Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 28
Filter
Add more filters










Publication year range
1.
J Med Chem ; 67(9): 7260-7275, 2024 May 09.
Article in English | MEDLINE | ID: mdl-38651218

ABSTRACT

Artificial intelligence (AI) de novo molecular generation is a highly promising strategy in the drug discovery, with deep reinforcement learning (RL) models emerging as powerful tools. This study introduces a fragment-by-fragment growth RL forward molecular generation and optimization strategy based on a low activity lead compound. This process integrates fragment growth-based reaction templates, while target docking and drug-likeness prediction were simultaneously performed. This comprehensive approach considers molecular similarity, internal diversity, synthesizability, and effectiveness, thereby enhancing the quality and efficiency of molecular generation. Finally, a series of tyrosinase inhibitors were generated and synthesized. Most compounds exhibited more improved activity than lead, with an optimal candidate compound surpassing the effects of kojic acid and demonstrating significant antipigmentation activity in a zebrafish model. Furthermore, metabolic stability studies indicated susceptibility to hepatic metabolism. The proposed AI structural optimization strategies will play a promising role in accelerating the drug discovery and improving traditional efficiency.


Subject(s)
Artificial Intelligence , Enzyme Inhibitors , Monophenol Monooxygenase , Zebrafish , Animals , Monophenol Monooxygenase/antagonists & inhibitors , Monophenol Monooxygenase/metabolism , Enzyme Inhibitors/pharmacology , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/chemical synthesis , Molecular Docking Simulation , Structure-Activity Relationship , Molecular Structure , Humans , Drug Discovery
2.
Eur J Med Chem ; 267: 116152, 2024 Mar 05.
Article in English | MEDLINE | ID: mdl-38278079

ABSTRACT

Photodamage is the result of prolonged exposure of the skin to sunlight. This exposure causes an overexpression of matrix metalloproteinases (MMPs), leading to the abnormal degradation of collagen in the skin tissue and resulting in skin aging and damage. This review presents a detailed overview of MMPs as a potential target for addressing skin aging. Specifically, we elucidated the precise mechanisms by which MMP inhibitors exert their anti-photoaging effects. Furthermore, we comprehensively analyzed the current research progress on MMP inhibitors that demonstrate significant inhibitory activity against MMPs and anti-skin photoaging effects. The review also provides insights into the structure-activity relationships of these inhibitors. Our objective in conducting this review is to provide valuable practical information to researchers engaged in investigations on anti-skin photoaging.


Subject(s)
Skin Aging , Matrix Metalloproteinase Inhibitors/pharmacology , Ultraviolet Rays , Skin , Matrix Metalloproteinases/metabolism
3.
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
4.
J Virol ; 97(7): e0032823, 2023 07 27.
Article in English | MEDLINE | ID: mdl-37338350

ABSTRACT

Hepatitis B virus (HBV) chronically infects approximately 300 million people worldwide, and permanently repressing transcription of covalently closed circular DNA (cccDNA), the episomal viral DNA reservoir, is an attractive approach toward curing HBV. However, the mechanism underlying cccDNA transcription is only partially understood. In this study, by illuminating cccDNA of wild-type HBV (HBV-WT) and transcriptionally inactive HBV that bears a deficient HBV X gene (HBV-ΔX), we found that the HBV-ΔX cccDNA more frequently colocalizes with promyelocytic leukemia (PML) bodies than that of HBV-WT cccDNA. A small interfering RNA (siRNA) screen targeting 91 PML body-related proteins identified SMC5-SMC6 localization factor 2 (SLF2) as a host restriction factor of cccDNA transcription, and subsequent studies showed that SLF2 mediates HBV cccDNA entrapment in PML bodies by interacting with the SMC5/6 complex. We further showed that the region of SLF2 comprising residues 590 to 710 interacts with and recruits the SMC5/6 complex to PML bodies, and the C-terminal domain of SLF2 containing this region is necessary for repression of cccDNA transcription. Our findings shed new light on cellular mechanisms that inhibit HBV infection and lend further support for targeting the HBx pathway to repress HBV activity. IMPORTANCE Chronic HBV infection remains a major public health problem worldwide. Current antiviral treatments rarely cure the infection, as they cannot clear the viral reservoir, cccDNA, in the nucleus. Therefore, permanently silencing HBV cccDNA transcription represents a promising approach for a cure of HBV infection. Our study provides new insights into the cellular mechanisms that restrict HBV infection, revealing the role of SLF2 in directing HBV cccDNA to PML bodies for transcriptional repression. These findings have important implications for the development of antiviral therapies against HBV.


