Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
1.
Mol Ther ; 29(4): 1572-1584, 2021 04 07.
Artigo em Inglês | MEDLINE | ID: mdl-33429083

RESUMO

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.


Assuntos
Antígeno CD47/genética , Carcinoma Hepatocelular/tratamento farmacológico , Glipicanas/genética , Neoplasias Hepáticas/tratamento farmacológico , Animais , Anticorpos Biespecíficos/farmacologia , Anticorpos Monoclonais/farmacologia , Antígeno CD47/antagonistas & inibidores , Antígeno CD47/imunologia , Carcinoma Hepatocelular/genética , Carcinoma Hepatocelular/imunologia , Carcinoma Hepatocelular/patologia , Linhagem Celular Tumoral , Glipicanas/antagonistas & inibidores , Glipicanas/imunologia , Humanos , Imunidade Inata/efeitos dos fármacos , Imunidade Inata/imunologia , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/imunologia , Neoplasias Hepáticas/patologia , Camundongos , Ensaios Antitumorais Modelo de Xenoenxerto
2.
PLoS Pathog ; 13(9): e1006658, 2017 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-28945802

RESUMO

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.


Assuntos
DNA Circular/biossíntese , Vírus da Hepatite B/fisiologia , Hepatite B Crônica/virologia , Replicação Viral/efeitos dos fármacos , Antivirais/farmacologia , Linhagem Celular , DNA Circular/genética , DNA Viral , DNA Polimerase Dirigida por DNA/metabolismo , Hepatócitos/virologia , Humanos , Nucleocapsídeo/efeitos dos fármacos , Nucleocapsídeo/genética , Reação em Cadeia da Polimerase em Tempo Real , Replicação Viral/fisiologia
3.
PLoS Pathog ; 12(10): e1005893, 2016 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-27783675

RESUMO

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.


Assuntos
DNA Circular/genética , DNA Viral/genética , DNA Polimerase Dirigida por DNA/metabolismo , Hepatite B/genética , Southern Blotting , Linhagem Celular , DNA Circular/metabolismo , DNA Viral/metabolismo , Ensaio de Imunoadsorção Enzimática , Imunofluorescência , Técnicas de Inativação de Genes , Hepatite B/metabolismo , Vírus da Hepatite B/genética , Vírus da Hepatite B/metabolismo , Hepatócitos/virologia , Humanos , Reação em Cadeia da Polimerase , RNA Interferente Pequeno , Transfecção , Replicação Viral/genética
4.
J Virol ; 90(19): 8866-74, 2016 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-27466423

RESUMO

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.


Assuntos
Substituição de Aminoácidos , Hepatite D/virologia , Vírus Delta da Hepatite/fisiologia , Transportadores de Ânions Orgânicos Dependentes de Sódio/genética , Transportadores de Ânions Orgânicos Dependentes de Sódio/metabolismo , Receptores Virais/genética , Receptores Virais/metabolismo , Simportadores/genética , Simportadores/metabolismo , Animais , Modelos Animais de Doenças , Suscetibilidade a Doenças , Camundongos , Proteínas Mutantes/genética , Proteínas Mutantes/metabolismo
5.
PLoS Pathog ; 11(4): e1004840, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25902143

RESUMO

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.


Assuntos
Hepatite D/metabolismo , Vírus Delta da Hepatite/fisiologia , Hepatócitos/metabolismo , Interações Hospedeiro-Patógeno , Transportadores de Ânions Orgânicos Dependentes de Sódio/metabolismo , Simportadores/metabolismo , Animais , Anticorpos Monoclonais/farmacologia , Anticorpos Antivirais/farmacologia , Células Cultivadas , Cruzamentos Genéticos , Feminino , Hepatite D/tratamento farmacológico , Hepatite D/patologia , Hepatite D/virologia , Vírus Delta da Hepatite/efeitos dos fármacos , Vírus Delta da Hepatite/imunologia , Hepatócitos/efeitos dos fármacos , Hepatócitos/patologia , Hepatócitos/virologia , Interações Hospedeiro-Patógeno/efeitos dos fármacos , Humanos , Imunidade Inata/efeitos dos fármacos , Masculino , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Transportadores de Ânions Orgânicos Dependentes de Sódio/genética , Receptor de Interferon alfa e beta/genética , Receptor de Interferon alfa e beta/metabolismo , Proteínas Recombinantes de Fusão/metabolismo , Organismos Livres de Patógenos Específicos , Simportadores/genética , Proteínas do Envelope Viral/antagonistas & inibidores , Proteínas do Envelope Viral/metabolismo
6.
J Virol ; 89(14): 7202-13, 2015 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-25926653

RESUMO

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.


Assuntos
Coronavirus/fisiologia , Hemaglutininas Virais/metabolismo , Ácido N-Acetilneuramínico/metabolismo , Receptores Virais/análise , Glicoproteína da Espícula de Coronavírus/metabolismo , Proteínas Virais de Fusão/metabolismo , Ligação Viral , Células Cultivadas , Coronavirus/enzimologia , Humanos
7.
J Virol ; 88(1): 237-48, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-24155382

RESUMO

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.


Assuntos
Febre/virologia , Cadeias Pesadas de Miosina/fisiologia , Phlebovirus/patogenicidade , Trombocitopenia/virologia , Animais , Sequência de Bases , Western Blotting , Linhagem Celular , Primers do DNA , Humanos , Microscopia Confocal , Cadeias Pesadas de Miosina/genética , Reação em Cadeia da Polimerase , RNA Interferente Pequeno/genética
8.
PLoS Pathog ; 9(9): e1003613, 2013 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-24068929

RESUMO

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.


Assuntos
DNA Circular/metabolismo , DNA Viral/metabolismo , Epigênese Genética , Vírus da Hepatite B do Pato/metabolismo , Hepatócitos/virologia , Interferon-alfa/metabolismo , Transcrição Gênica , Acetilação/efeitos dos fármacos , Animais , Proteínas Aviárias/antagonistas & inibidores , Proteínas Aviárias/biossíntese , Proteínas Aviárias/metabolismo , Linhagem Celular , Galinhas , Regulação para Baixo/efeitos dos fármacos , Epigênese Genética/efeitos dos fármacos , Infecções por Hepadnaviridae/metabolismo , Infecções por Hepadnaviridae/virologia , Vírus da Hepatite B do Pato/efeitos dos fármacos , Hepatócitos/efeitos dos fármacos , Hepatócitos/metabolismo , Inibidores de Histona Desacetilases/farmacologia , Histona Desacetilases/química , Histona Desacetilases/metabolismo , Histonas/metabolismo , Lisina/química , Biossíntese de Proteínas/efeitos dos fármacos , Processamento de Proteína Pós-Traducional/efeitos dos fármacos , Inibidores da Síntese de Proteínas/farmacologia , Transcrição Gênica/efeitos dos fármacos
9.
J Virol ; 87(14): 7977-91, 2013 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-23678176

RESUMO

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.


Assuntos
Vírus da Hepatite B/metabolismo , Vírus Delta da Hepatite/metabolismo , Transportadores de Ânions Orgânicos Dependentes de Sódio/metabolismo , Simportadores/metabolismo , Internalização do Vírus , Animais , Linhagem Celular Tumoral , Chlorocebus aethiops , Ensaio de Imunoadsorção Enzimática , Fluoresceína-5-Isotiocianato , Antígenos de Superfície da Hepatite B/metabolismo , Humanos , Camundongos , Microscopia de Fluorescência , Mutação/genética , Transportadores de Ânions Orgânicos Dependentes de Sódio/genética , Precursores de Proteínas/metabolismo , Simportadores/genética , Ácido Taurocólico/metabolismo , Trítio , Células Vero
10.
J Virol ; 87(12): 7176-84, 2013 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-23596296

RESUMO

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.


Assuntos
Atelinae/virologia , Hepadnaviridae/patogenicidade , Hepatócitos/virologia , Transportadores de Ânions Orgânicos Dependentes de Sódio/metabolismo , Receptores Virais/metabolismo , Simportadores/metabolismo , Tupaia/virologia , Sequência de Aminoácidos , Animais , Células Cultivadas , Hepadnaviridae/genética , Hepadnaviridae/metabolismo , Infecções por Hepadnaviridae/virologia , Antígenos de Superfície da Hepatite B/química , Antígenos de Superfície da Hepatite B/genética , Antígenos de Superfície da Hepatite B/metabolismo , Humanos , Dados de Sequência Molecular , Precursores de Proteínas/química , Precursores de Proteínas/genética , Precursores de Proteínas/metabolismo
11.
J Biol Chem ; 287(9): 6406-20, 2012 Feb 24.
Artigo em Inglês | MEDLINE | ID: mdl-22219187

RESUMO

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.


Assuntos
Antígenos CD36/metabolismo , Enterovirus Humano A/crescimento & desenvolvimento , Infecções por Enterovirus/virologia , Proteínas de Membrana Lisossomal/metabolismo , Receptores Depuradores/metabolismo , Proteínas Virais de Fusão/metabolismo , Animais , Antígenos CD36/química , Antígenos CD36/genética , Linhagem Celular Tumoral , Quirópteros , Cricetinae , Enterovirus Humano A/genética , Infecções por Enterovirus/metabolismo , Técnicas de Silenciamento de Genes , Proteínas de Fluorescência Verde/genética , Células HEK293 , Humanos , Rim/citologia , Proteínas de Membrana Lisossomal/química , Proteínas de Membrana Lisossomal/genética , Camundongos , Estrutura Terciária de Proteína , Receptores Depuradores/química , Receptores Depuradores/genética , Rabdomiossarcoma , Proteínas Virais de Fusão/genética
12.
Nat Commun ; 14(1): 5191, 2023 08 25.
Artigo em Inglês | MEDLINE | ID: mdl-37626079

RESUMO

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.


Assuntos
Enzima de Conversão de Angiotensina 2 , COVID-19 , Masculino , Animais , Cricetinae , Humanos , Vacinas contra COVID-19 , SARS-CoV-2/genética , Mesocricetus , Imunoglobulina M
13.
Cell Rep ; 35(13): 109288, 2021 06 29.
Artigo em Inglês | MEDLINE | ID: mdl-34192543

RESUMO

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.


Assuntos
Cromossomos Humanos Par 19/genética , Genoma Humano , Vírus da Hepatite B/genética , Hepatite B/genética , Hepatite B/virologia , Plasmídeos/genética , Transcrição Gênica , Sequência de Bases , Células Cultivadas , DNA Viral/genética , Genoma Viral , Células Hep G2 , Hepatócitos/patologia , Hepatócitos/virologia , Heterocromatina/metabolismo , Humanos
14.
Methods Mol Biol ; 1540: 1-14, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-27975303

RESUMO

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.


Assuntos
Vírus da Hepatite B/fisiologia , Hepatite B/metabolismo , Hepatite B/virologia , Transportadores de Ânions Orgânicos Dependentes de Sódio/metabolismo , Receptores Virais/metabolismo , Simportadores/metabolismo , Linhagem Celular , Células Cultivadas , DNA Circular , DNA Viral , Ensaio de Imunoadsorção Enzimática , Imunofluorescência , Expressão Gênica , Células Hep G2 , Hepatite B/genética , Antígenos de Superfície da Hepatite B/imunologia , Antígenos E da Hepatite B/imunologia , Humanos , Transportadores de Ânions Orgânicos Dependentes de Sódio/genética , RNA Viral , Receptores Virais/genética , Simportadores/genética , Replicação Viral
15.
Elife ; 62017 09 26.
Artigo em Inglês | MEDLINE | ID: mdl-28949917

RESUMO

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.


Assuntos
Anticorpos Neutralizantes/administração & dosagem , Anticorpos Antivirais/administração & dosagem , Antivirais/administração & dosagem , Hepatite B/terapia , Imunoterapia/métodos , Animais , Anticorpos Monoclonais/administração & dosagem , Quimioprevenção/métodos , Modelos Animais de Doenças , Hepatite B/prevenção & controle , Vírus da Hepatite B/efeitos dos fármacos , Vírus Delta da Hepatite/efeitos dos fármacos , Humanos , Camundongos , Resultado do Tratamento
16.
Elife ; 1: e00049, 2012 Nov 13.
Artigo em Inglês | MEDLINE | ID: mdl-23150796

RESUMO

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.


Assuntos
Vírus da Hepatite B/metabolismo , Vírus Delta da Hepatite/metabolismo , Hepatócitos/metabolismo , Transportadores de Ânions Orgânicos Dependentes de Sódio/metabolismo , Receptores Virais/metabolismo , Simportadores/metabolismo , Proteínas do Envelope Viral/metabolismo , Sequência de Aminoácidos , Animais , Transporte Biológico , Linhagem Celular , Expressão Gênica , Vírus da Hepatite B/química , Vírus da Hepatite B/genética , Vírus Delta da Hepatite/química , Vírus Delta da Hepatite/genética , Hepatócitos/patologia , Hepatócitos/virologia , Humanos , Fígado/metabolismo , Fígado/patologia , Fígado/virologia , Dados de Sequência Molecular , Transportadores de Ânions Orgânicos Dependentes de Sódio/química , Transportadores de Ânions Orgânicos Dependentes de Sódio/genética , Peptídeos/química , Processos Fotoquímicos , Cultura Primária de Células , Ligação Proteica , Estrutura Terciária de Proteína , Receptores Virais/química , Receptores Virais/genética , Simportadores/química , Simportadores/genética , Ácido Taurocólico/metabolismo , Tupaia , Proteínas do Envelope Viral/química , Proteínas do Envelope Viral/genética , Internalização do Vírus
17.
Elife ; 32012 Nov 13.
Artigo em Inglês | MEDLINE | ID: mdl-25409679

RESUMO

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.


Assuntos
Vírus da Hepatite B/metabolismo , Vírus Delta da Hepatite/metabolismo , Transportadores de Ânions Orgânicos Dependentes de Sódio/metabolismo , Receptores Virais/metabolismo , Simportadores/metabolismo , Sequência de Aminoácidos , Animais , Linhagem Celular Tumoral , Células Cultivadas , Suscetibilidade a Doenças , Hepatite B/patologia , Hepatite B/virologia , Hepatite D/patologia , Hepatite D/virologia , Hepatócitos/metabolismo , Hepatócitos/patologia , Hepatócitos/virologia , Humanos , Ligantes , Dados de Sequência Molecular , Transportadores de Ânions Orgânicos Dependentes de Sódio/química , Ligação Proteica , Estrutura Terciária de Proteína , Receptores Virais/química , Reprodutibilidade dos Testes , Simportadores/química , Tupaiidae , Vírion/metabolismo
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa