture model for studying HCV but also provides some insights into understanding the role of E1 in the HCV life cycle.IMPORTANCE Hepatitis C virus (HCV), an enveloped RNA virus, encodes two envelope proteins, E1 and E2, that form a heterodimeric complex to mediate virus entry. Compared to E2, the biological functions of E1 in the virus life cycle are not adequately investigated. Here we developed a new cell culture model for single-cycle HCV infection based on the trans-complementation of E1. The HCV genome lacking the E1-encoding sequence can be efficiently rescued for virus production by the ectopic expression of E1 in trans This new model renders a unique system to dissect functional domains and motifs in E1. Using this system, we found that a putative fusion peptide in E1 is a multifunctional structural element contributing to both HCV entry and morphogenesis. Our work has provided a new cell culture model to study HCV and provides insights into understanding the biological roles of E1 in the HCV life cycle.
Collapse
|
7
|
Bose M, Mullick R, Das S, Das S, Karande AA. Combination of neutralizing monoclonal antibodies against Hepatitis C virus E2 protein effectively blocks virus infection. Virus Res 2016; 224:46-57. [PMID: 27574733 DOI: 10.1016/j.virusres.2016.08.010] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2016] [Revised: 08/23/2016] [Accepted: 08/25/2016] [Indexed: 01/17/2023]
Abstract
Hepatitis C virus (HCV) represents a major global health threat. The envelope glycoproteins, E1-E2 of HCV play an important role in infection by binding to hepatocyte surface receptors leading to viral entry. Several regions on the E1-E2 are conserved for maintaining structural stability, despite the high mutation rate of HCV. Identification of antigenic determinants in these domains would aid in the development of anti-virals. The present study was aimed to delineate neutralizing epitopes by generating monoclonal antibodies (mAbs) to envelope proteins that can block virus binding and entry. Using HCV-like particles (HCV-LPs) corresponding to genotype 3a (prevalent in India), we obtained three mAbs specific for the E2 protein that significantly inhibited virus binding to hepatoma cells. Using overlapping protein fragments and peptides of the E2 protein, the epitopes corresponding to the mAbs were delineated. MAbs H6D3 and A10F2 recognise sequential linear epitopes, whereas, mAb E3D8 recognises a discontinuous epitope. The epitope of mAb E3D8 overlaps with the CD81 receptor-binding site and that of mAb A10F2 with the hypervariable region 2 of the E2 protein. The epitopes corresponding to these mAbs are distinct and unique. A combination of these antibodies significantly inhibited HCV binding and entry in both HCV pseudoparticle (in vitro) and HCV cell culture (ex vivo) system compared to the mAbs alone (P<0.0001). In conclusion, our findings support the potential of employing a cocktail of neutralizing mAbs in the management of HCV infection.
Collapse
Affiliation(s)
- Mihika Bose
- Department of Biochemistry, Indian Institute of Science, Bangalore 560 012, India
| | - Ranajoy Mullick
- Department of Microbiology and Cell Biology, Indian Institute of Science, Bangalore 560 012, India
| | - Soma Das
- Department of Biochemistry, Indian Institute of Science, Bangalore 560 012, India
| | - Saumitra Das
- Department of Microbiology and Cell Biology, Indian Institute of Science, Bangalore 560 012, India
| | - Anjali A Karande
- Department of Biochemistry, Indian Institute of Science, Bangalore 560 012, India.
| |
Collapse
|
8
|
Tang L, Marcell L, Kottilil S. Systemic manifestations of hepatitis C infection. Infect Agent Cancer 2016; 11:29. [PMID: 27222662 PMCID: PMC4878040 DOI: 10.1186/s13027-016-0076-7] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2015] [Accepted: 05/10/2016] [Indexed: 12/19/2022] Open
Abstract
Chronic hepatitis C (HCV) is a common infection affecting 185 million people worldwide. The most common manifestation of chronic HCV is progressive liver fibrosis, cirrhosis, liver failure and hepatocellular carcinoma. However, several systemic manifestations of HCV have been recognized and reported in the literature. The purpose of this review is to assimilate published literature based on evidence to categorize these extrahepatic manifestations with the likelihood of a causal association with HCV. Exciting recent developments have resulted in simple all oral interferon-free highly effective therapy for HCV. However, this treatment is also expensive and less accessible to most affected individuals as treatment recommendations are based on stage of liver fibrosis. Expanding the scope of HCV therapy to those with extrahepatic manifestations beyond what is currently recommended will significantly reduce the morbidity and mortality in this aging population.
Collapse
Affiliation(s)
- Lydia Tang
- Division of Clinical Care and Research, Institute of Human Virology, University of Maryland School of Medicine, 725 W Lombard St, Room S222, Baltimore, MD USA
| | - Lauren Marcell
- Division of Clinical Care and Research, Institute of Human Virology, University of Maryland School of Medicine, 725 W Lombard St, Room S222, Baltimore, MD USA
| | - Shyam Kottilil
- Division of Clinical Care and Research, Institute of Human Virology, University of Maryland School of Medicine, 725 W Lombard St, Room S222, Baltimore, MD USA
| |
Collapse
|
9
|
Mueller JL, King LY, Johnson KB, Gao T, Nephew LD, Kothari D, Simpson MA, Zheng H, Wei L, Corey KE, Misdraji J, Lee JH, Lin MV, Gogela NA, Fuchs BC, Tanabe KK, Gordon FD, Curry MP, Chung RT. Impact of EGF, IL28B, and PNPLA3 polymorphisms on the outcome of allograft hepatitis C: a multicenter study. Clin Transplant 2016; 30:452-460. [PMID: 26854475 PMCID: PMC4868041 DOI: 10.1111/ctr.12710] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/01/2016] [Indexed: 01/03/2023]
Abstract
Hepatitis C virus (HCV) infection is accelerated following liver transplantation (LT). Single nucleotide polymorphisms (SNPs) near the epidermal growth factor (EGF) (rs4444903), IL28B (rs12979860), and PNPLA3 (rs738409) loci are associated with treatment response, fibrosis, and hepatocellular carcinoma in non-transplant hepatitis C, but allograft population data are limited. We sought to determine the role of these SNPs in 264 patients with HCV who underwent LT between 1990 and 2008. Genotypes were determined from donor wedge/allograft biopsies and recipient explants. Cox proportional hazards model was used to assess time to cirrhosis, liver-related death, and retransplantation, adjusting for donor age and sustained virological response (SVR). Over a median follow-up of 6.3 yr, a trend toward increased progression to graft cirrhosis was observed among recipients of an EGF non-AA vs. AA donor liver (adjusted HR 2.01; 95% CI 0.93-4.34; p = 0.08). No other genotypes predicted cirrhosis development or graft survival. The CC IL28B variant in both recipients and donors was associated with increased rate of SVR (R-CC/D-CC 8/12[67%], R-non-CC/D-CC or R-CC/D-non-CC 23/52[44%], R-non-CC/D-non-CC 12/45[27%], p linear trend = 0.009). Recipient EGF, IL28B, and PNPLA3, and donor IL28B and PNPLA3 genotypes do not predict adverse outcomes in HCV LT recipients. A potential association exists between donor EGF genotype and cirrhosis.
Collapse
Affiliation(s)
- Jessica L. Mueller
- Division of Gastroenterology, Department of Medicine, Massachusetts General Hospital, Boston, MA
- Harvard Medical School, Boston, MA
| | - Lindsay Y. King
- Division of Gastroenterology, Department of Medicine, Massachusetts General Hospital, Boston, MA
- Harvard Medical School, Boston, MA
| | - Kara B. Johnson
- Division of Gastroenterology, Department of Medicine, Massachusetts General Hospital, Boston, MA
- Harvard Medical School, Boston, MA
| | - Tian Gao
- Division of Gastroenterology, Department of Medicine, Boston Medical Center, Boston, MA
| | - Lauren D. Nephew
- Division of Gastroenterology, Department of Medicine, University of Pennsylvania Health System, Philadelphia, PA
| | - Darshan Kothari
- Harvard Medical School, Boston, MA
- Division of Gastroenterology, Department of Medicine, Beth Israel Deaconness Medical Center, Boston
| | - Mary Ann Simpson
- Division of Gastroenterology, Department of Medicine, Lahey Hospital & Medical Center, Burlington
| | - Hui Zheng
- Biostatistics Center, Massachusetts General Hospital, Boston, MA, USA
| | - Lan Wei
- Division of Surgical Oncology, Department of Surgery, Massachusetts General Hospital, Boston, MA, USA
| | - Kathleen E. Corey
- Division of Gastroenterology, Department of Medicine, Massachusetts General Hospital, Boston, MA
- Harvard Medical School, Boston, MA
| | - Joseph Misdraji
- Department of Pathology, Massachusetts General Hospital, Boston, MA, USA
| | - Joon Hyoek Lee
- Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea
| | - M. Valerie Lin
- Division of Gastroenterology, Department of Medicine, Massachusetts General Hospital, Boston, MA
- Harvard Medical School, Boston, MA
| | - Neliswa A. Gogela
- Division of Gastroenterology, Department of Medicine, Massachusetts General Hospital, Boston, MA
- Harvard Medical School, Boston, MA
| | - Bryan C. Fuchs
- Harvard Medical School, Boston, MA
- Division of Surgical Oncology, Department of Surgery, Massachusetts General Hospital, Boston, MA, USA
| | - Kenneth K. Tanabe
- Harvard Medical School, Boston, MA
- Division of Surgical Oncology, Department of Surgery, Massachusetts General Hospital, Boston, MA, USA
| | - Fredric D. Gordon
- Harvard Medical School, Boston, MA
- Division of Gastroenterology, Department of Medicine, Lahey Hospital & Medical Center, Burlington
| | - Michael P. Curry
- Harvard Medical School, Boston, MA
- Division of Gastroenterology, Department of Medicine, Beth Israel Deaconness Medical Center, Boston
| | - Raymond T. Chung
- Division of Gastroenterology, Department of Medicine, Massachusetts General Hospital, Boston, MA
- Harvard Medical School, Boston, MA
| |
Collapse
|
10
|
Abstract
Tight junctions (TJs) are highly specialized membrane domains involved in many important cellular processes such as the regulation of the passage of ions and macromolecules across the paracellular space and the establishment of cell polarity in epithelial cells. Over the past few years there has been increasing evidence that different components of the TJs can be hijacked by viruses in order to complete their infectious cycle. Viruses from at least nine different families of DNA and RNA viruses have been reported to use TJ proteins in their benefit. For example, TJ proteins such as JAM-A or some members of the claudin family of proteins are used by members of the Reoviridae family and hepatitis C virus as receptors or co-receptors during their entry into their host cells. Reovirus, in addition, takes advantage of the TJ protein Junction Adhesion Molecule-A (JAM-A) to achieve its hematogenous dissemination. Some other viruses are capable of regulating the expression or the localization of TJ proteins to induce cell transformation or to improve the efficiency of their exit process. This review encompasses the importance of TJs for viral entry, replication, dissemination, and egress, and makes a clear statement of the importance of studying these proteins to gain a better understanding of the replication strategies used by viruses that infect epithelial and/or endothelial cells.
Collapse
|
11
|
Simon TG, Butt AA. Lipid dysregulation in hepatitis C virus, and impact of statin therapy upon clinical outcomes. World J Gastroenterol 2015; 21:8293-8303. [PMID: 26217081 PMCID: PMC4507099 DOI: 10.3748/wjg.v21.i27.8293] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/13/2015] [Revised: 04/17/2015] [Accepted: 06/10/2015] [Indexed: 02/06/2023] Open
Abstract
The hepatitis C virus (HCV) is one of the most common causes of chronic liver disease and the leading indication for liver transplantation worldwide. Every aspect of the HCV life cycle is closely tied to human lipid metabolism. The virus circulates as a lipid-rich particle, utilizing lipoprotein cell receptors to gain entry into the hepatocyte. It has also been shown to upregulate lipid biosynthesis and impair lipid degradation, resulting in significant intracellular lipid accumulation and circulating hypocholesterolemia. Patients with chronic hepatitis C (CHC) are at increased risk of hepatic steatosis, fibrosis, and cardiovascular disease including accelerated atherosclerosis. HMG CoA Reductase inhibitors, or statins, have been shown to play an important role in the modulation of hepatic steatosis and fibrosis, and recent attention has focused upon their potential therapeutic role in CHC. This article reviews the hepatitis C viral life cycle as it impacts host lipoproteins and lipid metabolism. It then describes the pathogenesis of HCV-related hepatic steatosis, hypocholesterolemia and atherosclerosis, and finally describes the promising anti-viral and anti-fibrotic effects of statins, for the treatment of CHC.
Collapse
|
12
|
Zhou Y, Zhang Y, Moorman JP, Yao ZQ, Jia ZS. Viral (hepatitis C virus, hepatitis B virus, HIV) persistence and immune homeostasis. Immunology 2014; 143:319-30. [PMID: 24965611 DOI: 10.1111/imm.12349] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2014] [Revised: 06/18/2014] [Accepted: 06/19/2014] [Indexed: 12/14/2022] Open
Abstract
Immune homeostasis is a host characteristic that maintains biological balance within a host. Humans have evolved many host defence mechanisms that ensure the survival of individuals upon encountering a pathogenic infection, with recovery or persistence from a viral infection being determined by both viral factors and host immunity. Chronic viral infections, such as hepatitis B virus, hepatitis C virus and HIV, often result in chronic fluctuating viraemia in the face of host cellular and humoral immune responses, which are dysregulated by multi-faceted mechanisms that are incompletely understood. This review attempts to illuminate the mechanisms involved in this process, focusing on immune homeostasis in the setting of persistent viral infection from the aspects of host defence mechanism, including interferon-stimulated genes, apolipoprotein B mRNA editing enzyme catalytic polypeptide 3 (APOBEC3), autophagy and interactions of various immune cells, cytokines and regulatory molecules.
Collapse
Affiliation(s)
- Yun Zhou
- Department of Infectious Diseases, Tangdu Hospital, The Fourth Military Medical University, Xi'an, China; Center for Inflammation, Infectious Diseases, and Immunity, Quillen College of Medicine, East Tennessee State University, Johnson City, TN, USA
| | | | | | | | | |
Collapse
|
13
|
Redwan EM, EL-Fakharany EM, Uversky VN, Linjawi MH. Screening the anti infectivity potentials of native N- and C-lobes derived from the camel lactoferrin against hepatitis C virus. BMC COMPLEMENTARY AND ALTERNATIVE MEDICINE 2014; 14:219. [PMID: 24993815 PMCID: PMC4086701 DOI: 10.1186/1472-6882-14-219] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/02/2014] [Accepted: 06/30/2014] [Indexed: 02/06/2023]
Abstract
BACKGROUND Hepatitis C virus (HCV) infection represents a worldwide health threat that still needs efficient protective vaccine and/or effective drug. The traditional medicine, such as camel milk, is heavily used by the large sector of HCV patients to control the infection due to the high cost of the available standard therapy. Camel milk contains lactoferrin, which plays an important and multifunctional role in innate immunity and specific host defense against microbial infection. Continuing the analysis of the effectiveness of camel lactoferrin against HCV, the current study aimed to separate and purify the native N- and C-lobes from the proteolytically cleaved camel lactoferrin (cLF) and to compare their in vitro activities against the HCV infection in Huh7.5 cells in order to determine the most active domain. METHODS Lactoferrin and its digested N- and C-lobes were purified by Mono S 5/50 GL column and Superdex 200 5/150 column. The purified proteins were assessed through three venues: 1. To inhibit intracellular replication, HCV infected cells were treated with the proteins at different concentrations and time intervals; 2. The proteins were directly incubated with the viral particles (neutralization) and then such neutralized viruses were used to infect cells; 3. The cells were protected with proteins before exposure to the virus. The antiviral potentials of the cLf and its lobes were determined using three techniques: 1. RT-nested PCR, 2. Real-time PCR, and 3. Flow cytometry. RESULTS N- and C-lobes were purified in two consecutive steps; using Mono-S and Superdex 200 columns. The molecular mass of N- and C-lobes was about 40 kDa. cLF and its lobes could prevent HCV entry into Huh 7.5 cells with activity reached 100% through direct interaction with the virus. The inhibition of intracellular viral replication by N-lobe is 2-fold and 3-fold more effective than that of the cLF and C-lobe, respectively. CONCLUSION Generated native N- and C-lobes from camel lactoferrin demonstrated a range of noticeably different potentials against HCV cellular infectivity. The anti-HCV activities were sorted as N-lobe > cLf > C-lobe.
Collapse
Affiliation(s)
- Elrashdy M Redwan
- Biological Science Department, Faculty of Science, King Abdulaziz University, P.O. Box 80203, Jeddah 21589, Kingdom of Saudi Arabia
- Therapeutic and Protective Proteins Laboratory, Protein Research Department, Genetic Engineering and Biotechnology Research Institute, City for Scientific Research and Technology Applications, New Borg EL-Arab, Alexandria 21394, Egypt
| | - Esmail M EL-Fakharany
- Therapeutic and Protective Proteins Laboratory, Protein Research Department, Genetic Engineering and Biotechnology Research Institute, City for Scientific Research and Technology Applications, New Borg EL-Arab, Alexandria 21394, Egypt
| | - Vladimir N Uversky
- Biological Science Department, Faculty of Science, King Abdulaziz University, P.O. Box 80203, Jeddah 21589, Kingdom of Saudi Arabia
- Department of Molecular Medicine, USF Health Byrd Alzheimer’s Research Institute, Morsani College of Medicine, University of South Florida, Tampa, FL 33612, USA
| | - Mustafa H Linjawi
- College of Applied Medical Sciences, King Abdulaziz University, P.O. Box 80203, Jeddah 21589, Kingdom of Saudi Arabia
| |
Collapse
|
14
|
Deng A, Chen C, Ishizaka Y, Chen X, Sun B, Yang R. Human immunodeficiency virus type 1 Vpr increases hepatitis C virus RNA replication in cell culture. Virus Res 2014; 184:93-102. [PMID: 24589706 DOI: 10.1016/j.virusres.2014.02.017] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2013] [Revised: 02/17/2014] [Accepted: 02/18/2014] [Indexed: 01/02/2023]
Abstract
Human immunodeficiency virus (HIV) coinfection with hepatitis C virus (HCV) is associated with an increased HCV RNA level, as well as a more rapid progression to cirrhosis and end-stage liver disease. However, the mechanism underlying this effect is largely unknown. Here, we investigated the role of HIV-1 Vpr in HCV infection and clearly demonstrated that Vpr increased the replication of both the infectious HCV full-length genome and the subgenomic replicon. We also demonstrated that Vpr increased HCV infection by enhancing RNA replication but not viral entry or translation. Further, we showed that Vpr could partially overcome the anti-HCV effect of PEG-IFN. Our findings not only partially explain the clinical observation that patients coinfected with HIV and HCV have higher levels of HCV RNA and viral load than HCV mono-infected patients but also provide important information for HCV treatment in HIV/HCV coinfected patients.
Collapse
Affiliation(s)
- Amei Deng
- Research Group of HIV Molecular Epidemiology and Virology, Center for Emerging Infectious Disease, The State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, Hubei 430071, PR China
| | - Chao Chen
- Research Group of HIV Molecular Epidemiology and Virology, Center for Emerging Infectious Disease, The State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, Hubei 430071, PR China
| | - Yukihito Ishizaka
- Department of Intractable Diseases, National Center for Global Health and Medicine, 1-21-1 Toyama, Shinjuku-ku, Tokyo 162-8655, Japan
| | - Xinwen Chen
- Research Group of HIV Molecular Epidemiology and Virology, Center for Emerging Infectious Disease, The State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, Hubei 430071, PR China
| | - Binlian Sun
- Research Group of HIV Molecular Epidemiology and Virology, Center for Emerging Infectious Disease, The State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, Hubei 430071, PR China.
| | - Rongge Yang
- Research Group of HIV Molecular Epidemiology and Virology, Center for Emerging Infectious Disease, The State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, Hubei 430071, PR China.
| |
Collapse
|
15
|
Potisopon S, Priet S, Selisko B, Canard B. Comparison of dengue virus and HCV: from impact on global health to their RNA-dependent RNA polymerases. Future Virol 2014. [DOI: 10.2217/fvl.13.121] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
ABSTRACT: Upon the discovery of HCV, dengue virus (DENV) and other flaviviruses have served as models to unravel the biology and mechanisms at play during HCV replication. HCV research has rapidly become a well-established field. Recently, several specific anti-HCV antiviral drugs have been discovered and approved for use in the clinic. Now, the strong emergence of DENV in the world and the associated increasing burden is casting light back to dengue virology and anti-dengue drug discovery. HCV polymerase (NS5B) is a prime target in antiviral therapies, and the analogous DENV polymerase (NS5) is also becoming one. Although both enzymes share common fold and function to some extent, a significant amount of unique structural and functional features have to be clearly delineated to efficiently translate drug design potential between these two essential enzymes.
Collapse
Affiliation(s)
- Supanee Potisopon
- AFMB Laboratoire d’Architecture et Fonction des Macromolécules Biologiques, Aix-Marseille Université & CNRS, UMR 7257, Parc Scientifique et Technologique de Luminy, Case 932, 163 Avenue de Luminy, 13288 Marseille Cedex 09, France
| | - Stéphane Priet
- AFMB Laboratoire d’Architecture et Fonction des Macromolécules Biologiques, Aix-Marseille Université & CNRS, UMR 7257, Parc Scientifique et Technologique de Luminy, Case 932, 163 Avenue de Luminy, 13288 Marseille Cedex 09, France
| | - Barbara Selisko
- AFMB Laboratoire d’Architecture et Fonction des Macromolécules Biologiques, Aix-Marseille Université & CNRS, UMR 7257, Parc Scientifique et Technologique de Luminy, Case 932, 163 Avenue de Luminy, 13288 Marseille Cedex 09, France
| | - Bruno Canard
- AFMB Laboratoire d’Architecture et Fonction des Macromolécules Biologiques, Aix-Marseille Université & CNRS, UMR 7257, Parc Scientifique et Technologique de Luminy, Case 932, 163 Avenue de Luminy, 13288 Marseille Cedex 09, France
| |
Collapse
|
16
|
Nepomnyashchikh GI, Bakarev MA, Nepomnyashchikh DL, Yudanov AV, Kapustina VI, Migus'kova EI, Postnikova OA, Vinogradova EV, Nokhrina ZV, Savchenko SA. Role of lipid infiltration of hepatocytes in the morphogenesis of chronic hepatitis C. Bull Exp Biol Med 2013; 156:281-4. [PMID: 24319768 DOI: 10.1007/s10517-013-2330-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Immunohistochemical and PCR analysis of the structure of lipid-containing hepatocytes and replication of chronic hepatitis C viral particles revealed no positive correlation between the presence of HCV RNA in blood specimens, number of infected hepatocytes (by expression of HCV NS3Ag) with clinical biochemical parameters and intensity of structural changes in biopsy specimens. Small-vesicular subcytolemmal lipid infiltration of hepatocytes was detected, presumably associated with hepatitis C virus replication phase. Characteristic subcytolemmal location of the lipid droplets can reflect their involvement in virus transport from the cell and indicate a virus-induced nature of lipid metabolism disorders.
Collapse
Affiliation(s)
- G I Nepomnyashchikh
- Research Institute of Regional Pathology and Pathomorphology, Siberian Division of the Russian Academy of Medical Sciences, Novosibirsk, Russia.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
17
|
Macarthur KL, Smolic R, Smolic MV, Wu CH, Wu GY. Update on the Development of Anti-Viral Agents Against Hepatitis C. J Clin Transl Hepatol 2013; 1:9-21. [PMID: 26357602 PMCID: PMC4521270 DOI: 10.14218/jcth.2013.007xx] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/12/2013] [Revised: 05/14/2013] [Accepted: 05/14/2013] [Indexed: 02/06/2023] Open
Abstract
Hepatitis C virus (HCV) infects nearly 170 million people worldwide and causes chronic hepatitis, cirrhosis, and hepatocellular carcinoma. The search for a drug regimen that maximizes efficacy and minimizes side effects is quickly evolving. This review will discuss a wide range of drug targets currently in all phases of development for the treatment of HCV. Direct data from agents in phase III/IV clinical trials will be presented, along with reported side-effect profiles. The mechanism of action of all treatments and resistance issues are highlighted. Special attention is given to available trial data supporting interferon-free treatment regimens. HCV has become an increasingly important public health concern, and it is important for physicians to stay up to date on the rapidly growing novel therapeutic options.
Collapse
Affiliation(s)
| | | | | | - Catherine H. Wu
- Department of Medicine, Division of Gastroenterology-Hepatology, University of Connecticut Health Center, Farmington, CT, USA
| | - George Y. Wu
- Department of Medicine, Division of Gastroenterology-Hepatology, University of Connecticut Health Center, Farmington, CT, USA
| |
Collapse
|
18
|
Zhang S, Kodys K, Babcock GJ, Szabo G. CD81/CD9 tetraspanins aid plasmacytoid dendritic cells in recognition of hepatitis C virus-infected cells and induction of interferon-alpha. Hepatology 2013; 58:940-9. [PMID: 22577054 PMCID: PMC4511847 DOI: 10.1002/hep.25827] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/29/2012] [Accepted: 05/01/2012] [Indexed: 12/11/2022]
Abstract
UNLABELLED Recognition of hepatitis C virus (HCV)-infected hepatocyes and interferon (IFN) induction are critical in antiviral immune response. We hypothesized that cell-cell contact between plasmacytoid dendritic cells (pDCs) and HCV-infected cells was required for IFN-α induction through the involvement of cell-surface molecules. Coculture of human peripheral blood mononuclear cells (PBMCs) with genotype 1a full-length (FL) HCV genomic replicon cells or genotype 2a Japanese fulminant hepatitis type 1 (JFH-1) virus-infected hepatoma cells (JFH-1), and not with uninfected hepatoma cells (Huh7.5), induced IFN-α production. Depletion of pDCs from PBMCs attenuated IFN-α release, and purified pDCs produced high levels of IFN-α after coculture with FL replicons or JFH-1-infected cells. IFN-α induction by HCV-containing hepatoma cells required viral replication, direct cell-cell contact with pDCs, and receptor-mediated endocytosis. We determined that the tetraspanin proteins, CD81 and CD9, and not other HCV entry receptors, were required for IFN-α induction in pDCs by HCV-infected hepatoma cells. Disruption of cholesterol-rich membrane microdomains, the localization site of CD81, or inhibition of the CD81 downstream molecule, Rac GTPase, inhibited IFN-α production. IFN-α induction involved HCV RNA and Toll-like receptor (TLR) 7. IFN-α production by HCV-infected hepatoma cells was decreased in pDCs from HCV-infected patients, compared to healthy controls. We found that preexposure of healthy PBMCs to HCV viral particles attenuated IFN-α induction by HCV-infected hepatoma cells or TLR ligands, and this inhibitory effect could be prevented by an anti-HCV envelope glycoprotein 2-blocking antibody. CONCLUSION Our novel data show that recognition of HCV-infected hepatoma cells by pDCs involves CD81- and CD9-associated membrane microdomains and induces potent IFN-α production.
Collapse
|
19
|
Abstract
Hepatitis C virus (HCV) is a hepatotropic virus and a major cause of chronic hepatitis and liver disease worldwide. Initial interactions between HCV virions and hepatocytes are required for productive viral infection and initiation of the viral life cycle. Furthermore, HCV entry contributes to the tissue tropism and species specificity of this virus. The elucidation of these interactions is critical, not only to understand the pathogenesis of HCV infection, but also to design efficient antiviral strategies and vaccines. This review summarizes our current knowledge of the host factors required for the HCV-host interactions during HCV binding and entry, our understanding of the molecular mechanisms underlying HCV entry into target cells, and the relevance of HCV entry for the pathogenesis of liver disease, antiviral therapy, and vaccine development.
Collapse
|
20
|
Inhibition of hepatitis C virus infection by DNA aptamer against envelope protein. Antimicrob Agents Chemother 2013; 57:4937-44. [PMID: 23877701 DOI: 10.1128/aac.00897-13] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Hepatitis C virus (HCV) envelope protein (E1E2) is essential for virus binding to host cells. Aptamers have been demonstrated to have strong promising applications in drug development. In the current study, a cDNA fragment encoding the entire E1E2 gene of HCV was cloned. E1E2 protein was expressed and purified. Aptamers for E1E2 were selected by the method of selective evolution of ligands by exponential enrichment (SELEX), and the antiviral actions of the aptamers were examined. The mechanism of their antiviral activity was investigated. The data show that selected aptamers for E1E2 specifically recognize the recombinant E1E2 protein and E1E2 protein from HCV-infected cells. CD81 protein blocks the binding of aptamer E1E2-6 to E1E2 protein. Aptamers against E1E2 inhibit HCV infection in an infectious cell culture system although they have no effect on HCV replication in a replicon cell line. Beta interferon (IFN-β) and IFN-stimulated genes (ISGs) are not induced in virus-infected hepatocytes with aptamer treatment, suggesting that E1E2-specific aptamers do not induce innate immunity. E2 protein is essential for the inhibition of HCV infection by aptamer E1E2-6, and the aptamer binding sites are located in E2. Q412R within E1E2 is the major resistance substitution identified. The data indicate that an aptamer against E1E2 exerts its antiviral effects through inhibition of virus binding to host cells. Aptamers against E1E2 can be used with envelope protein to understand the mechanisms of HCV entry and fusion. The aptamers may hold promise for development as therapeutic drugs for hepatitis C patients.
Collapse
|
21
|
Inhibition of hepatitis C virus infection by DNA aptamer against envelope protein. Antimicrob Agents Chemother 2013. [PMID: 23877701 DOI: 10.1128/aac.00897] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Hepatitis C virus (HCV) envelope protein (E1E2) is essential for virus binding to host cells. Aptamers have been demonstrated to have strong promising applications in drug development. In the current study, a cDNA fragment encoding the entire E1E2 gene of HCV was cloned. E1E2 protein was expressed and purified. Aptamers for E1E2 were selected by the method of selective evolution of ligands by exponential enrichment (SELEX), and the antiviral actions of the aptamers were examined. The mechanism of their antiviral activity was investigated. The data show that selected aptamers for E1E2 specifically recognize the recombinant E1E2 protein and E1E2 protein from HCV-infected cells. CD81 protein blocks the binding of aptamer E1E2-6 to E1E2 protein. Aptamers against E1E2 inhibit HCV infection in an infectious cell culture system although they have no effect on HCV replication in a replicon cell line. Beta interferon (IFN-β) and IFN-stimulated genes (ISGs) are not induced in virus-infected hepatocytes with aptamer treatment, suggesting that E1E2-specific aptamers do not induce innate immunity. E2 protein is essential for the inhibition of HCV infection by aptamer E1E2-6, and the aptamer binding sites are located in E2. Q412R within E1E2 is the major resistance substitution identified. The data indicate that an aptamer against E1E2 exerts its antiviral effects through inhibition of virus binding to host cells. Aptamers against E1E2 can be used with envelope protein to understand the mechanisms of HCV entry and fusion. The aptamers may hold promise for development as therapeutic drugs for hepatitis C patients.
Collapse
|
22
|
|
23
|
Crystal structure of glycoprotein E2 from bovine viral diarrhea virus. Proc Natl Acad Sci U S A 2013; 110:6805-10. [PMID: 23569276 DOI: 10.1073/pnas.1300524110] [Citation(s) in RCA: 99] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Pestiviruses, including bovine viral diarrhea virus, are important animal pathogens and are closely related to hepatitis C virus, which remains a major global health threat. They have an outer lipid envelope bearing two glycoproteins, E1 and E2, required for cell entry. They deliver their genome into the host cell cytoplasm by fusion of their envelope with a cellular membrane. The crystal structure of bovine viral diarrhea virus E2 reveals a unique protein architecture consisting of two Ig-like domains followed by an elongated β-stranded domain with a new fold. E2 forms end-to-end homodimers with a conserved C-terminal motif rich in aromatic residues at the contact. A disulfide bond across the interface explains the acid resistance of pestiviruses and their requirement for a redox activation step to initiate fusion. From the structure of E2, we propose alternative possible membrane fusion mechanisms. We expect the pestivirus fusion apparatus to be conserved in hepatitis C virus.
Collapse
|
24
|
Koutsoudakis G, Forns X, Pérez-Del-Pulgar S. [The molecular biology of hepatitis C virus]. GASTROENTEROLOGIA Y HEPATOLOGIA 2013; 36:280-93. [PMID: 23490024 DOI: 10.1016/j.gastrohep.2012.11.005] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/09/2012] [Accepted: 11/13/2012] [Indexed: 12/12/2022]
Abstract
Since the discovery of the hepatitis C virus (HCV), a plethora of experimental models have evolved, allowing the virus's life cycle and the pathogenesis of associated liver diseases to be investigated. These models range from inoculation of cultured cells with serum from patients with hepatitis C to the use of surrogate models for the study of specific stages of the HCV life cycle: retroviral pseudoparticles for the study of HCV entry, replicons for the study of HCV replication, and the HCV cell culture model, which reproduces the entire life cycle (replication and production of infectious particles). The use of these tools has been and remains crucial to identify potential therapeutic targets in the different stages of the virus's life cycle and to screen new antiviral drugs. A clear example is the recent approval of two viral protease inhibitors (boceprevir and telaprevir) in combination with pegylated interferon and ribavirin for the treatment of chronic hepatitis C. This review analyzes the advances made in the molecular biology of HCV and highlights possible candidates as therapeutic targets for the treatment of HCV infection.
Collapse
Affiliation(s)
- George Koutsoudakis
- Servicio de Hepatología, Hospital Clínic, Institut d'Investigacions Biomèdiques August Pi i Sunyer, Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas, Barcelona, España
| | | | | |
Collapse
|
25
|
Identification of hepatitis C virus inhibitors targeting different aspects of infection using a cell-based assay. Antimicrob Agents Chemother 2012; 56:6109-20. [PMID: 22948883 DOI: 10.1128/aac.01413-12] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
With 2 to 3% of the worldwide population chronically infected, hepatitis C virus (HCV) infection continues to be a major health care burden. Unfortunately, current interferon-based treatment options are not effective in all patients and are associated with significant side effects. Consequently, there is an ongoing need to identify and develop new anti-HCV therapies. Toward this goal, we previously developed a cell-based HCV infection assay for antiviral compound screening based on a low-multiplicity-of-infection approach that uniquely allows for the identification of antiviral compounds that target cell culture-derived HCV (HCVcc) at any step of the viral infection cycle. Using this assay, here we report the screening of the NCI Diversity Set II library, containing 1,974 synthesized chemical compounds, and the identification of compounds with specific anti-HCV activity. In combination with toxicity counterscreening, we identified 30 hits from the compound library, 13 of which showed reproducible and dose-dependent inhibition of HCV with mean therapeutic indices (50% cytotoxic concentration [CC(50)]/50% effective concentration [EC(50)]) of greater than 6. Using HCV pseudotype and replicon systems of multiple HCV genotypes, as well as infectious HCVcc-based assembly and secretion analysis, we determined that different compounds within this group of candidate inhibitors target different steps of viral infection. The compounds identified not only will serve as biological probes to study and further dissect the biology of viral infection but also should facilitate the development of new anti-HCV therapeutic treatments.
Collapse
|
26
|
Morin TJ, Broering TJ, Leav BA, Blair BM, Rowley KJ, Boucher EN, Wang Y, Cheslock PS, Knauber M, Olsen DB, Ludmerer SW, Szabo G, Finberg RW, Purcell RH, Lanford RE, Ambrosino DM, Molrine DC, Babcock GJ. Human monoclonal antibody HCV1 effectively prevents and treats HCV infection in chimpanzees. PLoS Pathog 2012; 8:e1002895. [PMID: 22952447 PMCID: PMC3431327 DOI: 10.1371/journal.ppat.1002895] [Citation(s) in RCA: 136] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2012] [Accepted: 07/23/2012] [Indexed: 12/11/2022] Open
Abstract
Hepatitis C virus (HCV) infection is a leading cause of liver transplantation and there is an urgent need to develop therapies to reduce rates of HCV infection of transplanted livers. Approved therapeutics for HCV are poorly tolerated and are of limited efficacy in this patient population. Human monoclonal antibody HCV1 recognizes a highly-conserved linear epitope of the HCV E2 envelope glycoprotein (amino acids 412–423) and neutralizes a broad range of HCV genotypes. In a chimpanzee model, a single dose of 250 mg/kg HCV1 delivered 30 minutes prior to infusion with genotype 1a H77 HCV provided complete protection from HCV infection, whereas a dose of 50 mg/kg HCV1 did not protect. In addition, an acutely-infected chimpanzee given 250 mg/kg HCV1 42 days following exposure to virus had a rapid reduction in viral load to below the limit of detection before rebounding 14 days later. The emergent virus displayed an E2 mutation (N415K/D) conferring resistance to HCV1 neutralization. Finally, three chronically HCV-infected chimpanzees were treated with a single dose of 40 mg/kg HCV1 and viral load was reduced to below the limit of detection for 21 days in one chimpanzee with rebounding virus displaying a resistance mutation (N417S). The other two chimpanzees had 0.5–1.0 log10 reductions in viral load without evidence of viral resistance to HCV1. In vitro testing using HCV pseudovirus (HCVpp) demonstrated that the sera from the poorly-responding chimpanzees inhibited the ability of HCV1 to neutralize HCVpp. Measurement of antibody responses in the chronically-infected chimpanzees implicated endogenous antibody to E2 and interference with HCV1 neutralization although other factors may also be responsible. These data suggest that human monoclonal antibody HCV1 may be an effective therapeutic for the prevention of graft infection in HCV-infected patients undergoing liver transplantation. The majority of individuals infected with hepatitis C virus (HCV) become chronically infected and many go on to develop liver failure requiring liver transplantation. Unfortunately, the transplanted liver becomes infected with HCV in nearly 100% of transplant patients. Current treatments for HCV are poorly tolerated after liver transplantation and graft health is compromised by infection. We have developed a monoclonal antibody called HCV1 that blocks HCV from infecting liver cells in culture. Using chimpanzees as a model for HCV infection, we demonstrate that HCV1 has the ability to prevent HCV infection. We also show that HCV1 can treat chimpanzees chronically infected with HCV and reduce plasma viral load to below the level of detection for a period of 7 to 21 days. The virus that reemerges in the treated chimpanzees was resistant to HCV1 neutralization demonstrating target engagement. Given the ability of HCV1 to protect chimpanzees from HCV infection, we speculate that HCV1 may be beneficial in HCV- infected patients undergoing liver transplant.
Collapse
Affiliation(s)
- Trevor J. Morin
- MassBiologics, University of Massachusetts Medical School, Boston, Massachusetts, United States of America
| | - Teresa J. Broering
- MassBiologics, University of Massachusetts Medical School, Boston, Massachusetts, United States of America
| | - Brett A. Leav
- MassBiologics, University of Massachusetts Medical School, Boston, Massachusetts, United States of America
| | - Barbra M. Blair
- MassBiologics, University of Massachusetts Medical School, Boston, Massachusetts, United States of America
| | - Kirk J. Rowley
- MassBiologics, University of Massachusetts Medical School, Boston, Massachusetts, United States of America
| | - Elisabeth N. Boucher
- MassBiologics, University of Massachusetts Medical School, Boston, Massachusetts, United States of America
| | - Yang Wang
- MassBiologics, University of Massachusetts Medical School, Boston, Massachusetts, United States of America
| | - Peter S. Cheslock
- MassBiologics, University of Massachusetts Medical School, Boston, Massachusetts, United States of America
| | - Michael Knauber
- MassBiologics, University of Massachusetts Medical School, Boston, Massachusetts, United States of America
| | - David B. Olsen
- Merck Research Laboratories, West Point, Pennsylvania, United States of America
| | - Steve W. Ludmerer
- Merck Research Laboratories, West Point, Pennsylvania, United States of America
| | - Gyongyi Szabo
- Department of Medicine, University of Massachusetts Medical School, Worcester, Massachusetts, United States of America
| | - Robert W. Finberg
- Department of Medicine, University of Massachusetts Medical School, Worcester, Massachusetts, United States of America
| | - Robert H. Purcell
- National Institutes of Health, Bethesda, Maryland, United States of America
| | - Robert E. Lanford
- Department of Virology and Immunology, Texas Biomedical Research Institute and Southwest National Primate Research Center, San Antonio, Texas, United States of America
| | - Donna M. Ambrosino
- MassBiologics, University of Massachusetts Medical School, Boston, Massachusetts, United States of America
| | - Deborah C. Molrine
- MassBiologics, University of Massachusetts Medical School, Boston, Massachusetts, United States of America
| | - Gregory J. Babcock
- MassBiologics, University of Massachusetts Medical School, Boston, Massachusetts, United States of America
- * E-mail:
| |
Collapse
|
27
|
Ye J. Hepatitis C virus: a new class of virus associated with particles derived from very low-density lipoproteins. Arterioscler Thromb Vasc Biol 2012; 32:1099-103. [PMID: 22517369 DOI: 10.1161/atvbaha.111.241448] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Hepatitis C virus (HCV) infects 3% of the world population and is the leading cause of liver failure in the United States. A unique feature of HCV is that the viral particles are integral to very low-density lipoprotein (VLDL)-derived lipoprotein particles. The virus is assembled into VLDL in hepatocytes and released out of the cells together with VLDL. The virus then infects more hepatocytes by entering the cells through the low-density lipoprotein receptor, which mediates uptake of majorities of VLDL-derived lipoprotein particles. These observations suggest that HCV may belong to a novel class of viruses that is associated with VLDL. Understanding the relationship between HCV and VLDL metabolism may reveal new strategies to treat HCV infection.
Collapse
Affiliation(s)
- Jin Ye
- Department of Molecular Genetics, University of Texas Southwestern Medical Center, Dallas, TX 75390-9046, USA.
| |
Collapse
|
28
|
Lacek K, Vercauteren K, Grzyb K, Naddeo M, Verhoye L, Słowikowski MP, Fafi-Kremer S, Patel AH, Baumert TF, Folgori A, Leroux-Roels G, Cortese R, Meuleman P, Nicosia A. Novel human SR-BI antibodies prevent infection and dissemination of HCV in vitro and in humanized mice. J Hepatol 2012; 57:17-23. [PMID: 22414763 DOI: 10.1016/j.jhep.2012.02.018] [Citation(s) in RCA: 66] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/16/2011] [Revised: 01/09/2012] [Accepted: 02/01/2012] [Indexed: 12/15/2022]
Abstract
BACKGROUND & AIMS Hepatitis C virus (HCV)-induced end-stage liver disease is currently the major indication for liver transplantation in the Western world. After transplantation, the donor liver almost inevitably becomes infected by the circulating virus and disease progression is accelerated in immune suppressed transplant patients. The current standard therapy, based on pegylated interferon and ribavirin, induces severe side effects and is often ineffective in this population. Therefore, new strategies to prevent graft re-infection are urgently needed. We have previously shown that monoclonal antibodies (mAbs) against the HCV co-receptor scavenger receptor class B type I (SR-BI/Cla1) inhibit infection by different HCV genotypes in cell culture. METHODS Using phage display libraries, we have generated a large set of novel human mAbs against SR-BI and evaluated their effectiveness in preventing HCV infection and direct cell-to-cell spread in vitro and in vivo using uPA-SCID mice with a humanized liver. RESULTS Eleven human monoclonal antibodies were generated that specifically recognize SR-BI. Two antibodies, mAb8 and mAb151, displayed the highest binding and inhibitory properties and also interfered with direct cell-to-cell spread in vitro. Studies in humanized mice showed that both antibodies were capable of preventing HCV infection and could block intrahepatic spread and virus amplification when administered 3 days after infection. Interestingly, anti-SR-BI therapy was effective against an HCV variant that escaped the control of the adaptive immune response in a liver transplant patient. CONCLUSIONS The anti-SR-BI mAbs generated in this study may represent novel therapeutic tools to prevent HCV re-infection of liver allografts.
Collapse
|
29
|
Rivero-Juarez A, Camacho A, Caruz A, Neukam K, Gonzalez R, Di Lello FA, Perez-Camacho I, Mesa P, Torre-Cisneros J, Peña J, Pineda JA, Rivero A. LDLr genotype modifies the impact of IL28B on HCV viral kinetics after the first weeks of treatment with PEG-IFN/RBV in HIV/HCV patients. AIDS 2012; 26:1009-15. [PMID: 22382144 DOI: 10.1097/qad.0b013e3283528b1c] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
OBJECTIVE To evaluate the effect of low-density lipoprotein receptor (LDLr) and IL28B genotypes on hepatitis C virus (HCV) viral kinetics in the first 4 weeks of treatment with pegylated-interferon (PEG-IFN)/ribavirin (RBV) in HIV patients co-infected with HCV genotype 1. METHODS HIV patients co-infected with HCV genotype 1 and naïve to PEG-IFN/RBV treatment were enrolled in a prospective study. HCV RNA viral loads were measured at baseline and at weeks 1, 2 and 4 after start of therapy. Differences in viral load decline were evaluated for IL28B (CC versus non-CC) and LDLr (CC versus non-CC) genotypes between baseline and weeks 1, 2 and 4. Additionally, the effect of LDLr genotype on HCV viral decline in IL28B CC genotype patients (CC/CC versus CC/non-CC) was analyzed. RESULTS Eighty-seven HIV/HCV genotype 1 co-infected patients were included in the study. Patients carrying the LDLr-CC or IL28B-CC genotypes showed greater HCV viral decline than those with IL28B non-CC or LDLr non-CC genotypes at every time-point analyzed. CC/CC patients had higher rapid virological response (RVR) rates than CC/non-CC patients (41.2 versus 13.3%; P < 0.001). Moreover, at all time points, the CC/CC pattern was associated with greater HCV viral decline than the CC/non-CC genotype (week 1: 1.18 ± 0.51 versus 0.31 ± 0.29, P = 0.041; week 2: 1.55 ± 0.81 versus 0.93 ± 0.73, P = 0.032; week 4: 2.23 ± 1.1 versus 1.5 ± 0.94, P = 0.039). CONCLUSION The LDLr genotype impacts on viral kinetics during the first days of starting treatment with PEG-IFN/RBV in HIV/HCV genotype 1 co-infected patients, and modifies the impact of IL28B on HCV viral decay.
Collapse
|
30
|
Khattab MA, Eslam M, Aly MM, Shatat M, Mousa YI, Abd-Aalhalim H, Aly H, Shaker Y. Serum lipids and chronic hepatitis C genotype 4: interaction and significance. Ann Hepatol 2012; 11:37-46. [PMID: 22166559 DOI: 10.1016/s1665-2681(19)31484-x] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 08/29/2023]
Abstract
BACKGROUND & AIM Metabolic abnormalities are common in chronic hepatitis C infection (CHC). However, the genotypic differences of these disarrangements in patients infected with CHC genotype 4 (HCV-4) and its association with liver histology and viral loads remain unknown. MATERIAL AND METHODS We consecutively enrolled 183 HCV-4 patients and 106 healthy matched controls; to compare metabolic profiles and assess pattern of association of HCV RNA levels as well as histological factors with the serum lipid profile. RESULTS HCV-4 infection is associated with higher homeostasis model assessment of insulin resistance (HOMA-IR) index, despite that, a favourable lipid pattern, consisting of an elevation in HDL- C and a reduction in serum cholesterol (TC), LDL-C and triglyceride (TG) levels, in comparison with normal matched adults. Significant fibrosis was independently associated with HOMA-IR, portal/periportal inflammation grade, serum cholesterol and age. Univariate association was elucidated between lower LDL-C and TC and Metavir activity score and between higher TG and TC and steatosis. In multivariate analysis, severe hepatitis activity, milder hepatic fibrosis, and triglyceride levels are associated with higher HCV RNA levels. CONCLUSION HCV-4 is associated with wide metabolic changes. A proportional relationship is found between serum lipid profiles and hepatitis C viral load and liver histology in patients with HCV-4.
Collapse
|
31
|
In vitro systems for the study of hepatitis C virus infection. Int J Hepatol 2012; 2012:292591. [PMID: 23056952 PMCID: PMC3465938 DOI: 10.1155/2012/292591] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/26/2012] [Revised: 07/03/2012] [Accepted: 07/17/2012] [Indexed: 12/22/2022] Open
Abstract
The study of a virus is made possible by the availability of culture systems in which the viral lifecycle can be realized. Such systems support robust virus entry, replication, assembly, and secretion of nascent virions. Furthermore, culture models provide a platform in which therapeutic interventions can be devised or monitored. Hepatitis C virus (HCV) has a restricted tropism to human and chimpanzees; thus investigations of HCV biology have been hindered for many years due to a lack of small animal models. Nevertheless, significant efforts have been directed at developing cell culture models to elucidate the viral lifecycle in vitro. HCV primarily infects liver parenchymal cells commonly known as hepatocytes. The liver is a highly specialized and complex organ and the development of in vitro systems that reflects this complexity has proven difficult. Consequently, host cell receptor molecules that potentiate HCV infection were identified over a decade after the virus was discovered. A summary of the various HCV in vitro culture models, their advantages, and disadvantages are described.
Collapse
|
32
|
Darling JM, Lemon SM, Fried MW. Hepatitis C. SCHIFF'S DISEASES OF THE LIVER 2011:582-652. [DOI: 10.1002/9781119950509.ch25] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
|
33
|
MacPherson JI, Sidders B, Wieland S, Zhong J, Targett-Adams P, Lohmann V, Backes P, Delpuech-Adams O, Chisari F, Lewis M, Parkinson T, Robertson DL. An integrated transcriptomic and meta-analysis of hepatoma cells reveals factors that influence susceptibility to HCV infection. PLoS One 2011; 6:e25584. [PMID: 22046242 PMCID: PMC3201949 DOI: 10.1371/journal.pone.0025584] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2011] [Accepted: 09/06/2011] [Indexed: 02/06/2023] Open
Abstract
Hepatitis C virus (HCV) is a global problem. To better understand HCV infection researchers employ in vitro HCV cell-culture (HCVcc) systems that use Huh-7 derived hepatoma cells that are particularly permissive to HCV infection. A variety of hyper-permissive cells have been subcloned for this purpose. In addition, subclones of Huh-7 which have evolved resistance to HCV are available. However, the mechanisms of susceptibility or resistance to infection among these cells have not been fully determined. In order to elucidate mechanisms by which hepatoma cells are susceptible or resistant to HCV infection we performed genome-wide expression analyses of six Huh-7 derived cell cultures that have different levels of permissiveness to infection. A great number of genes, representing a wide spectrum of functions are differentially expressed between cells. To focus our investigation, we identify host proteins from HCV replicase complexes, perform gene expression analysis of three HCV infected cells and conduct a detailed analysis of differentially expressed host factors by integrating a variety of data sources. Our results demonstrate that changes relating to susceptibility to HCV infection in hepatoma cells are linked to the innate immune response, secreted signal peptides and host factors that have a role in virus entry and replication. This work identifies both known and novel host factors that may influence HCV infection. Our findings build upon current knowledge of the complex interplay between HCV and the host cell, which could aid development of new antiviral strategies.
Collapse
Affiliation(s)
- Jamie I. MacPherson
- Computational and Evolutionary Biology, Faculty of Life Sciences, University of Manchester, Manchester, United Kingdom
| | - Ben Sidders
- Pfizer Global Research and Development, Sandwich, United Kingdom
| | - Stefan Wieland
- The Scripps Research Institute, La Jolla, California, United States ofAmerica
| | - Jin Zhong
- The Scripps Research Institute, La Jolla, California, United States ofAmerica
| | | | - Volker Lohmann
- Department of Infectious Diseases, Molecular Virology, University of Heidelberg, Heidelberg, Germany
| | - Perdita Backes
- Department of Infectious Diseases, Molecular Virology, University of Heidelberg, Heidelberg, Germany
| | | | - Francis Chisari
- The Scripps Research Institute, La Jolla, California, United States ofAmerica
| | - Marilyn Lewis
- Pfizer Global Research and Development, Sandwich, United Kingdom
| | - Tanya Parkinson
- Pfizer Global Research and Development, Sandwich, United Kingdom
| | - David L. Robertson
- Computational and Evolutionary Biology, Faculty of Life Sciences, University of Manchester, Manchester, United Kingdom
| |
Collapse
|
34
|
Abstract
Cholesterol is an essential molecule for the life cycle of the hepatitis C virus (HCV). This review focuses on the roles of cholesterol in HCV infection and introduces HCV events related to cholesterol metabolism and applications for cholesterol metabolism as a therapeutic target. HCV appears to alter host lipid metabolism into its preferable state, which is clinically recognized as steatosis and hypocholesterolemia. While hepatic fatty acid and triglyceride syntheses are upregulated in chronic hepatitis C patients, no direct evidence of increased hepatic de novo cholesterol biosynthesis has been obtained. Impaired VLDL secretion from hepatocytes is suggested to increase intracellular cholesterol concentrations, which may lead to hypocholesterolemia. Clinically, lower serum cholesterol levels are associated with lower rates of sustained virological responses (SVR) to pegylated-interferon plus ribavirin therapy, but the reason remains unclear. Clinical trials targeting HMG-CoA reductase, the rate-limiting enzyme in the cholesterol biosynthetic pathway, are being conducted using statins. Anti-HCV actions by statins appear to be caused by the inhibition of geranylgeranyl pyrophosphate synthesis rather than their cholesterol lowering effects. Other compounds that block various steps of cholesterol metabolic pathways have also been studied to develop new strategies for the complete eradication of this virus.
Collapse
Affiliation(s)
- Akira Honda
- Department of Gastroenterology, Tokyo Medical University Ibaraki Medical Center, Ibaraki, Japan
| | | |
Collapse
|
35
|
Lupberger J, Zeisel MB, Xiao F, Thumann C, Fofana I, Zona L, Davis C, Mee CJ, Turek M, Gorke S, Royer C, Fischer B, Zahid MN, Lavillette D, Fresquet J, Cosset FL, Rothenberg SM, Pietschmann T, Patel AH, Pessaux P, Doffoël M, Raffelsberger W, Poch O, Mckeating JA, Brino L, Baumert TF. EGFR and EphA2 are host factors for hepatitis C virus entry and possible targets for antiviral therapy. Nat Med 2011; 17:589-95. [PMID: 21516087 PMCID: PMC3938446 DOI: 10.1038/nm.2341] [Citation(s) in RCA: 562] [Impact Index Per Article: 40.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2010] [Accepted: 03/03/2011] [Indexed: 02/07/2023]
Abstract
Hepatitis C virus (HCV) is a major cause of liver disease, but therapeutic options are limited and there are no prevention strategies. Viral entry is the first step of infection and requires the cooperative interaction of several host cell factors. Using a functional RNAi kinase screen, we identified epidermal growth factor receptor and ephrin receptor A2 as host cofactors for HCV entry. Blocking receptor kinase activity by approved inhibitors broadly impaired infection by all major HCV genotypes and viral escape variants in cell culture and in a human liver chimeric mouse model in vivo. The identified receptor tyrosine kinases (RTKs) mediate HCV entry by regulating CD81-claudin-1 co-receptor associations and viral glycoprotein-dependent membrane fusion. These results identify RTKs as previously unknown HCV entry cofactors and show that tyrosine kinase inhibitors have substantial antiviral activity. Inhibition of RTK function may constitute a new approach for prevention and treatment of HCV infection.
Collapse
Affiliation(s)
- Joachim Lupberger
- Interaction Virus-Hôte et Maladies du Foie
INSERM : U748Université de Strasbourg - Faculté de Médecine 3 Rue Koeberle 67000 Strasbourg, FR
| | - Mirjam B. Zeisel
- Interaction Virus-Hôte et Maladies du Foie
INSERM : U748Université de Strasbourg - Faculté de Médecine 3 Rue Koeberle 67000 Strasbourg, FR
| | - Fei Xiao
- Interaction Virus-Hôte et Maladies du Foie
INSERM : U748Université de Strasbourg - Faculté de Médecine 3 Rue Koeberle 67000 Strasbourg, FR
| | - Christine Thumann
- Interaction Virus-Hôte et Maladies du Foie
INSERM : U748Université de Strasbourg - Faculté de Médecine 3 Rue Koeberle 67000 Strasbourg, FR
| | - Isabel Fofana
- Interaction Virus-Hôte et Maladies du Foie
INSERM : U748Université de Strasbourg - Faculté de Médecine 3 Rue Koeberle 67000 Strasbourg, FR
| | - Laetitia Zona
- Interaction Virus-Hôte et Maladies du Foie
INSERM : U748Université de Strasbourg - Faculté de Médecine 3 Rue Koeberle 67000 Strasbourg, FR
| | - Christopher Davis
- Hepatitis C Research Group
University of BirminghamDivision of Immunity and InfectionEdgbaston, Birmingham B15 2TT, GB
| | - Christopher J. Mee
- Hepatitis C Research Group
University of BirminghamDivision of Immunity and InfectionEdgbaston, Birmingham B15 2TT, GB
| | - Marine Turek
- Interaction Virus-Hôte et Maladies du Foie
INSERM : U748Université de Strasbourg - Faculté de Médecine 3 Rue Koeberle 67000 Strasbourg, FR
| | - Sebastian Gorke
- Interaction Virus-Hôte et Maladies du Foie
INSERM : U748Université de Strasbourg - Faculté de Médecine 3 Rue Koeberle 67000 Strasbourg, FR
- Department of Medicine II
University of FreiburgFahnenbergplatz, 79085 Freiburg im Breisgau, DE
| | - Cathy Royer
- Interaction Virus-Hôte et Maladies du Foie
INSERM : U748Université de Strasbourg - Faculté de Médecine 3 Rue Koeberle 67000 Strasbourg, FR
| | - Benoit Fischer
- IGBMC, Institut de Génétique et de Biologie Moléculaire et Cellulaire
INSERM : U964CNRS : UMR7104Université de StrasbourgParc D'Innovation - 1 Rue Laurent Fries - BP 10142 - 67404 Illkirch Cedex, FR
| | - Muhammad N. Zahid
- Interaction Virus-Hôte et Maladies du Foie
INSERM : U748Université de Strasbourg - Faculté de Médecine 3 Rue Koeberle 67000 Strasbourg, FR
| | - Dimitri Lavillette
- Virologie Humaine
INSERM : U758IFR128École Normale Supérieure - Lyon46, Allee D'Italie 69364 Lyon Cedex 07, FR
| | - Judith Fresquet
- Virologie Humaine
INSERM : U758IFR128École Normale Supérieure - Lyon46, Allee D'Italie 69364 Lyon Cedex 07, FR
| | - François-Loïc Cosset
- Virologie Humaine
INSERM : U758IFR128École Normale Supérieure - Lyon46, Allee D'Italie 69364 Lyon Cedex 07, FR
| | - S Michael Rothenberg
- Massachusetts General Hospital Cancer Center
Howard Hughes Medical InstituteHarvard Medical School55 Fruit St, Boston, MA 02114, US
| | - Thomas Pietschmann
- TWINCORE, Division of Experimental Virology
Centre for Experimental and Clinical Infection Research HannoverMedical School Hannover (MHH)Helmholtz Centre for Infection Research (HZI)Feodor-Lynen-Straße 7 D-30625 Hannover, DE
| | - Arvind H. Patel
- MRC Virology Unit
University of Glasgow - Institute of VirologyGlasgow, Glasgow City G12 8QQ, GB
| | - Patrick Pessaux
- Service d'Hépato-Gastroentérologie
Nouvel Hôpital CivilHôpitaux Universitaires de Strasbourg (HUS)1 Place de l'Hôpital 67000 Strasbourg, FR
| | - Michel Doffoël
- Interaction Virus-Hôte et Maladies du Foie
INSERM : U748Université de Strasbourg - Faculté de Médecine 3 Rue Koeberle 67000 Strasbourg, FR
- Service d'Hépato-Gastroentérologie
Nouvel Hôpital CivilHôpitaux Universitaires de Strasbourg (HUS)1 Place de l'Hôpital 67000 Strasbourg, FR
| | - Wolfgang Raffelsberger
- IGBMC, Institut de Génétique et de Biologie Moléculaire et Cellulaire
INSERM : U964CNRS : UMR7104Université de StrasbourgParc D'Innovation - 1 Rue Laurent Fries - BP 10142 - 67404 Illkirch Cedex, FR
| | - Olivier Poch
- IGBMC, Institut de Génétique et de Biologie Moléculaire et Cellulaire
INSERM : U964CNRS : UMR7104Université de StrasbourgParc D'Innovation - 1 Rue Laurent Fries - BP 10142 - 67404 Illkirch Cedex, FR
| | - Jane A. Mckeating
- Hepatitis C Research Group
University of BirminghamDivision of Immunity and InfectionEdgbaston, Birmingham B15 2TT, GB
| | - Laurent Brino
- IGBMC, Institut de Génétique et de Biologie Moléculaire et Cellulaire
INSERM : U964CNRS : UMR7104Université de StrasbourgParc D'Innovation - 1 Rue Laurent Fries - BP 10142 - 67404 Illkirch Cedex, FR
| | - Thomas F. Baumert
- Interaction Virus-Hôte et Maladies du Foie
INSERM : U748Université de Strasbourg - Faculté de Médecine 3 Rue Koeberle 67000 Strasbourg, FR
- Service d'Hépato-Gastroentérologie
Nouvel Hôpital CivilHôpitaux Universitaires de Strasbourg (HUS)1 Place de l'Hôpital 67000 Strasbourg, FR
| |
Collapse
|
36
|
Multifaceted roles for lipids in viral infection. Trends Microbiol 2011; 19:368-75. [PMID: 21530270 PMCID: PMC3130080 DOI: 10.1016/j.tim.2011.03.007] [Citation(s) in RCA: 252] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2011] [Revised: 03/21/2011] [Accepted: 03/22/2011] [Indexed: 02/07/2023]
Abstract
Viruses have evolved complex and dynamic interactions with their host cell. In recent years we have gained insight into the expanding roles for host lipids in the virus life cycle. In particular, viruses target lipid signaling, synthesis, and metabolism to remodel their host cells into an optimal environment for their replication. This review highlights examples from different viruses that illustrate the importance of these diverse virus–lipid interactions.
Collapse
|
37
|
Xue HL, Feng GH, Dou XG. New advances in the development of experimental models of hepatitis C virus infection. Shijie Huaren Xiaohua Zazhi 2011; 19:1269-1274. [DOI: 10.11569/wcjd.v19.i12.1269] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Hepatitis C virus (HCV) infection is another common cause of chronic hepatitis, liver cirrhosis, and hepatocellular carcinoma after hepatitis B virus. It is very difficult to study the variety and replication of HCV and interplay between HCV and the host and to develop new antiviral drugs and vaccines against HCV infection because of lack of susceptible hosts and stable and convenient experimental models of HCV infection. In this paper, we review recent advances in the development of experimental models of HCV infection.
Collapse
|
38
|
Scavenger receptor class B type I and the hypervariable region-1 of hepatitis C virus in cell entry and neutralisation. Expert Rev Mol Med 2011; 13:e13. [PMID: 21489334 DOI: 10.1017/s1462399411001785] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Hepatitis C virus (HCV) infection is a leading cause of chronic liver disease worldwide and represents a major public health problem. Viral attachment and entry - the first encounter of the virus with the host cell - are major targets of neutralising immune responses. Thus, a detailed understanding of the HCV entry process offers interesting opportunities for the development of novel therapeutic strategies. Different cellular or soluble host factors mediate HCV entry, and considerable progress has been made in recent years to decipher how they induce HCV attachment, internalisation and membrane fusion. Among these factors, the scavenger receptor class B type I (SR-BI/SCARB1) is essential for HCV replication in vitro, through its interaction with the HCV E1E2 surface glycoproteins and, more particularly, the HVR1 segment located in the E2 protein. SR-BI is an interesting receptor because HCV, whose replication cycle intersects with lipoprotein metabolism, seems to exploit some aspects of its physiological functions, such as cholesterol transfer from high-density lipoprotein (HDL), during cell entry. SR-BI is also involved in neutralisation attenuation and therefore could be an important target for therapeutic intervention. Recent results suggest that it should be possible to identify inhibitors of the interaction of HCV with SR-BI that do not impair its important physiological properties, as discussed in this review.
Collapse
|
39
|
Tong Y, Zhu Y, Xia X, Liu Y, Feng Y, Hua X, Chen Z, Ding H, Gao L, Wang Y, Feitelson MA, Zhao P, Qi ZT. Tupaia CD81, SR-BI, claudin-1, and occludin support hepatitis C virus infection. J Virol 2011; 85:2793-802. [PMID: 21177818 PMCID: PMC3067968 DOI: 10.1128/jvi.01818-10] [Citation(s) in RCA: 69] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2010] [Accepted: 12/14/2010] [Indexed: 12/19/2022] Open
Abstract
Hepatitis C virus (HCV)-related research has been hampered by the lack of appropriate small-animal models. It has been reported that tree shrews, or tupaias (Tupaia belangeri), can be infected with serum-derived HCV. However, these reports do not firmly establish the tupaia as a reliable model of HCV infection. Human CD81, scavenger receptor class B type I (SR-BI), claudin 1 (CLDN1), and occludin (OCLN) are considered essential receptors or coreceptors for HCV cell entry. In the present study, the roles of these tupaia orthologs in HCV infection were assessed. Both CD81 and SR-BI of tupaia were found to be able to bind with HCV envelope protein 2 (E2). In comparison with human CD81, tupaia CD81 exhibited stronger binding activity with E2 and increased HCV pseudoparticle (HCVpp) cell entry 2-fold. The 293T cells transfected with tupaia CLDN1 became susceptible to HCVpp infection. Moreover, simultaneous transfection of the four tupaia factors into mouse NIH 3T3 cells made the cells susceptible to HCVpp infection. HCVpp of diverse genotypes were able to infect primary tupaia hepatocytes (PTHs), and this infection could be blocked by either anti-CD81 or anti-SR-BI. PTHs could be infected by cell culture-produced HCV (HCVcc) and did produce infectious progeny virus in culture supernatant. These findings indicate that PTHs possess all of the essential factors required for HCV entry and support the complete HCV infection cycle. This highlights both the mechanisms of susceptibility of tupaia to HCV infection and the possibility of using tupaia as a promising small-animal model in HCV study.
Collapse
Affiliation(s)
- Yimin Tong
- Department of Microbiology, Shanghai Key Laboratory of Medical Biodefense, Second Military Medical University, Shanghai 200433, China, Faculty of Life Science and Technology, Kunming University of Science and Technology, Kunming, Yunnan 650224, China, Department of Infectious Diseases, Affiliated Changhai Hospital, Second Military Medical University, Shanghai 200433, China, Department of Biology, College of Science and Technology, Temple University, Philadelphia, Pennsylvania
| | - Yongzhe Zhu
- Department of Microbiology, Shanghai Key Laboratory of Medical Biodefense, Second Military Medical University, Shanghai 200433, China, Faculty of Life Science and Technology, Kunming University of Science and Technology, Kunming, Yunnan 650224, China, Department of Infectious Diseases, Affiliated Changhai Hospital, Second Military Medical University, Shanghai 200433, China, Department of Biology, College of Science and Technology, Temple University, Philadelphia, Pennsylvania
| | - Xueshan Xia
- Department of Microbiology, Shanghai Key Laboratory of Medical Biodefense, Second Military Medical University, Shanghai 200433, China, Faculty of Life Science and Technology, Kunming University of Science and Technology, Kunming, Yunnan 650224, China, Department of Infectious Diseases, Affiliated Changhai Hospital, Second Military Medical University, Shanghai 200433, China, Department of Biology, College of Science and Technology, Temple University, Philadelphia, Pennsylvania
| | - Yuan Liu
- Department of Microbiology, Shanghai Key Laboratory of Medical Biodefense, Second Military Medical University, Shanghai 200433, China, Faculty of Life Science and Technology, Kunming University of Science and Technology, Kunming, Yunnan 650224, China, Department of Infectious Diseases, Affiliated Changhai Hospital, Second Military Medical University, Shanghai 200433, China, Department of Biology, College of Science and Technology, Temple University, Philadelphia, Pennsylvania
| | - Yue Feng
- Department of Microbiology, Shanghai Key Laboratory of Medical Biodefense, Second Military Medical University, Shanghai 200433, China, Faculty of Life Science and Technology, Kunming University of Science and Technology, Kunming, Yunnan 650224, China, Department of Infectious Diseases, Affiliated Changhai Hospital, Second Military Medical University, Shanghai 200433, China, Department of Biology, College of Science and Technology, Temple University, Philadelphia, Pennsylvania
| | - Xian Hua
- Department of Microbiology, Shanghai Key Laboratory of Medical Biodefense, Second Military Medical University, Shanghai 200433, China, Faculty of Life Science and Technology, Kunming University of Science and Technology, Kunming, Yunnan 650224, China, Department of Infectious Diseases, Affiliated Changhai Hospital, Second Military Medical University, Shanghai 200433, China, Department of Biology, College of Science and Technology, Temple University, Philadelphia, Pennsylvania
| | - Zhihui Chen
- Department of Microbiology, Shanghai Key Laboratory of Medical Biodefense, Second Military Medical University, Shanghai 200433, China, Faculty of Life Science and Technology, Kunming University of Science and Technology, Kunming, Yunnan 650224, China, Department of Infectious Diseases, Affiliated Changhai Hospital, Second Military Medical University, Shanghai 200433, China, Department of Biology, College of Science and Technology, Temple University, Philadelphia, Pennsylvania
| | - Hui Ding
- Department of Microbiology, Shanghai Key Laboratory of Medical Biodefense, Second Military Medical University, Shanghai 200433, China, Faculty of Life Science and Technology, Kunming University of Science and Technology, Kunming, Yunnan 650224, China, Department of Infectious Diseases, Affiliated Changhai Hospital, Second Military Medical University, Shanghai 200433, China, Department of Biology, College of Science and Technology, Temple University, Philadelphia, Pennsylvania
| | - Li Gao
- Department of Microbiology, Shanghai Key Laboratory of Medical Biodefense, Second Military Medical University, Shanghai 200433, China, Faculty of Life Science and Technology, Kunming University of Science and Technology, Kunming, Yunnan 650224, China, Department of Infectious Diseases, Affiliated Changhai Hospital, Second Military Medical University, Shanghai 200433, China, Department of Biology, College of Science and Technology, Temple University, Philadelphia, Pennsylvania
| | - Yongzhi Wang
- Department of Microbiology, Shanghai Key Laboratory of Medical Biodefense, Second Military Medical University, Shanghai 200433, China, Faculty of Life Science and Technology, Kunming University of Science and Technology, Kunming, Yunnan 650224, China, Department of Infectious Diseases, Affiliated Changhai Hospital, Second Military Medical University, Shanghai 200433, China, Department of Biology, College of Science and Technology, Temple University, Philadelphia, Pennsylvania
| | - Mark A. Feitelson
- Department of Microbiology, Shanghai Key Laboratory of Medical Biodefense, Second Military Medical University, Shanghai 200433, China, Faculty of Life Science and Technology, Kunming University of Science and Technology, Kunming, Yunnan 650224, China, Department of Infectious Diseases, Affiliated Changhai Hospital, Second Military Medical University, Shanghai 200433, China, Department of Biology, College of Science and Technology, Temple University, Philadelphia, Pennsylvania
| | - Ping Zhao
- Department of Microbiology, Shanghai Key Laboratory of Medical Biodefense, Second Military Medical University, Shanghai 200433, China, Faculty of Life Science and Technology, Kunming University of Science and Technology, Kunming, Yunnan 650224, China, Department of Infectious Diseases, Affiliated Changhai Hospital, Second Military Medical University, Shanghai 200433, China, Department of Biology, College of Science and Technology, Temple University, Philadelphia, Pennsylvania
| | - Zhong-Tian Qi
- Department of Microbiology, Shanghai Key Laboratory of Medical Biodefense, Second Military Medical University, Shanghai 200433, China, Faculty of Life Science and Technology, Kunming University of Science and Technology, Kunming, Yunnan 650224, China, Department of Infectious Diseases, Affiliated Changhai Hospital, Second Military Medical University, Shanghai 200433, China, Department of Biology, College of Science and Technology, Temple University, Philadelphia, Pennsylvania
| |
Collapse
|
40
|
Mechanism of inhibition of enveloped virus membrane fusion by the antiviral drug arbidol. PLoS One 2011; 6:e15874. [PMID: 21283579 PMCID: PMC3026800 DOI: 10.1371/journal.pone.0015874] [Citation(s) in RCA: 94] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2010] [Accepted: 11/28/2010] [Indexed: 12/13/2022] Open
Abstract
The broad-spectrum antiviral arbidol (Arb) inhibits cell entry of enveloped viruses by blocking viral fusion with host cell membrane. To better understand Arb mechanism of action, we investigated its interactions with phospholipids and membrane peptides. We demonstrate that Arb associates with phospholipids in the micromolar range. NMR reveals that Arb interacts with the polar head-group of phospholipid at the membrane interface. Fluorescence studies of interactions between Arb and either tryptophan derivatives or membrane peptides reconstituted into liposomes show that Arb interacts with tryptophan in the micromolar range. Interestingly, apparent binding affinities between lipids and tryptophan residues are comparable with those of Arb IC50 of the hepatitis C virus (HCV) membrane fusion. Since tryptophan residues of membrane proteins are known to bind preferentially at the membrane interface, these data suggest that Arb could increase the strength of virus glycoprotein's interactions with the membrane, due to a dual binding mode involving aromatic residues and phospholipids. The resulting complexation would inhibit the expected viral glycoprotein conformational changes required during the fusion process. Our findings pave the way towards the design of new drugs exhibiting Arb-like interfacial membrane binding properties to inhibit early steps of virus entry, i.e., attractive targets to combat viral infection.
Collapse
|
41
|
Pezacki JP, Singaravelu R, Lyn RK. Host-virus interactions during hepatitis C virus infection: a complex and dynamic molecular biosystem. MOLECULAR BIOSYSTEMS 2010; 6:1131-42. [PMID: 20549003 DOI: 10.1039/b924668c] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
The hepatitis C virus (HCV) is a global health issue with no vaccine available and limited clinical treatment options. Like other obligate parasites, HCV requires host cellular components of an infected individual to propagate. These host-virus interactions during HCV infection are complex and dynamic and involve the hijacking of host cell environments, enzymes and pathways. Understanding this unique molecular biosystem has the potential to yield new and exciting strategies for therapeutic intervention. Advances in genomics and proteomics have opened up new possibilities for the rapid measurement of global changes at the transcriptional and translational levels during infection. However, these techniques only yield snapshots of host-virus interactions during HCV infection. Other new methods that involve the imaging of biomolecular interactions during HCV infection are required to identify key interactions that may be transient and dynamic. Herein we highlight systems biology based strategies that have helped to identify key host-virus interactions during HCV replication and infection. Novel biophysical tools are also highlighted for identification and visualization of activities and interactions between HCV and its host hepatocyte. As some of these methods mature, we expect them to pave the way forward for further exploration of this complex biosystem and elucidation of mechanisms for HCV pathogenesis and carcinogenesis.
Collapse
Affiliation(s)
- John Paul Pezacki
- Steacie Institute for Molecular Sciences, National Research Council of Canada, 100 Sussex Dr., Ottawa, Ontario, Canada.
| | | | | |
Collapse
|
42
|
Fafi-Kremer S, Fofana I, Soulier E, Carolla P, Meuleman P, Leroux-Roels G, Patel AH, Cosset FL, Pessaux P, Doffoël M, Wolf P, Stoll-Keller F, Baumert TF. Viral entry and escape from antibody-mediated neutralization influence hepatitis C virus reinfection in liver transplantation. ACTA ACUST UNITED AC 2010; 207:2019-31. [PMID: 20713596 PMCID: PMC2931157 DOI: 10.1084/jem.20090766] [Citation(s) in RCA: 114] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
End-stage liver disease caused by chronic hepatitis C virus (HCV) infection is a leading cause for liver transplantation (LT). Due to viral evasion from host immune responses and the absence of preventive antiviral strategies, reinfection of the graft is universal. The mechanisms by which the virus evades host immunity to reinfect the liver graft are unknown. In a longitudinal analysis of six HCV-infected patients undergoing LT, we demonstrate that HCV variants reinfecting the liver graft were characterized by efficient entry and poor neutralization by antibodies present in pretransplant serum compared with variants not detected after transplantation. Monoclonal antibodies directed against HCV envelope glycoproteins or a cellular entry factor efficiently cross-neutralized infection of human hepatocytes by patient-derived viral isolates that were resistant to autologous host-neutralizing responses. These findings provide significant insights into the molecular mechanisms of viral evasion during HCV reinfection and suggest that viral entry is a viable target for prevention of HCV reinfection of the liver graft.
Collapse
Affiliation(s)
- Samira Fafi-Kremer
- Institut National de la Santé et de la Recherche Médicale, Unité 748, Strasbourg, France
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Uprichard SL. Hepatitis C virus experimental model systems and antiviral drug research. Virol Sin 2010; 25:227-45. [PMID: 20960298 DOI: 10.1007/s12250-010-3134-0] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2010] [Accepted: 04/18/2010] [Indexed: 12/27/2022] Open
Abstract
An estimated 130 million people worldwide are chronically infected with hepatitis C virus (HCV) making it a leading cause of liver disease worldwide. Because the currently available therapy of pegylated interferon-alpha and ribavirin is only effective in a subset of patients, the development of new HCV antivirals is a healthcare imperative. This review discusses the experimental models available for HCV antiviral drug research, recent advances in HCV antiviral drug development, as well as active research being pursued to facilitate development of new HCV-specific therapeutics.
Collapse
Affiliation(s)
- Susan L Uprichard
- Department of Medicine, University of Illinois at Chicago, Chicago, IL 60612, USA.
| |
Collapse
|
44
|
Katsarou K, Lavdas AΑ, Tsitoura P, Serti E, Markoulatos P, Mavromara P, Georgopoulou U. Endocytosis of hepatitis C virus non-enveloped capsid-like particles induces MAPK-ERK1/2 signaling events. Cell Mol Life Sci 2010; 67:2491-506. [PMID: 20358251 PMCID: PMC11115770 DOI: 10.1007/s00018-010-0351-5] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2009] [Revised: 03/08/2010] [Accepted: 03/11/2010] [Indexed: 12/12/2022]
Abstract
Although HCV is an enveloped virus, naked nucleocapsids have been reported in the serum of infected patients. The HCV core particle serves as a protective capsid shell for the viral genome and recombinant in vitro assembled HCV core particles induce strong specific immunity. We investigated the post-binding mechanism of recombinant core particle uptake and its intracellular fate. In hepatic cells, these particles are internalized, most likely in a clathrin-dependent pathway, reaching early to late endosomes and finally lysosomes. The endocytic acidic milieu is implicated in trafficking process. Using specific phosphoantibodies, signaling pathway inhibitors and chemical agents, ERK(1/2) was found to be activated in a sustained way after endocytosis, followed by downstream immediate early genes (c-fos and egr-1) modulation. We propose that the intriguing properties of cellular internalization of HCV non-enveloped particles can induce specific ERK(1/2)-MAPKs events that could be important in HCV life cycle and pathogenesis of HCV infection.
Collapse
Affiliation(s)
| | - Alexandros Α. Lavdas
- Laboratory of Cellular and Molecular Neurobiology, Hellenic Pasteur Institute, Athens, Greece
| | - Panagiota Tsitoura
- Present Address: Insect Molecular Genetics and Biotechnology, Institute of Biology, NCSR Demokritos, Athens, Greece
| | - Elisavet Serti
- Molecular Virology Laboratory, Hellenic Pasteur Institute, Athens, Greece
| | | | - Penelope Mavromara
- Molecular Virology Laboratory, Hellenic Pasteur Institute, Athens, Greece
| | | |
Collapse
|
45
|
Silberstein E, Mihalik K, Ulitzky L, Plant EP, Puig M, Gagneten S, Yu MYW, Kaushik-Basu N, Feinstone SM, Taylor DR. Persistent growth of a human plasma-derived hepatitis C virus genotype 1b isolate in cell culture. PLoS Pathog 2010; 6:e1000910. [PMID: 20502631 PMCID: PMC2873922 DOI: 10.1371/journal.ppat.1000910] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2009] [Accepted: 04/16/2010] [Indexed: 01/13/2023] Open
Abstract
HCV (hepatitis C virus) research, including therapeutics and vaccine development, has been hampered by the lack of suitable tissue culture models. Development of cell culture systems for the growth of the most drug-resistant HCV genotype (1b) as well as natural isolates has remained a challenge. Transfection of cultured cells with adenovirus-associated RNA(I) (VA RNA(I)), a known interferon (IFN) antagonist and inhibitor of dsRNA-mediated antiviral pathways, enhanced the growth of plasma-derived HCV genotype 1b. Furthermore, persistent viral growth was achieved after passaging through IFN-alpha/beta-deficient VeroE6 cells for 2 years. Persistently infected cells were maintained in culture for an additional 4 years, and the virus rescued from these cells induced strong cytopathic effect (CPE). Using a CPE-based assay, we measured inhibition of viral production by anti-HCV specific inhibitors, including 2'-C-Methyl-D-Adenosine, demonstrating its utility for the evaluation of HCV antivirals. This virus constitutes a novel tool for the study of one of the most relevant strains of HCV, genotype 1b, which will now be available for HCV life cycle research and useful for the development of new therapeutics.
Collapse
Affiliation(s)
- Erica Silberstein
- Division of Emerging and Transfusion-transmitted Diseases, Center for Biologics Evaluation and Research, Food and Drug Administration, Bethesda, Maryland, United States of America
| | - Kathleen Mihalik
- Division of Viral Products, Center for Biologics Evaluation and Research, Food and Drug Administration, Bethesda, Maryland, United States of America
| | - Laura Ulitzky
- Division of Emerging and Transfusion-transmitted Diseases, Center for Biologics Evaluation and Research, Food and Drug Administration, Bethesda, Maryland, United States of America
| | - Ewan P. Plant
- Division of Emerging and Transfusion-transmitted Diseases, Center for Biologics Evaluation and Research, Food and Drug Administration, Bethesda, Maryland, United States of America
| | - Montserrat Puig
- Division of Therapeutic Proteins, Center for Drug Evaluation and Research, Food and Drug Administration, Bethesda, Maryland, United States of America
| | - Sara Gagneten
- Division of Viral Products, Center for Biologics Evaluation and Research, Food and Drug Administration, Bethesda, Maryland, United States of America
| | - Mei-ying W. Yu
- Division of Hematology, Center for Biologics Evaluation and Research, Food and Drug Administration, Bethesda, Maryland, United States of America
| | - Neerja Kaushik-Basu
- Department of Biochemistry and Molecular Biology, University of Medicine and Dentistry of New Jersey, Newark, New Jersey, United States of America
| | - Stephen M. Feinstone
- Division of Viral Products, Center for Biologics Evaluation and Research, Food and Drug Administration, Bethesda, Maryland, United States of America
| | - Deborah R. Taylor
- Division of Emerging and Transfusion-transmitted Diseases, Center for Biologics Evaluation and Research, Food and Drug Administration, Bethesda, Maryland, United States of America
| |
Collapse
|
46
|
Samuel CE. Thematic minireview series: elucidating hepatitis C virus-host interactions at the biochemical level. J Biol Chem 2010; 285:22723-4. [PMID: 20457611 DOI: 10.1074/jbc.r110.142240] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Affiliation(s)
- Charles E Samuel
- Department of Molecular, Cellular and Developmental Biology and the Biomolecular Sciences and Engineering Program, University of California, Santa Barbara, California 93106, USA.
| |
Collapse
|
47
|
The disulfide bonds in glycoprotein E2 of hepatitis C virus reveal the tertiary organization of the molecule. PLoS Pathog 2010; 6:e1000762. [PMID: 20174556 PMCID: PMC2824758 DOI: 10.1371/journal.ppat.1000762] [Citation(s) in RCA: 189] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2009] [Accepted: 01/12/2010] [Indexed: 02/07/2023] Open
Abstract
Hepatitis C virus (HCV), a major cause of chronic liver disease in humans, is the focus of intense research efforts worldwide. Yet structural data on the viral envelope glycoproteins E1 and E2 are scarce, in spite of their essential role in the viral life cycle. To obtain more information, we developed an efficient production system of recombinant E2 ectodomain (E2e), truncated immediately upstream its trans-membrane (TM) region, using Drosophila melanogaster cells. This system yields a majority of monomeric protein, which can be readily separated chromatographically from contaminating disulfide-linked aggregates. The isolated monomeric E2e reacts with a number of conformation-sensitive monoclonal antibodies, binds the soluble CD81 large external loop and efficiently inhibits infection of Huh7.5 cells by infectious HCV particles (HCVcc) in a dose-dependent manner, suggesting that it adopts a native conformation. These properties of E2e led us to experimentally determine the connectivity of its 9 disulfide bonds, which are strictly conserved across HCV genotypes. Furthermore, circular dichroism combined with infrared spectroscopy analyses revealed the secondary structure contents of E2e, indicating in particular about 28% β-sheet, in agreement with the consensus secondary structure predictions. The disulfide connectivity pattern, together with data on the CD81 binding site and reported E2 deletion mutants, enabled the threading of the E2e polypeptide chain onto the structural template of class II fusion proteins of related flavi- and alphaviruses. The resulting model of the tertiary organization of E2 gives key information on the antigenicity determinants of the virus, maps the receptor binding site to the interface of domains I and III, and provides insight into the nature of a putative fusogenic conformational change. Little is known about the structure of the envelope glycoproteins of the hepatitis C virus (HCV), in spite of their essential role in the viral cycle of this major human pathogen. Here, we determined the connectivity of the 9 disulfide bonds formed by the strictly conserved 18 cysteines of the ectodomain of HCV glycoprotein E2. We show that this information, together with important functional data available in the literature, impose important restrictions to the possible three-dimensional fold of the molecule. Indeed, these constraints allow the unambiguous threading of the predicted secondary structure elements along the polypeptide chain onto the template provided by the crystal structures of related flavi- and alphavirus class II fusion proteins. The resulting model of the tertiary organization of E2 shows the amino acid distribution among the characteristic class II domains, places the CD81 binding site at the interface of domains I and III, and highlights the location of a candidate fusion loop.
Collapse
|
48
|
Gouttenoire J, Penin F, Moradpour D. Hepatitis C virus nonstructural protein 4B: a journey into unexplored territory. Rev Med Virol 2010; 20:117-29. [DOI: 10.1002/rmv.640] [Citation(s) in RCA: 98] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
49
|
Quer J, Buti M, Cubero M, Guardia J, Esteban R, Esteban JI. New strategies for the treatment of hepatitis C virus infection and implications of resistance to new direct-acting antiviral agents. Infect Drug Resist 2010; 3:133-45. [PMID: 21694902 PMCID: PMC3108733 DOI: 10.2147/idr.s7136] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2010] [Indexed: 12/22/2022] Open
Abstract
Persistent hepatitis C virus (HCV) infection is a leading cause of chronic hepatitis, cirrhosis, and hepatocellular carcinoma and the major indication for liver transplantation in adults. Current standard of care treatment (SOC) with pegylated-interferon-α 2 and ribavirin (RBV) has a limited efficacy and is associated with significant side effects frequently associated with poor compliance or treatment discontinuation, requiring specialized and frequent monitoring. To overcome the limited efficacy of SOC, more than 50 direct-acting antiviral agents (DAA) designed to target viral-encoded proteins essential in the HCV life cycle are currently under development. The rapid selection of resistant mutants associated with the quasispecies nature of HCV with high mutation and replication rates is one of the main challenges for the new HCV therapies. Predictive host and viral factors together with combination of DAAs with or without IFN and/or RBV need to be accurately evaluated to design the most effective individualized treatment strategy within the shortest time interval and with minimum side effects.
Collapse
Affiliation(s)
- Josep Quer
- Liver Unit, Internal Medicine Hospital Universitari Vall d'Hebron, Institut de Recerca (VHIR), Barcelona, Spain
| | | | | | | | | | | |
Collapse
|
50
|
Syed GH, Amako Y, Siddiqui A. Hepatitis C virus hijacks host lipid metabolism. Trends Endocrinol Metab 2010; 21:33-40. [PMID: 19854061 PMCID: PMC2818172 DOI: 10.1016/j.tem.2009.07.005] [Citation(s) in RCA: 276] [Impact Index Per Article: 18.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/30/2009] [Revised: 07/17/2009] [Accepted: 07/24/2009] [Indexed: 12/19/2022]
Abstract
Hepatitis C virus (HCV) enhances its replication by modulating host cell lipid metabolism. HCV circulates in the blood in association with lipoproteins. HCV infection is associated with enhanced lipogenesis, reduced secretion, and beta-oxidation of lipids. HCV-induced imbalance in lipid homeostasis leads to steatosis. Many lipids are crucial for the virus life cycle, and inhibitors of cholesterol/fatty acid biosynthetic pathways inhibit virus replication, maturation and secretion. HCV negatively modulates the synthesis and secretion of very low-density lipoproteins (VLDL). Components involved in VLDL assembly are also required for HCV morphogenesis/secretion, suggesting that HCV co-opts the VLDL secretory pathway for its own secretion. This review highlights HCV-altered lipid metabolic events that aid the virus life cycle and ultimately promote liver disease.
Collapse
|