Subject(s)
Hepatitis B , Leukemia , Humans , Hepatitis B virus/genetics , Hepatitis B virus/metabolism , DNA, Circular/genetics , DNA, Circular/metabolism , Antiviral Agents/pharmacology , DNA, Viral/genetics , DNA, Viral/metabolism , Promyelocytic Leukemia Protein/genetics , Promyelocytic Leukemia Protein/metabolism , Virus Replication/genetics , Chromosomal Proteins, Non-Histone/metabolism , Cell Cycle Proteins/metabolism
5.
Antiviral Res ; 216: 105642, 2023 08.
Article in English | MEDLINE | ID: mdl-37253400

ABSTRACT

Covalently closed circular (ccc) DNA is the template for hepatitis B virus (HBV) replication. The lack of small animal models for characterizing chronic HBV infection has hampered research progress in HBV pathogenesis and drug development. Here, we generated a spatiotemporally controlled recombinant cccDNA (rcccDNA) mouse model by combining Cre/loxP-mediated DNA recombination with the liver-specific "Tet-on/Cre" system. The mouse model harbors three transgenes: a single copy of the HBV genome (integrated at the Rosa26 locus, RHBV), H11-albumin-rtTA (spatiotemporal conditional module), and (tetO)7-Cre (tetracycline response element), and is named as RHTC mouse. By supplying the RHTC mice with doxycycline (DOX)-containing drinking water for two days, the animals generate rcccDNA in hepatocytes, and the rcccDNA supports active HBV gene expression and can maintain HBV viremia persistence for over 60 weeks. Persistent HBV gene expression induces intrahepatic inflammation, fibrosis, and dysplastic pathology, which closely mirrors the disease progression in clinical patients. Bepirovirsen, an antisense oligonucleotide (ASO) targeting all HBV RNA species, showed dose-dependent antiviral effects in the RHTC mouse model. The spatiotemporally controlled rcccDNA mouse is convenient and reliable, providing versatile small animal model for studying cccDNA-centric HBV biology as well as evaluating antiviral therapeutics.


Subject(s)
Hepatitis B, Chronic , Hepatitis B , Mice , Animals , Hepatitis B virus/physiology , DNA, Viral/genetics , DNA, Viral/metabolism , Hepatitis B, Chronic/genetics , DNA, Circular/genetics , DNA, Circular/metabolism , Antiviral Agents/therapeutic use , Disease Models, Animal , Virus Replication , Hepatitis B/drug therapy
6.
J Virol ; 97(1): e0171722, 2023 01 31.
Article in English | MEDLINE | ID: mdl-36475867

ABSTRACT

Hepatitis B virus (HBV) infection remains a public health problem worldwide. Persistent HBV infection relies on active transcription of the covalently closed circular DNA (cccDNA) in hepatocytes, which is less understood at the single-cell level. In this study, we isolated primary human hepatocytes from liver-humanized FRG mice infected with HBV and examined cccDNA transcripts in single cells based on 5' end sequencing. Our 5' transcriptome sequencing (RNA-seq) analysis unambiguously assigns different viral transcripts with overlapping 3' sequences and quantitatively measures viral transcripts for structural genes (3.5 kb, 2.4 kb, and 2.1 kb) and the nonstructural X gene (0.7 kb and related) in single cells. We found that an infected cell either can generate all viral transcripts, signifying active transcription, or presents only transcripts from the X gene and its associated enhancer I domain and no structural gene transcripts. Results from cell infection assays with recombinant HBV show that nonproductive transcription of cccDNA can be activated by incoming virus through superinfection. Moreover, upon HBV infection, cccDNA apparently can be transcribed in the absence of HBx and produces HBx, needed for productive transcription of other viral genes. These results shed new light on cccDNA transcription at the single-cell level and provide insights useful for improving the treatment strategy against chronic HBV infection. IMPORTANCE Hepatitis B virus (HBV) infection can be effectively suppressed but rarely cured by available drugs. Chronic HBV infection is based on persistence of covalently closed circular DNA (cccDNA) and continuous infection and reinfection with HBV in the liver. Understanding transcriptional regulation of cccDNA will help to achieve permanent transcriptional silencing, i.e., functional cure of HBV. In our study, we found that an infected cell either can generate all viral transcripts, signifying active transcription, or presents only transcripts from the X gene and its associated enhancer I domain and no structural gene transcripts. The nonproductive transcription of cccDNA can be activated by incoming virus through superinfection. Upon an infection, cccDNA apparently can be transcribed in the absence of HBx to produce HBx, necessary for subsequent transcription of other HBV genes. Our studies shed new light on the mechanism of HBV infection and may have implications for a functional cure regimen for HBV.


Subject(s)
DNA, Circular , Hepatitis B, Chronic , Superinfection , Animals , Humans , Mice , DNA, Circular/genetics , DNA, Viral/genetics , Hepatitis B virus/genetics , Hepatitis B, Chronic/genetics , Virus Replication/genetics , Hepatocytes , Viral Regulatory and Accessory Proteins/genetics
7.
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
8.
J Virol ; 95(11)2021 05 10.
Article in English | MEDLINE | ID: mdl-33692215

ABSTRACT

Within a year after its emergence, the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has infected over 100 million people worldwide with a death toll over 2 million. Vaccination remains the best hope to ultimately put this pandemic to an end. Here, using Trimer-Tag technology, we produced both wild-type (WT) and furin site mutant (MT) S-Trimers for COVID-19 vaccine studies. Cryo-EM structures of the WT and MT S-Trimers, determined at 3.2 Å and 2.6 Å respectively, revealed that both antigens adopt a tightly closed conformation and their structures are essentially identical to that of the previously solved full-length WT S protein in detergent. The tightly closed conformation is stabilized by fatty acid and polysorbate 80 binding at the receptor binding domains (RBDs) and the N terminal domains (NTDs) respectively. Additionally, we identified an important pH switch in the WT S-Trimer that shows dramatic conformational change and accounts for its increased stability at lower pH. These results validate Trimer-Tag as a platform technology in production of metastable WT S-Trimer as a candidate for COVID-19 subunit vaccine.IMPORTANCEEffective vaccine against SARS-CoV-2 is critical to end the COVID-19 pandemic. Here, using Trimer-Tag technology, we are able to produce stable and large quantities of WT S-Trimer, a subunit vaccine candidate for COVID-19 with high safety and efficacy from animal and Phase 1 clinical trial studies. Cryo-EM structures of the S-Trimer subunit vaccine candidate show that it predominately adopts tightly closed pre-fusion state, and resembles that of the native and full-length spike in detergent, confirming its structural integrity. WT S-Trimer is currently being evaluated in global Phase 2/3 clinical trial. Combining with published structures of the S protein, we also propose a model to dissect the conformation change of the spike protein before receptor binding.

9.
Nat Microbiol ; 4(3): 429-437, 2019 03.
Article in English | MEDLINE | ID: mdl-30617349

ABSTRACT

Severe fever with thrombocytopenia syndrome phlebovirus (SFTSV), listed in the World Health Organization Prioritized Pathogens, is an emerging phlebovirus with a high fatality1-4. Owing to the lack of therapies and vaccines5,6, there is a pressing need to understand SFTSV pathogenesis. SFSTV non-structural protein (NSs) has been shown to block type I interferon induction7-11 and facilitate disease progression12,13. Here, we report that SFTSV-NSs targets the tumour progression locus 2 (TPL2)-A20-binding inhibitor of NF-κB activation 2 (ABIN2)-p105 complex to induce the expression of interleukin-10 (IL-10) for viral pathogenesis. Using a combination of reverse genetics, a TPL2 kinase inhibitor and Tpl2-/- mice showed that NSs interacted with ABIN2 and promoted TPL2 complex formation and signalling activity, resulting in the marked upregulation of Il10 expression. Whereas SFTSV infection of wild-type mice led to rapid weight loss and death, Tpl2-/- mice or Il10-/- mice survived an infection. Furthermore, SFTSV-NSs P102A and SFTSV-NSs K211R that lost the ability to induce TPL2 signalling and IL-10 production showed drastically reduced pathogenesis. Remarkably, the exogenous administration of recombinant IL-10 effectively rescued the attenuated pathogenic activity of SFTSV-NSs P102A, resulting in a lethal infection. Our study demonstrates that SFTSV-NSs targets the TPL2 signalling pathway to induce immune-suppressive IL-10 cytokine production as a means to dampen the host defence and promote viral pathogenesis.


Subject(s)
Host-Pathogen Interactions , MAP Kinase Kinase Kinases/metabolism , Phlebovirus/pathogenicity , Proto-Oncogene Proteins/metabolism , Signal Transduction , Viral Nonstructural Proteins/genetics , Adaptor Proteins, Signal Transducing , Animals , Bunyaviridae Infections/immunology , Bunyaviridae Infections/pathology , Female , HEK293 Cells , HeLa Cells , Humans , Interleukin-10/administration & dosage , Interleukin-10/genetics , MAP Kinase Kinase Kinases/immunology , Male , Mice , Mice, 129 Strain , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Phlebovirus/drug effects , Proto-Oncogene Proteins/immunology , RAW 264.7 Cells , Reverse Genetics
10.
J Virol ; 92(8)2018 04 15.
Article in English | MEDLINE | ID: mdl-29437960

ABSTRACT

Multiple steps of the life cycle of hepatitis B virus (HBV) are known to be coupled to hepatic metabolism. However, the details of involvement of the hepatic metabolic milieu in HBV infection remain incompletely understood. Hepatic lipid metabolism is controlled by a complicated transcription factor network centered on retinoid X receptor alpha (RXRα). Here, we report that RXRα negatively regulates HBV infection at an early stage in cell cultures. The RXR-specific agonist bexarotene inhibits HBV in HepG2 cells expressing the sodium taurocholate cotransporting polypeptide (NTCP) (HepG2-NTCP), HepaRG cells, and primary Tupaia hepatocytes (PTHs); reducing RXRα expression significantly enhanced HBV infection in the cells. Transcriptome sequencing (RNA-seq) analysis of HepG2-NTCP cells with a disrupted RXRα gene revealed that reduced gene expression in arachidonic acid (AA)/eicosanoid biosynthesis pathways, including the AA synthases phospholipase A2 group IIA (PLA2G2A), is associated with increased HBV infection. Moreover, exogenous treatment of AA inhibits HBV infection in HepG2-NTCP cells. These data demonstrate that RXRα is an important cellular factor in modulating HBV infection and implicate the participation of AA/eicosanoid biosynthesis pathways in the regulation of HBV infection.IMPORTANCE Understanding how HBV infection is connected with hepatic lipid metabolism may provide new insights into virus infection and its pathogenesis. By a series of genetic studies in combination with transcriptome analysis and pharmacological assays, we here investigated the role of cellular retinoid X receptor alpha (RXRα), a crucial transcription factor for controlling hepatic lipid metabolism, in de novo HBV infection in cell cultures. We found that silencing of RXRα resulted in elevated HBV covalently closed circular DNA (cccDNA) formation and viral antigen production, while activation of RXRα reduced HBV infection efficiency. Our results also showed that silencing phospholipase A2 group IIA (PLA2G2A), a key enzyme of arachidonic acid (AA) synthases, enhanced HBV infection efficiency in HepG2-NTCP cells and that exogenous AA treatment reduced de novo HBV infection in the cells. These findings unveil RXRα as an important cellular factor in modulating HBV infection and may point to a new strategy for host-targeted therapies against HBV.


Subject(s)
Gene Expression Regulation , Hepatitis B virus/metabolism , Hepatitis B/metabolism , Retinoid X Receptor alpha/biosynthesis , Animals , Hep G2 Cells , Hepatitis B/genetics , Humans , Tupaia
11.
J Infect Dis ; 218(2): 197-207, 2018 06 20.
Article in English | MEDLINE | ID: mdl-29346682

ABSTRACT

Although bats are known to harbor Middle East Respiratory Syndrome coronavirus (MERS-CoV)-related viruses, the role of bats in the evolutionary origin and pathway remains obscure. We identified a novel MERS-CoV-related betacoronavirus, Hp-BatCoV HKU25, from Chinese pipistrelle bats. Although it is closely related to MERS-CoV in most genome regions, its spike protein occupies a phylogenetic position between that of Ty-BatCoV HKU4 and Pi-BatCoV HKU5. Because Ty-BatCoV HKU4 but not Pi-BatCoV HKU5 can use the MERS-CoV receptor human dipeptidyl peptidase 4 (hDPP4) for cell entry, we tested the ability of Hp-BatCoV HKU25 to bind and use hDPP4. The HKU25-receptor binding domain (RBD) can bind to hDPP4 protein and hDPP4-expressing cells, but it does so with lower efficiency than that of MERS-RBD. Pseudovirus assays showed that HKU25-spike can use hDPP4 for entry to hDPP4-expressing cells, although with lower efficiency than that of MERS-spike and HKU4-spike. Our findings support a bat origin of MERS-CoV and suggest that bat CoV spike proteins may have evolved in a stepwise manner for binding to hDPP4.


Subject(s)
Betacoronavirus/physiology , Chiroptera , Dipeptidyl Peptidase 4/metabolism , Evolution, Molecular , Receptors, Virus/metabolism , Spike Glycoprotein, Coronavirus/metabolism , Virus Internalization , Animals , Betacoronavirus/classification , Betacoronavirus/genetics , Betacoronavirus/isolation & purification , HEK293 Cells , Humans , Phylogeny , Protein Binding , Sequence Analysis, DNA , Spike Glycoprotein, Coronavirus/genetics
12.
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
13.
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
14.
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
15.
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
16.
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
17.
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
18.
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
19.
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
20.
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
...