Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
Mais filtros

Base de dados
Tipo de documento
País de afiliação
Intervalo de ano de publicação
1.
PLoS Pathog ; 10(5): e1004166, 2014 May.
Artigo em Inglês | MEDLINE | ID: mdl-24874215

RESUMO

Coronaviruses raise serious concerns as emerging zoonotic viruses without specific antiviral drugs available. Here we screened a collection of 16671 diverse compounds for anti-human coronavirus 229E activity and identified an inhibitor, designated K22, that specifically targets membrane-bound coronaviral RNA synthesis. K22 exerts most potent antiviral activity after virus entry during an early step of the viral life cycle. Specifically, the formation of double membrane vesicles (DMVs), a hallmark of coronavirus replication, was greatly impaired upon K22 treatment accompanied by near-complete inhibition of viral RNA synthesis. K22-resistant viruses contained substitutions in non-structural protein 6 (nsp6), a membrane-spanning integral component of the viral replication complex implicated in DMV formation, corroborating that K22 targets membrane bound viral RNA synthesis. Besides K22 resistance, the nsp6 mutants induced a reduced number of DMVs, displayed decreased specific infectivity, while RNA synthesis was not affected. Importantly, K22 inhibits a broad range of coronaviruses, including Middle East respiratory syndrome coronavirus (MERS-CoV), and efficient inhibition was achieved in primary human epithelia cultures representing the entry port of human coronavirus infection. Collectively, this study proposes an evolutionary conserved step in the life cycle of positive-stranded RNA viruses, the recruitment of cellular membranes for viral replication, as vulnerable and, most importantly, druggable target for antiviral intervention. We expect this mode of action to serve as a paradigm for the development of potent antiviral drugs to combat many animal and human virus infections.


Assuntos
Antivirais/farmacologia , Infecções por Coronavirus/virologia , Coronavirus , RNA Viral/genética , Vírus Sinciciais Respiratórios , Replicação Viral/efeitos dos fármacos , Animais , Linhagem Celular , Membrana Celular/metabolismo , Infecções por Coronavirus/prevenção & controle , Humanos , Internalização do Vírus/efeitos dos fármacos
2.
Viruses ; 13(5)2021 05 12.
Artigo em Inglês | MEDLINE | ID: mdl-34065826

RESUMO

The contribution of virus components to liberation of herpes simplex virus type 2 (HSV-2) progeny virions from the surface of infected cells is poorly understood. We report that the HSV-2 mutant deficient in the expression of a mucin-like membrane-associated glycoprotein G (mgG) exhibited defect in the release of progeny virions from infected cells manifested by ~2 orders of magnitude decreased amount of infectious virus in a culture medium as compared to native HSV-2. Electron microscopy revealed that the mgG deficient virions were produced in infected cells and present at the cell surface. These virions could be forcibly liberated to a nearly native HSV-2 level by the treatment of cells with glycosaminoglycan (GAG)-mimicking oligosaccharides. Comparative assessment of the interaction of mutant and native virions with surface-immobilized chondroitin sulfate GAG chains revealed that while the mutant virions associated with GAGs ~fourfold more extensively, the lateral mobility of bound virions was much poorer than that of native virions. These data indicate that the mgG of HSV-2 balances the virus interaction with GAG chains, a feature critical to prevent trapping of the progeny virions at the surface of infected cells.


Assuntos
Glicoproteínas/metabolismo , Herpes Simples/virologia , Herpesvirus Humano 2/fisiologia , Proteínas do Envelope Viral/metabolismo , Liberação de Vírus , Membrana Celular/metabolismo , Células Cultivadas , Glicoproteínas/genética , Herpesvirus Humano 2/ultraestrutura , Interações Hospedeiro-Patógeno , Humanos , Mutação , Proteínas do Envelope Viral/genética , Vírion/ultraestrutura
3.
J Virol ; 81(24): 13424-34, 2007 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-17928351

