Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
Add more filters










Publication year range
1.
Nat Commun ; 14(1): 5191, 2023 08 25.
Article in English | MEDLINE | ID: mdl-37626079

ABSTRACT

Many of the currently available COVID-19 vaccines and therapeutics are not effective against newly emerged SARS-CoV-2 variants. Here, we developed the metallo-enzyme domain of angiotensin converting enzyme 2 (ACE2)-the cellular receptor of SARS-CoV-2-into an IgM-like inhalable molecule (HH-120). HH-120 binds to the SARS-CoV-2 Spike (S) protein with high avidity and confers potent and broad-spectrum neutralization activity against all known SARS-CoV-2 variants of concern. HH-120 was developed as an inhaled formulation that achieves appropriate aerodynamic properties for rodent and monkey respiratory system delivery, and we found that early administration of HH-120 by aerosol inhalation significantly reduced viral loads and lung pathology scores in male golden Syrian hamsters infected by the SARS-CoV-2 ancestral strain (GDPCC-nCoV27) and the Delta variant. Our study presents a meaningful advancement in the inhalation delivery of large biologics like HH-120 (molecular weight (MW) ~ 1000 kDa) and demonstrates that HH-120 can serve as an efficacious, safe, and convenient agent against SARS-CoV-2 variants. Finally, given the known role of ACE2 in viral reception, it is conceivable that HH-120 has the potential to be efficacious against additional emergent coronaviruses.


Subject(s)
Angiotensin-Converting Enzyme 2 , COVID-19 , Male , Animals , Cricetinae , Humans , COVID-19 Vaccines , SARS-CoV-2/genetics , Mesocricetus , Immunoglobulin M
2.
Cell Rep ; 35(13): 109288, 2021 06 29.
Article in English | MEDLINE | ID: mdl-34192543

ABSTRACT

The hepatitis B virus (HBV) infects 257 million people worldwide. HBV infection requires establishment and persistence of covalently closed circular (ccc) DNA, a viral episome, in nucleus. Here, we study cccDNA spatial localization in the 3D host genome by using chromosome conformation capture-based sequencing analysis and fluorescence in situ hybridization (FISH). We show that transcriptionally inactive cccDNA is not randomly distributed in host nucleus. Rather, it is preferentially accumulated at specialized areas, including regions close to chromosome 19 (chr.19). Activation of the cccDNA is apparently associated with its re-localization, from a pre-established heterochromatin hub formed by 5 regions of chr.19 to transcriptionally active regions formed by chr.19 and nearby chromosomes including chr.16, 17, 20, and 22. This active versus inactive positioning at discrete regions of the host genome is primarily controlled by the viral HBx protein and by host factors including the structural maintenance of chromosomes protein 5/6 (SMC5/6) complex.


Subject(s)
Chromosomes, Human, Pair 19/genetics , Genome, Human , Hepatitis B virus/genetics , Hepatitis B/genetics , Hepatitis B/virology , Plasmids/genetics , Transcription, Genetic , Base Sequence , Cells, Cultured , DNA, Viral/genetics , Genome, Viral , Hep G2 Cells , Hepatocytes/pathology , Hepatocytes/virology , Heterochromatin/metabolism , Humans
3.
Mol Ther ; 29(4): 1572-1584, 2021 04 07.
Article in English | MEDLINE | ID: mdl-33429083

ABSTRACT

Glypican-3 (GPC3) is a well-characterized hepatocellular carcinoma (HCC)-associated antigen, yet anti-GPC3 therapies have achieved only minimal clinical progress. CD47 is a ubiquitously expressed innate immune checkpoint that promotes evasion of tumors from immune surveillance. Given both the specific expression of GPC3 in HCC and the known phagocytosis inhibitory effect of CD47 in liver cancer, we hypothesized that a bispecific antibody (BsAb) that co-engages with GPC3 and CD47 may offer excellent antitumor efficacy with minimal toxicity. Here, we generated a novel BsAb: GPC3/CD47 biAb. With the use of both in vitro and in vivo assays, we found that GPC3/CD47 biAb exerts strong antitumor activity preferentially against dual antigen-expressing tumor cells. In hCD47/human signal regulatory protein alpha (hCD47/hSIRPα) humanized mice, GPC3/CD47 biAb had an extended serum half-life without causing systemic toxicity. Importantly, GPC3/CD47 biAb induced enhanced Fc-mediated effector functions to dual antigen-expressing HCC cells in vitro, and both macrophages and neutrophils are required for its strong efficacy against xenograft HCC tumors. Notably, GPC3/CD47 biAb outperformed monotherapies and a combination therapy with anti-CD47 and anti-GPC3 monoclonal antibodies (mAbs) in a xenograft HCC model. Our study illustrates a strategy for improving HCC treatment by boosting innate immune responses and presents new insights to inform antibody design for the future development of innovative immune therapies.


Subject(s)
CD47 Antigen/genetics , Carcinoma, Hepatocellular/drug therapy , Glypicans/genetics , Liver Neoplasms/drug therapy , Animals , Antibodies, Bispecific/pharmacology , Antibodies, Monoclonal/pharmacology , CD47 Antigen/antagonists & inhibitors , CD47 Antigen/immunology , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/immunology , Carcinoma, Hepatocellular/pathology , Cell Line, Tumor , Glypicans/antagonists & inhibitors , Glypicans/immunology , Humans , Immunity, Innate/drug effects , Immunity, Innate/immunology , Liver Neoplasms/genetics , Liver Neoplasms/immunology , Liver Neoplasms/pathology , Mice , Xenograft Model Antitumor Assays
4.
PLoS Pathog ; 13(9): e1006658, 2017 Sep.
Article in English | MEDLINE | ID: mdl-28945802

ABSTRACT

Hepatitis B virus (HBV) core protein assembles viral pre-genomic (pg) RNA and DNA polymerase into nucleocapsids for reverse transcriptional DNA replication to take place. Several chemotypes of small molecules, including heteroaryldihydropyrimidines (HAPs) and sulfamoylbenzamides (SBAs), have been discovered to allosterically modulate core protein structure and consequentially alter the kinetics and pathway of core protein assembly, resulting in formation of irregularly-shaped core protein aggregates or "empty" capsids devoid of pre-genomic RNA and viral DNA polymerase. Interestingly, in addition to inhibiting nucleocapsid assembly and subsequent viral genome replication, we have now demonstrated that HAPs and SBAs differentially modulate the biosynthesis of covalently closed circular (ccc) DNA from de novo infection and intracellular amplification pathways by inducing disassembly of nucleocapsids derived from virions as well as double-stranded DNA-containing progeny nucleocapsids in the cytoplasm. Specifically, the mistimed cuing of nucleocapsid uncoating prevents cccDNA formation during de novo infection of hepatocytes, while transiently accelerating cccDNA synthesis from cytoplasmic progeny nucleocapsids. Our studies indicate that elongation of positive-stranded DNA induces structural changes of nucleocapsids, which confers ability of mature nucleocapsids to bind CpAMs and triggers its disassembly. Understanding the molecular mechanism underlying the dual effects of the core protein allosteric modulators on nucleocapsid assembly and disassembly will facilitate the discovery of novel core protein-targeting antiviral agents that can more efficiently suppress cccDNA synthesis and cure chronic hepatitis B.


Subject(s)
DNA, Circular/biosynthesis , Hepatitis B virus/physiology , Hepatitis B, Chronic/virology , Virus Replication/drug effects , Antiviral Agents/pharmacology , Cell Line , DNA, Circular/genetics , DNA, Viral , DNA-Directed DNA Polymerase/metabolism , Hepatocytes/virology , Humans , Nucleocapsid/drug effects , Nucleocapsid/genetics , Real-Time Polymerase Chain Reaction , Virus Replication/physiology
5.
Elife ; 62017 09 26.
Article in English | MEDLINE | ID: mdl-28949917

ABSTRACT

Hepatitis B virus (HBV) infection is a major global health problem. Currently-available therapies are ineffective in curing chronic HBV infection. HBV and its satellite hepatitis D virus (HDV) infect hepatocytes via binding of the preS1 domain of its large envelope protein to sodium taurocholate cotransporting polypeptide (NTCP). Here, we developed novel human monoclonal antibodies that block the engagement of preS1 with NTCP and neutralize HBV and HDV with high potency. One antibody, 2H5-A14, functions at picomolar level and exhibited neutralization-activity-mediated prophylactic effects. It also acts therapeutically by eliciting antibody-Fc-dependent immunological effector functions that impose durable suppression of viral infection in HBV-infected mice, resulting in reductions in the levels of the small envelope antigen and viral DNA, with no emergence of escape mutants. Our results illustrate a novel antibody-Fc-dependent approach for HBV treatment and suggest 2H5-A14 as a novel clinical candidate for HBV prevention and treatment of chronic HBV infection.


Subject(s)
Antibodies, Neutralizing/administration & dosage , Antibodies, Viral/administration & dosage , Antiviral Agents/administration & dosage , Hepatitis B/therapy , Immunotherapy/methods , Animals , Antibodies, Monoclonal/administration & dosage , Chemoprevention/methods , Disease Models, Animal , Hepatitis B/prevention & control , Hepatitis B virus/drug effects , Hepatitis Delta Virus/drug effects , Humans , Mice , Treatment Outcome
6.
Methods Mol Biol ; 1540: 1-14, 2017.
Article in English | MEDLINE | ID: mdl-27975303

ABSTRACT

Sodium taurocholate cotransporting polypeptide (NTCP) has been identified as a functional receptor for hepatitis B virus (HBV). Expressing human NTCP in human hepatoma HepG2 cells (HepG2-NTCP) renders these cells susceptible for HBV infection. The HepG2-NTCP stably transfected cell line provides a much-needed and easily accessible platform for studying the virus. HepG2-NTCP cells could also be used to identify chemicals targeting key steps of the virus life cycle including HBV covalent closed circular (ccc) DNA, and enable the development of novel antivirals against the infection.Many factors may contribute to the efficiency of HBV infection on HepG2-NTCP cells, with clonal differences among cell line isolates, the source of viral inoculum, and infection medium among the most critical ones. Here, we provide detailed protocols for efficient HBV infection of HepG2-NTCP cells in culture; generation and selection of single cell clones of HepG2-NTCP; production of infectious HBV virion stock through DNA transfection of recombinant plasmid that enables studying primary clinical HBV isolates; and assessing the infection with immunostaining of HBV antigens and Southern blot analysis of HBV cccDNA.


Subject(s)
Hepatitis B virus/physiology , Hepatitis B/metabolism , Hepatitis B/virology , Organic Anion Transporters, Sodium-Dependent/metabolism , Receptors, Virus/metabolism , Symporters/metabolism , Cell Line , Cells, Cultured , DNA, Circular , DNA, Viral , Enzyme-Linked Immunosorbent Assay , Fluorescent Antibody Technique , Gene Expression , Hep G2 Cells , Hepatitis B/genetics , Hepatitis B Surface Antigens/immunology , Hepatitis B e Antigens/immunology , Humans , Organic Anion Transporters, Sodium-Dependent/genetics , RNA, Viral , Receptors, Virus/genetics , Symporters/genetics , Virus Replication
7.
PLoS Pathog ; 12(10): e1005893, 2016 Oct.
Article in English | MEDLINE | ID: mdl-27783675

ABSTRACT

Hepatitis B virus (HBV) infection of hepatocytes begins by binding to its cellular receptor sodium taurocholate cotransporting polypeptide (NTCP), followed by the internalization of viral nucleocapsid into the cytoplasm. The viral relaxed circular (rc) DNA genome in nucleocapsid is transported into the nucleus and converted into covalently closed circular (ccc) DNA to serve as a viral persistence reservoir that is refractory to current antiviral therapies. Host DNA repair enzymes have been speculated to catalyze the conversion of rcDNA to cccDNA, however, the DNA polymerase(s) that fills the gap in the plus strand of rcDNA remains to be determined. Here we conducted targeted genetic screening in combination with chemical inhibition to identify the cellular DNA polymerase(s) responsible for cccDNA formation, and exploited recombinant HBV with capsid coding deficiency which infects HepG2-NTCP cells with similar efficiency of wild-type HBV to assure cccDNA synthesis is exclusively from de novo HBV infection. We found that DNA polymerase κ (POLK), a Y-family DNA polymerase with maximum activity in non-dividing cells, substantially contributes to cccDNA formation during de novo HBV infection. Depleting gene expression of POLK in HepG2-NTCP cells by either siRNA knockdown or CRISPR/Cas9 knockout inhibited the conversion of rcDNA into cccDNA, while the diminished cccDNA formation in, and hence the viral infection of, the knockout cells could be effectively rescued by ectopic expression of POLK. These studies revealed that POLK is a crucial host factor required for cccDNA formation during a de novo HBV infection and suggest that POLK may be a potential target for developing antivirals against HBV.


Subject(s)
DNA, Circular/genetics , DNA, Viral/genetics , DNA-Directed DNA Polymerase/metabolism , Hepatitis B/genetics , Blotting, Southern , Cell Line , DNA, Circular/metabolism , DNA, Viral/metabolism , Enzyme-Linked Immunosorbent Assay , Fluorescent Antibody Technique , Gene Knockout Techniques , Hepatitis B/metabolism , Hepatitis B virus/genetics , Hepatitis B virus/metabolism , Hepatocytes/virology , Humans , Polymerase Chain Reaction , RNA, Small Interfering , Transfection , Virus Replication/genetics
8.
J Virol ; 90(19): 8866-74, 2016 10 01.
Article in English | MEDLINE | ID: mdl-27466423

ABSTRACT

UNLABELLED: Sodium taurocholate cotransporting polypeptide (NTCP) was identified as a functional receptor for hepatitis D virus (HDV) and its helper hepatitis B virus (HBV). In cultured cell lines, HDV infection through mouse NTCP is restricted by residues 84 to 87 of the receptor. This study shows that mice with these three amino acids altered their corresponding human residues (H84R, T86K, and S87N) in endogenous mouse NTCP support de novo HDV infection in vivo HDV infection was documented by the presence of replicative forms of HDV RNA and HDV proteins in liver cells at day 6 after viral inoculation. Monoclonal antibody specifically binding to the motif centered on K86 in NTCP partially inhibited HDV infection. These studies demonstrated specific interaction between the receptor and the viral envelopes in vivo and established a novel mouse model with minimal genetic manipulation for studying HDV infection. The model will also be useful for evaluating entry inhibitors against HDV and its helper HBV. IMPORTANCE: NTCP was identified as a functional receptor for both HDV and HBV in cell cultures. We recently showed that neonatal C57BL/6 transgenic (Tg) mice exogenously expressing human NTCP (hNTCP-Tg) in liver support transient HDV infection. In this study, we introduced alterations of three amino acids in the endogenous NTCP of FVB mice through genome editing. The mice with the humanized NTCP residues (H84R, T86K, and S87N) are susceptible to HDV infection, and the infection can be established in both neonatal and adult mice with this editing. We also developed a monoclonal antibody specifically targeting the region of NTCP centered on lysine residue 86, and it can differentiate the modified mouse NTCP from that of the wild type and partially inhibited HDV infection. These studies shed new light on NTCP-mediated HDV infection in vivo, and the NTCP-modified mice provide a useful animal model for studying HDV infection and evaluating antivirals against the infection.


Subject(s)
Amino Acid Substitution , Hepatitis D/virology , Hepatitis Delta Virus/physiology , Organic Anion Transporters, Sodium-Dependent/genetics , Organic Anion Transporters, Sodium-Dependent/metabolism , Receptors, Virus/genetics , Receptors, Virus/metabolism , Symporters/genetics , Symporters/metabolism , Animals , Disease Models, Animal , Disease Susceptibility , Mice , Mutant Proteins/genetics , Mutant Proteins/metabolism
9.
J Virol ; 89(14): 7202-13, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25926653

ABSTRACT

UNLABELLED: Human coronavirus (hCoV) HKU1 is one of six hCoVs identified to date and the only one with an unidentified cellular receptor. hCoV-HKU1 encodes a hemagglutinin-esterase (HE) protein that is unique to the group a betacoronaviruses (group 2a). The function of HKU1-HE remains largely undetermined. In this study, we examined binding of the S1 domain of hCoV-HKU1 spike to a panel of cells and found that the S1 could specifically bind on the cell surface of a human rhabdomyosarcoma cell line, RD. Pretreatment of RD cells with neuraminidase (NA) and trypsin greatly reduced the binding, suggesting that the binding was mediated by sialic acids on glycoproteins. However, unlike other group 2a CoVs, e.g., hCoV-OC43, for which 9-O-acetylated sialic acid (9-O-Ac-Sia) serves as a receptor determinant, HKU1-S1 bound with neither 9-O-Ac-Sia-containing glycoprotein(s) nor rat and mouse erythrocytes. Nonetheless, the HKU1-HE was similar to OC43-HE, also possessed sialate-O-acetylesterase activity, and acted as a receptor-destroying enzyme (RDE) capable of eliminating the binding of HKU1-S1 to RD cells, whereas the O-acetylesterase-inactive HKU1-HE mutant lost this capacity. Using primary human ciliated airway epithelial (HAE) cell cultures, the only in vitro replication model for hCoV-HKU1 infection, we confirmed that pretreatment of HAE cells with HE but not the enzymatically inactive mutant blocked hCoV-HKU1 infection. These results demonstrate that hCoV-HKU1 exploits O-Ac-Sia as a cellular attachment receptor determinant to initiate the infection of host cells and that its HE protein possesses the corresponding sialate-O-acetylesterase RDE activity. IMPORTANCE: Human coronaviruses (hCoV) are important human respiratory pathogens. Among the six hCoVs identified to date, only hCoV-HKU1 has no defined cellular receptor. It is also unclear whether hemagglutinin-esterase (HE) protein plays a role in viral entry. In this study, we found that, similarly to other members of the group 2a CoVs, sialic acid moieties on glycoproteins are critical receptor determinants for the hCoV-HKU1 infection. Interestingly, the virus seems to employ a type of sialic acid different from those employed by other group 2a CoVs. In addition, we determined that the HKU1-HE protein is an O-acetylesterase and acts as a receptor-destroying enzyme (RDE) for hCoV-HKU1. This is the first study to demonstrate that hCoV-HKU1 uses certain types of O-acetylated sialic acid residues on glycoproteins to initiate the infection of host cells and that the HKU1-HE protein possesses sialate-O-acetylesterase RDE activity.


Subject(s)
Coronavirus/physiology , Hemagglutinins, Viral/metabolism , N-Acetylneuraminic Acid/metabolism , Receptors, Virus/analysis , Spike Glycoprotein, Coronavirus/metabolism , Viral Fusion Proteins/metabolism , Virus Attachment , Cells, Cultured , Coronavirus/enzymology , Humans
10.
PLoS Pathog ; 11(4): e1004840, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25902143

ABSTRACT

Hepatitis D virus (HDV) is the smallest virus known to infect human. About 15 million people worldwide are infected by HDV among those 240 million infected by its helper hepatitis B virus (HBV). Viral hepatitis D is considered as one of the most severe forms of human viral hepatitis. No specific antivirals are currently available to treat HDV infection and antivirals against HBV do not ameliorate hepatitis D. Liver sodium taurocholate co-transporting polypeptide (NTCP) was recently identified as a common entry receptor for HDV and HBV in cell cultures. Here we show HDV can infect mice expressing human NTCP (hNTCP-Tg). Antibodies against critical regions of HBV envelope proteins blocked HDV infection in the hNTCP-Tg mice. The infection was acute yet HDV genome replication occurred efficiently, evident by the presence of antigenome RNA and edited RNA species specifying large delta antigen in the livers of infected mice. The resolution of HDV infection appears not dependent on adaptive immune response, but might be facilitated by innate immunity. Liver RNA-seq analyses of HDV infected hNTCP-Tg and type I interferon receptor 1 (IFNα/ßR1) null hNTCP-Tg mice indicated that in addition to induction of type I IFN response, HDV infection was also associated with up-regulation of novel cellular genes that may modulate HDV infection. Our work has thus proved the concept that NTCP is a functional receptor for HDV infection in vivo and established a convenient small animal model for investigation of HDV pathogenesis and evaluation of antiviral therapeutics against the early steps of infection for this important human pathogen.


Subject(s)
Hepatitis D/metabolism , Hepatitis Delta Virus/physiology , Hepatocytes/metabolism , Host-Pathogen Interactions , Organic Anion Transporters, Sodium-Dependent/metabolism , Symporters/metabolism , Animals , Antibodies, Monoclonal/pharmacology , Antibodies, Viral/pharmacology , Cells, Cultured , Crosses, Genetic , Female , Hepatitis D/drug therapy , Hepatitis D/pathology , Hepatitis D/virology , Hepatitis Delta Virus/drug effects , Hepatitis Delta Virus/immunology , Hepatocytes/drug effects , Hepatocytes/pathology , Hepatocytes/virology , Host-Pathogen Interactions/drug effects , Humans , Immunity, Innate/drug effects , Male , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Organic Anion Transporters, Sodium-Dependent/genetics , Receptor, Interferon alpha-beta/genetics , Receptor, Interferon alpha-beta/metabolism , Recombinant Fusion Proteins/metabolism , Specific Pathogen-Free Organisms , Symporters/genetics , Viral Envelope Proteins/antagonists & inhibitors , Viral Envelope Proteins/metabolism
11.
J Virol ; 88(1): 237-48, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24155382

ABSTRACT

Severe fever with thrombocytopenia syndrome virus (SFTSV) is a novel phlebovirus in the Bunyaviridae family. Most patients infected by SFTSV present with fever and thrombocytopenia, and up to 30% die due to multiple-organ dysfunction. The mechanisms by which SFTSV enters multiple cell types are unknown. SFTSV contains two species of envelope glycoproteins, Gn (44.2 kDa) and Gc (56 kDa), both of which are encoded by the M segment and are cleaved from a precursor polypeptide (about 116 kDa) in the endoplasmic reticulum (ER). Gn fused with an immunoglobulin Fc tag at its C terminus (Gn-Fc) bound to multiple cells susceptible to the infection of SFTSV and blocked viral infection of human umbilical vein endothelial cells (HUVECs). Immunoprecipitation assays following mass spectrometry analysis showed that Gn binds to nonmuscle myosin heavy chain IIA (NMMHC-IIA), a cellular protein with surface expression in multiple cell types. Small interfering RNA (siRNA) knockdown of NMMHC-IIA, but not the closely related NMMHC-IIB or NMMHC-IIC, reduced SFTSV infection, and NMMHC-IIA specific antibody blocked infection by SFTSV but not other control viruses. Overexpression of NMMHC-IIA in HeLa cells, which show limited susceptivity to SFTSV, markedly enhanced SFTSV infection of the cells. These results show that NMMHC-IIA is critical for the cellular entry of SFTSV. As NMMHC-IIA is essential for the normal functions of platelets and human vascular endothelial cells, it is conceivable that NMMHC-IIA directly contributes to the pathogenesis of SFTSV and may be a useful target for antiviral interventions against the viral infection.


Subject(s)
Fever/virology , Myosin Heavy Chains/physiology , Phlebovirus/pathogenicity , Thrombocytopenia/virology , Animals , Base Sequence , Blotting, Western , Cell Line , DNA Primers , Humans , Microscopy, Confocal , Myosin Heavy Chains/genetics , Polymerase Chain Reaction , RNA, Small Interfering/genetics
12.
PLoS Pathog ; 9(9): e1003613, 2013 Sep.
Article in English | MEDLINE | ID: mdl-24068929

ABSTRACT

Covalently closed circular DNA (cccDNA) of hepadnaviruses exists as an episomal minichromosome in the nucleus of infected hepatocyte and serves as the transcriptional template for viral mRNA synthesis. Elimination of cccDNA is the prerequisite for either a therapeutic cure or immunological resolution of HBV infection. Although accumulating evidence suggests that inflammatory cytokines-mediated cure of virally infected hepatocytes does occur and plays an essential role in the resolution of an acute HBV infection, the molecular mechanism by which the cytokines eliminate cccDNA and/or suppress its transcription remains elusive. This is largely due to the lack of convenient cell culture systems supporting efficient HBV infection and cccDNA formation to allow detailed molecular analyses. In this study, we took the advantage of a chicken hepatoma cell line that supports tetracycline-inducible duck hepatitis B virus (DHBV) replication and established an experimental condition mimicking the virally infected hepatocytes in which DHBV pregenomic (pg) RNA transcription and DNA replication are solely dependent on cccDNA. This cell culture system allowed us to demonstrate that cccDNA transcription required histone deacetylase activity and IFN-α induced a profound and long-lasting suppression of cccDNA transcription, which required protein synthesis and was associated with the reduction of acetylated histone H3 lysine 9 (H3K9) and 27 (H3K27) in cccDNA minichromosomes. Moreover, IFN-α treatment also induced a delayed response that appeared to accelerate the decay of cccDNA. Our studies have thus shed light on the molecular mechanism by which IFN-α noncytolytically controls hepadnavirus infection.


Subject(s)
DNA, Circular/metabolism , DNA, Viral/metabolism , Epigenesis, Genetic , Hepatitis B Virus, Duck/metabolism , Hepatocytes/virology , Interferon-alpha/metabolism , Transcription, Genetic , Acetylation/drug effects , Animals , Avian Proteins/antagonists & inhibitors , Avian Proteins/biosynthesis , Avian Proteins/metabolism , Cell Line , Chickens , Down-Regulation/drug effects , Epigenesis, Genetic/drug effects , Hepadnaviridae Infections/metabolism , Hepadnaviridae Infections/virology , Hepatitis B Virus, Duck/drug effects , Hepatocytes/drug effects , Hepatocytes/metabolism , Histone Deacetylase Inhibitors/pharmacology , Histone Deacetylases/chemistry , Histone Deacetylases/metabolism , Histones/metabolism , Lysine/chemistry , Protein Biosynthesis/drug effects , Protein Processing, Post-Translational/drug effects , Protein Synthesis Inhibitors/pharmacology , Transcription, Genetic/drug effects
13.
J Virol ; 87(14): 7977-91, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23678176

ABSTRACT

Human hepatitis B virus (HBV) and its satellite virus, hepatitis D virus (HDV), primarily infect humans, chimpanzees, or tree shrews (Tupaia belangeri). Viral infections in other species are known to be mainly restricted at the entry level since viral replication can be achieved in the cells by transfection of the viral genome. Sodium taurocholate cotransporting polypeptide (NTCP) is a functional receptor for HBV and HDV, and amino acids 157 to 165 of NTCP are critical for viral entry and likely limit viral infection of macaques. However, the molecular determinants for viral entry restriction in mouse NTCP (mNTCP) remain unclear. In this study, mNTCP was found to be unable to support either HBV or HDV infection, although it can bind to pre-S1 of HBV L protein and is functional in transporting substrate taurocholate; comprehensive swapping and point mutations of human NTCP (hNTCP) and mNTCP revealed molecular determinants restricting mNTCP for viral entry of HBV and HDV. Remarkably, when mNTCP residues 84 to 87 were substituted by human counterparts, mNTCP can effectively support viral infections. In addition, a number of cell lines, regardless of their species or tissue origin, supported HDV infection when transfected with hNTCP or mNTCP with residues 84 to 87 replaced by human counterparts, highlighting the central role of NTCP for viral infections mediated by HBV envelope proteins. These studies advance our understanding of NTCP-mediated viral entry of HBV and HDV and have important implications for developing the mouse model for their infections.


Subject(s)
Hepatitis B virus/metabolism , Hepatitis Delta Virus/metabolism , Organic Anion Transporters, Sodium-Dependent/metabolism , Symporters/metabolism , Virus Internalization , Animals , Cell Line, Tumor , Chlorocebus aethiops , Enzyme-Linked Immunosorbent Assay , Fluorescein-5-isothiocyanate , Hepatitis B Surface Antigens/metabolism , Humans , Mice , Microscopy, Fluorescence , Mutation/genetics , Organic Anion Transporters, Sodium-Dependent/genetics , Protein Precursors/metabolism , Symporters/genetics , Taurocholic Acid/metabolism , Tritium , Vero Cells
14.
J Virol ; 87(12): 7176-84, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23596296

ABSTRACT

Primary Tupaia hepatocytes (PTHs) are susceptible to woolly monkey hepatitis B virus (WMHBV) infection, but the identity of the cellular receptor(s) mediating WMHBV infection of PTHs remains unclear. Recently, sodium taurocholate cotransporting polypeptide (NTCP) was identified as a functional receptor for human hepatitis B virus (HBV) infection of primary human and Tupaia hepatocytes. In this study, a synthetic pre-S1 peptide from WMHBV was found to bind specifically to cells expressing Tupaia NTCP (tsNTCP) and it efficiently blocked WMHBV entry into PTHs; silencing of tsNTCP in PTHs significantly inhibited WMHBV infection. Ectopic expression of tsNTCP rendered HepG2 cells susceptible to WMHBV infection. These data demonstrate that tsNTCP is a functional receptor for WMHBV infection of PTHs. The result also indicates that NTCP's orthologs likely act as a common cellular receptor for all known primate hepadnaviruses.


Subject(s)
Atelinae/virology , Hepadnaviridae/pathogenicity , Hepatocytes/virology , Organic Anion Transporters, Sodium-Dependent/metabolism , Receptors, Virus/metabolism , Symporters/metabolism , Tupaia/virology , Amino Acid Sequence , Animals , Cells, Cultured , Hepadnaviridae/genetics , Hepadnaviridae/metabolism , Hepadnaviridae Infections/virology , Hepatitis B Surface Antigens/chemistry , Hepatitis B Surface Antigens/genetics , Hepatitis B Surface Antigens/metabolism , Humans , Molecular Sequence Data , Protein Precursors/chemistry , Protein Precursors/genetics , Protein Precursors/metabolism
15.
Elife ; 1: e00049, 2012 Nov 13.
Article in English | MEDLINE | ID: mdl-23150796

ABSTRACT

Human hepatitis B virus (HBV) infection and HBV-related diseases remain a major public health problem. Individuals coinfected with its satellite hepatitis D virus (HDV) have more severe disease. Cellular entry of both viruses is mediated by HBV envelope proteins. The pre-S1 domain of the large envelope protein is a key determinant for receptor(s) binding. However, the identity of the receptor(s) is unknown. Here, by using near zero distance photo-cross-linking and tandem affinity purification, we revealed that the receptor-binding region of pre-S1 specifically interacts with sodium taurocholate cotransporting polypeptide (NTCP), a multiple transmembrane transporter predominantly expressed in the liver. Silencing NTCP inhibited HBV and HDV infection, while exogenous NTCP expression rendered nonsusceptible hepatocarcinoma cells susceptible to these viral infections. Moreover, replacing amino acids 157-165 of nonfunctional monkey NTCP with the human counterpart conferred its ability in supporting both viral infections. Our results demonstrate that NTCP is a functional receptor for HBV and HDV.DOI:http://dx.doi.org/10.7554/eLife.00049.001.


Subject(s)
Hepatitis B virus/metabolism , Hepatitis Delta Virus/metabolism , Hepatocytes/metabolism , Organic Anion Transporters, Sodium-Dependent/metabolism , Receptors, Virus/metabolism , Symporters/metabolism , Viral Envelope Proteins/metabolism , Amino Acid Sequence , Animals , Biological Transport , Cell Line , Gene Expression , Hepatitis B virus/chemistry , Hepatitis B virus/genetics , Hepatitis Delta Virus/chemistry , Hepatitis Delta Virus/genetics , Hepatocytes/pathology , Hepatocytes/virology , Humans , Liver/metabolism , Liver/pathology , Liver/virology , Molecular Sequence Data , Organic Anion Transporters, Sodium-Dependent/chemistry , Organic Anion Transporters, Sodium-Dependent/genetics , Peptides/chemistry , Photochemical Processes , Primary Cell Culture , Protein Binding , Protein Structure, Tertiary , Receptors, Virus/chemistry , Receptors, Virus/genetics , Symporters/chemistry , Symporters/genetics , Taurocholic Acid/metabolism , Tupaia , Viral Envelope Proteins/chemistry , Viral Envelope Proteins/genetics , Virus Internalization
16.
J Biol Chem ; 287(9): 6406-20, 2012 Feb 24.
Article in English | MEDLINE | ID: mdl-22219187

ABSTRACT

Enterovirus 71 (EV71) is one of the major pathogens that cause hand, foot, and mouth disease outbreaks in young children in the Asia-Pacific region in recent years. Human scavenger receptor class B 2 (SCARB2) is the main cellular receptor for EV71 on target cells. The requirements of the EV71-SCARB2 interaction have not been fully characterized, and it has not been determined whether SCARB2 serves as an uncoating receptor for EV71. Here we compared the efficiency of the receptor from different species including human, horseshoe bat, mouse, and hamster and demonstrated that the residues between 144 and 151 are critical for SCARB2 binding to viral capsid protein VP1 of EV71 and seven residues from the human receptor could convert murine SCARB2, an otherwise inefficient receptor, to an efficient receptor for EV71 viral infection. We also identified that EV71 binds to SCARB2 via a canyon of VP1 around residue Gln-172. Soluble SCARB2 could convert the EV71 virions from 160 S to 135 S particles, indicating that SCARB2 is an uncoating receptor of the virus. The uncoating efficiency of SCARB2 significantly increased in an acidic environment (pH 5.6). These studies elucidated the viral capsid and receptor determinants of enterovirus 71 infection and revealed a possible target for antiviral interventions.


Subject(s)
CD36 Antigens/metabolism , Enterovirus A, Human/growth & development , Enterovirus Infections/virology , Lysosomal Membrane Proteins/metabolism , Receptors, Scavenger/metabolism , Viral Fusion Proteins/metabolism , Animals , CD36 Antigens/chemistry , CD36 Antigens/genetics , Cell Line, Tumor , Chiroptera , Cricetinae , Enterovirus A, Human/genetics , Enterovirus Infections/metabolism , Gene Knockdown Techniques , Green Fluorescent Proteins/genetics , HEK293 Cells , Humans , Kidney/cytology , Lysosomal Membrane Proteins/chemistry , Lysosomal Membrane Proteins/genetics , Mice , Protein Structure, Tertiary , Receptors, Scavenger/chemistry , Receptors, Scavenger/genetics , Rhabdomyosarcoma , Viral Fusion Proteins/genetics
17.
Elife ; 32012 Nov 13.
Article in English | MEDLINE | ID: mdl-25409679

ABSTRACT

Human hepatitis B virus (HBV) infection and HBV-related diseases remain a major public health problem. Individuals coinfected with its satellite hepatitis D virus (HDV) have more severe disease. Cellular entry of both viruses is mediated by HBV envelope proteins. The pre-S1 domain of the large envelope protein is a key determinant for receptor(s) binding. However, the identity of the receptor(s) is unknown. Here, by using near zero distance photo-cross-linking and tandem affinity purification, we revealed that the receptor-binding region of pre-S1 specifically interacts with sodium taurocholate cotransporting polypeptide (NTCP), a multiple transmembrane transporter predominantly expressed in the liver. Silencing NTCP inhibited HBV and HDV infection, while exogenous NTCP expression rendered nonsusceptible hepatocarcinoma cells susceptible to these viral infections. Moreover, replacing amino acids 157-165 of nonfunctional monkey NTCP with the human counterpart conferred its ability in supporting both viral infections. Our results demonstrate that NTCP is a functional receptor for HBV and HDV.


Subject(s)
Hepatitis B virus/metabolism , Hepatitis Delta Virus/metabolism , Organic Anion Transporters, Sodium-Dependent/metabolism , Receptors, Virus/metabolism , Symporters/metabolism , Amino Acid Sequence , Animals , Cell Line, Tumor , Cells, Cultured , Disease Susceptibility , Hepatitis B/pathology , Hepatitis B/virology , Hepatitis D/pathology , Hepatitis D/virology , Hepatocytes/metabolism , Hepatocytes/pathology , Hepatocytes/virology , Humans , Ligands , Molecular Sequence Data , Organic Anion Transporters, Sodium-Dependent/chemistry , Protein Binding , Protein Structure, Tertiary , Receptors, Virus/chemistry , Reproducibility of Results , Symporters/chemistry , Tupaiidae , Virion/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...