RESUMO

Variants of herpes simplex virus type 2 (HSV-2) generated by virus passage in GMK-AH1 cells in the presence of the sulfated oligosaccharide PI-88 were analyzed. Many of these variants were substantially resistant to PI-88 in their initial infection of cells and/or their cell-to-cell spread. The major alteration detected in all variants resistant to PI-88 in the initial infection of cells was a frameshift mutation(s) in the glycoprotein G (gG) gene that resulted in the lack of protein expression. Molecular transfer of the altered gG gene into the wild-type background confirmed that the gG-deficient recombinants were resistant to PI-88. In addition to PI-88, all gG-deficient variants of HSV-2 were resistant to the sulfated polysaccharide heparin. The gG-deficient virions were capable of attaching to cells, and this activity was relatively resistant to PI-88. In addition to having a drug-resistant phenotype, the gG-deficient variants were inefficiently released from infected cells. Purified gG bound to heparin and showed the cell-binding activity which was inhibited by PI-88. Many PI-88 variants produced syncytia in cultured cells and contained alterations in gB, including the syncytium-inducing L792P amino acid substitution. Although this phenotype can enhance the lateral spread of HSV in cells, it conferred no virus resistance to PI-88. Some PI-88 variants also contained occasional alterations in gC, gD, gE, gK, and UL24. In conclusion, we found that glycoprotein gG, a mucin-like component of the HSV-2 envelope, was targeted by sulfated oligo- and polysaccharides. This is a novel finding that suggests the involvement of HSV-2 gG in interactions with sulfated polysaccharides, including cell surface glycosaminoglycans.


Assuntos
Farmacorresistência Viral , Herpesvirus Humano 2/efeitos dos fármacos , Oligossacarídeos/farmacologia , Polissacarídeos/farmacologia , Proteínas do Envelope Viral/efeitos dos fármacos , Animais , Linhagem Celular , Mutação da Fase de Leitura , Herpesvirus Humano 2/genética , Herpesvirus Humano 2/fisiologia , Humanos , Rim/citologia , Rim/virologia , Dados de Sequência Molecular , Análise de Sequência de DNA , Proteínas do Envelope Viral/deficiência , Proteínas do Envelope Viral/genética , Proteínas do Envelope Viral/metabolismo , Ensaio de Placa Viral
4.
Sci Rep ; 6: 18745, 2016 Jan 07.
Artigo em Inglês | MEDLINE | ID: mdl-26738561

RESUMO

H9N2 avian influenza virus is a major cause of poultry production loss across Asia leading to the wide use of vaccines. Efficacy of vaccines is often compromised due to the rapid emergence of antigenic variants. To improve the effectiveness of vaccines in the field, a better understanding of the antigenic epitopes of the major antigen, hemagglutinin, is required. To address this, a panel of nine monoclonal antibodies were generated against a contemporary Pakistani H9N2 isolate, which represents a major Asian H9N2 viral lineage. Antibodies were characterized in detail and used to select a total of 26 unique 'escape' mutants with substitutions across nine different amino acid residues in hemagglutinin including seven that have not been described as antigenic determinants for H9N2 viruses before. Competition assays and structural mapping revealed two novel, discrete antigenic sites "H9-A" and "H9-B". Additionally, a second subset of escape mutants contained amino acid deletions within the hemagglutinin receptor binding site. This constitutes a novel method of escape for group 1 hemagglutinins and could represent an alternative means for H9N2 viruses to overcome vaccine induced immunity. These results will guide surveillance efforts for arising antigenic variants as well as evidence based vaccine seed selection and vaccine design.


Assuntos
Anticorpos Antivirais/química , Antígenos Virais/imunologia , Vírus da Influenza A Subtipo H9N2/imunologia , Sequência de Aminoácidos , Substituição de Aminoácidos , Animais , Anticorpos Monoclonais Murinos/química , Anticorpos Neutralizantes/química , Anticorpos Antivirais/sangue , Antígenos Virais/química , Galinhas/imunologia , Galinhas/virologia , Reações Cruzadas , Mapeamento de Epitopos , Humanos , Hibridomas , Evasão da Resposta Imune , Soros Imunes/química , Vírus da Influenza A Subtipo H9N2/química , Influenza Aviária/imunologia , Influenza Aviária/virologia , Influenza Humana/imunologia , Influenza Humana/virologia , Peptídeos e Proteínas de Sinalização Intracelular , Camundongos , Modelos Moleculares , Ligação Proteica , Conformação Proteica em alfa-Hélice
5.
APMIS ; 121(4): 280-9, 2013 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-23030500

RESUMO

Several herpesviruses induce expression of the selectin receptor sialyl-Lewis X (sLe(x) ) by activating transcription of one or more of silent host FUT genes, each one encoding a fucosyltransferase that catalyses the rate-limiting step of sLe(x) synthesis. The aim here was to identify the identity of the glycoconjugate associated with sLe(x) glycoepitope in herpes simplex virus type 1 (HSV-1) infected human diploid fibroblasts, using immunofluorescence confocal microscopy. Cells infected with all tested HSV-1 strains analysed demonstrated bright sLe(x) fluorescence, except for two mutant viruses that were unable to induce proper expression of viral glycoprotein gC-1: One gC-1 null mutant and another mutant expressing gC-1 devoid of its major O-glycan-containing region (aa 33-116). The sLe(x) reactivity of HSV-1 infected cells was abolished by mild alkali treatment. Altogether the results indicated that the detectable sLe(x) was associated with O-linked glycans, situated in the mucin region of gC-1. No evidence for sLe(x) (i) in other HSV-1 glycoproteins with mucin domains such as gI-1 or (ii) in host cell glycoproteins/glycolipids was found. Thus, the mucin domain of HSV-1 gC-1 may support expression of selectin ligands such as sLe(x) and other larger O-linked glycans in cell types lacking endogenous mucin domain-containing glycoproteins, optimized for O-glycan expression, provided that the adequate host glycosyltransferase genes are activated.


Assuntos
Herpesvirus Humano 1/fisiologia , Antígenos CD15/biossíntese , Proteínas do Envelope Viral/fisiologia , Fibroblastos/virologia , Fucosiltransferases/fisiologia , Glicosilação , Humanos , Ligantes , Mucinas/fisiologia , Antígeno Sialil Lewis X , Proteínas do Envelope Viral/química
6.
Antiviral Res ; 88(3): 317-24, 2010 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-20965215

RESUMO

To search for novel drugs against human respiratory syncytial virus (RSV), we have screened a diversity collection of 16,671 compounds for anti-RSV activity in cultures of HEp-2 cells. Two of the hit compounds, i.e., the N-(2-hydroxyethyl)-4-methoxy-N-methyl-3-(6-methyl[1,2,4]triazolo[3,4-a]phthalazin-3-yl)benzenesulfonamide (designated as P13) and the 1,4-bis(3-methyl-4-pyridinyl)-1,4-diazepane (designated as C15), reduced the virus infectivity with IC50 values of 0.11 and 0.13µM respectively. The concentration of P13 and C15 that reduced the viability of HEp-2 cells by 50% was 310 and 75µM respectively. Both P13 and C15 exhibited no direct virucidal activity or inhibitory effects on the virus attachment to cells. However, to inhibit formation of RSV-induced syncytial plaques P13 and C15 had to be present during the virus entry into the cells and the cell-to-cell transmission of the virus. The RSV multiplication in HEp-2 cells in the presence of P13 or C15 resulted in rapid selection of viral variants that were ∼1000 times less sensitive to these drugs than original virus. Sequencing of resistant viruses revealed presence of amino acid substitutions in the F protein of RSV, i.e., the D489G for C15-selected, and the T400I and N197T (some clones) for the P13-selected virus variants. In conclusion, we have identified two novel fusion inhibitors of RSV, and the detailed understanding of their mode of antiviral activity including selection for the drug resistant viral variants may help to develop selective and efficient anti-RSV drugs.


Assuntos
Substituição de Aminoácidos/efeitos dos fármacos , Antivirais/química , Antivirais/farmacologia , Azepinas/química , Azepinas/farmacologia , Farmacorresistência Viral , Fusão de Membrana/efeitos dos fármacos , Ftalazinas/química , Ftalazinas/farmacologia , Piridinas/química , Piridinas/farmacologia , Infecções por Vírus Respiratório Sincicial/genética , Vírus Sincicial Respiratório Humano/efeitos dos fármacos , Vírus Sincicial Respiratório Humano/genética , Seleção Genética/efeitos dos fármacos , Sulfonamidas/química , Sulfonamidas/farmacologia , Proteínas Virais de Fusão/genética , Ligação Viral/efeitos dos fármacos , Substituição de Aminoácidos/genética , Animais , Antivirais/uso terapêutico , Azepinas/uso terapêutico , Linhagem Celular , Cricetinae , Avaliação Pré-Clínica de Medicamentos , Humanos , Concentração Inibidora 50 , Ftalazinas/metabolismo , Ftalazinas/uso terapêutico , Piridinas/uso terapêutico , Infecções por Vírus Respiratório Sincicial/tratamento farmacológico , Vírus Sincicial Respiratório Humano/crescimento & desenvolvimento , Análise de Sequência , Sulfonamidas/metabolismo , Sulfonamidas/uso terapêutico , Proteínas Virais de Fusão/metabolismo
7.
Antiviral Res ; 86(2): 196-203, 2010 May.
Artigo em Inglês | MEDLINE | ID: mdl-20176055

RESUMO

Although sulfated polysaccharides potently inhibit the infectivity of herpes simplex virus (HSV) and human immunodeficiency virus in cultured cells, these compounds fail to show protective effects in humans, most likely due to their poor virucidal activity. Herein we report on sulfated oligosaccharide glycosides related to muparfostat (formerly known as PI-88) and their assessment for anti-HSV activity. Chemical modifications based on the introduction of specific hydrophobic groups at the reducing end of a sulfated oligosaccharide chain enhanced the compound's capability to inhibit the infection of cells by HSV-1 and HSV-2 and abrogated the cell-to-cell transmission of HSV-2. Furthermore, modification with a highly lipophilic cholestanyl group provided a compound with virucidal activity against HSV. This glycoside targeted the viral particle and, to a lesser degree, the cell, and exhibited an antiviral mode of action typical for sulfated polysaccharides and virucides, i.e., interference with the virus attachment to cells and irreversible inactivation of virus infectivity, respectively. The virucidal activity was decreased in the presence of human cervical secretions suggesting that higher doses of this glycoside might be needed for in vivo application. Altogether, the sulfated oligosaccharide-cholestanyl glycoside exhibits potent anti-HSV activity and is, therefore, a good candidate for development as a virucide.


Assuntos
Antivirais/farmacologia , Herpesvirus Humano 1/efeitos dos fármacos , Herpesvirus Humano 2/efeitos dos fármacos , Viabilidade Microbiana/efeitos dos fármacos , Oligossacarídeos/farmacologia , Animais , Antivirais/química , Secreções Corporais , Linhagem Celular , Chlorocebus aethiops , Herpesvirus Humano 1/fisiologia , Herpesvirus Humano 2/fisiologia , Humanos , Microscopia Eletrônica de Transmissão , Estrutura Molecular , Oligossacarídeos/química , Ensaio de Placa Viral
8.
Virology ; 400(2): 197-206, 2010 May 10.
Artigo em Inglês | MEDLINE | ID: mdl-20176392

RESUMO

Human antibodies specific for glycoprotein C (gC1) of herpes simplex virus type 1 (HSV-1) neutralized the virus infectivity and efficiently inhibited attachment of HSV-1 to human HaCaT keratinocytes and to murine mutant L cells expressing either heparan sulfate or chondroitin sulfate at the cell surface. Similar activities were observed with anti-gC1 monoclonal antibody B1C1. In addition to HaCaT and L cells, B1C1 antibody neutralized HSV-1 infectivity in simian GMK AH1 cells mildly pre-treated with heparinase III. Human anti-gC1 antibodies efficiently competed with the binding of gC1 to B1C1 antibody whose epitope overlaps a part of the attachment domain of gC1. Human anti-gC1 and B1C1 antibodies extended survival time of mice experimentally infected with HSV-1. We conclude that in HaCaT cells and in cell systems showing restricted expression of glycosaminoglycans, human and some monoclonal anti-gC1 antibodies can target the cell-binding domain of this protein and neutralize viral infectivity.


Assuntos
Anticorpos Neutralizantes/imunologia , Anticorpos Antivirais/imunologia , Herpesvirus Humano 1/imunologia , Proteínas do Envelope Viral/imunologia , Animais , Sítios de Ligação , Linhagem Celular , Chlorocebus aethiops , Modelos Animais de Doenças , Epitopos/imunologia , Heparitina Sulfato/metabolismo , Herpes Simples/imunologia , Humanos , Queratinócitos/virologia , Camundongos , Testes de Neutralização , Análise de Sobrevida , Ligação Viral
9.
Virology ; 367(2): 244-52, 2007 Oct 25.
Artigo em Inglês | MEDLINE | ID: mdl-17604805

RESUMO

Herpes simplex virus type 1 variants selected by virus propagation in cultured cells in the presence of the sulfated oligosaccharide PI-88 were analyzed. Many of these variants were substantially resistant to the presence of PI-88 during their initial infection of cells and/or their cell-to-cell spread. Nucleotide sequence analysis revealed that the deletion of amino acids 33-116 of gC but not lack of gC expression provided the virus with selective advantage to infect cells in the presence of PI-88. Purified gC (Delta33-116) was more resistant to PI-88 than unaltered protein in its binding to cells. Alterations that partly contributed to the virus resistance to PI-88 in its cell-to-cell spread activity were amino acid substitutions Q27R in gD and R770W in gB. These results suggest that PI-88 targets several distinct viral glycoproteins during the course of initial virus infection and cell-to-cell spread.


Assuntos
Resistência Microbiana a Medicamentos/genética , Herpesvirus Humano 1/efeitos dos fármacos , Herpesvirus Humano 1/genética , Oligossacarídeos/farmacologia , Proteínas do Envelope Viral/efeitos dos fármacos , Sequência de Aminoácidos , Aminoácidos/metabolismo , Animais , DNA Viral , Herpesvirus Humano 1/metabolismo , Humanos , Mutação , Células Tumorais Cultivadas , Proteínas do Envelope Viral/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA