Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Proc Natl Acad Sci U S A ; 119(4)2022 01 25.
Artigo em Inglês | MEDLINE | ID: mdl-35046043

RESUMO

Receptor usage defines cell tropism and contributes to cell entry and infection. Coxsackievirus B (CVB) engages coxsackievirus and adenovirus receptor (CAR), and selectively utilizes the decay-accelerating factor (DAF; CD55) to infect cells. However, the differential receptor usage mechanism for CVB remains elusive. This study identified VP3-234 residues (234Q/N/V/D/E) as critical population selection determinants during CVB3 virus evolution, contributing to diverse binding affinities to CD55. Cryoelectron microscopy (cryo-EM) structures of CD55-binding/nonbinding isolates and their complexes with CD55 or CAR were obtained under both neutral and acidic conditions, and the molecular mechanism of VP3-234 residues determining CD55 affinity/specificity for naturally occurring CVB3 strains was elucidated. Structural and biochemical studies in vitro revealed the dynamic entry process of CVB3 and the function of the uncoating receptor CAR with different pH preferences. This work provides detailed insight into the molecular mechanism of CVB infection and contributes to an in-depth understanding of enterovirus attachment receptor usage.


Assuntos
Antígenos CD55/metabolismo , Infecções por Coxsackievirus/metabolismo , Infecções por Coxsackievirus/virologia , Enterovirus Humano B/fisiologia , Interações Hospedeiro-Patógeno , Receptores Virais/metabolismo , Sequência de Aminoácidos , Substituição de Aminoácidos , Sítios de Ligação , Enterovirus Humano B/ultraestrutura , Humanos , Modelos Moleculares , Ligação Proteica , Conformação Proteica , Domínios e Motivos de Interação entre Proteínas , Receptores Virais/química , Relação Estrutura-Atividade , Ligação Viral
2.
Nat Commun ; 10(1): 1138, 2019 03 08.
Artigo em Inglês | MEDLINE | ID: mdl-30850609

RESUMO

Viruses from the genus Enterovirus are important human pathogens. Receptor binding or exposure to acidic pH in endosomes converts enterovirus particles to an activated state that is required for genome release. However, the mechanism of enterovirus uncoating is not well understood. Here, we use cryo-electron microscopy to visualize virions of human echovirus 18 in the process of genome release. We discover that the exit of the RNA from the particle of echovirus 18 results in a loss of one, two, or three adjacent capsid-protein pentamers. The opening in the capsid, which is more than 120 Å in diameter, enables the release of the genome without the need to unwind its putative double-stranded RNA segments. We also detect capsids lacking pentamers during genome release from echovirus 30. Thus, our findings uncover a mechanism of enterovirus genome release that could become target for antiviral drugs.


Assuntos
Capsídeo/ultraestrutura , Enterovirus Humano B/ultraestrutura , Genoma Viral , RNA Viral/genética , Vírion/ultraestrutura , Desenvelopamento do Vírus/genética , Animais , Capsídeo/química , Chlorocebus aethiops , Microscopia Crioeletrônica , Enterovirus Humano B/genética , Células Epiteliais/ultraestrutura , Células Epiteliais/virologia , Humanos , Concentração de Íons de Hidrogênio , Simulação de Dinâmica Molecular , RNA de Cadeia Dupla/química , RNA de Cadeia Dupla/genética , RNA Viral/química , Vírion/genética
3.
Nanoscale ; 7(41): 17457-67, 2015 Nov 07.
Artigo em Inglês | MEDLINE | ID: mdl-26440968

RESUMO

Visualization and tracking of viruses without compromising their functionality is crucial in order to understand virus targeting to cells and tissues, and to understand the subsequent subcellular steps leading to virus uncoating and replication. Enteroviruses are important human pathogens causing a vast number of acute infections, and are also suggested to contribute to the development of chronic diseases like type I diabetes. Here, we demonstrate a novel method to target site-specifically the hydrophobic pocket of enteroviruses. A probe, a derivative of Pleconaril, was developed and conjugated to various labels that enabled the visualization of enteroviruses under light and electron microscopes. The probe mildly stabilized the virus particle by increasing the melting temperature by 1-3 degrees, and caused a delay in the uncoating of the virus in the cellular endosomes, but could not however inhibit the receptor binding, cellular entry or infectivity of the virus. The hydrophobic pocket binding moiety of the probe was shown to bind to echovirus 1 particle by STD and tr-NOESY NMR methods. Furthermore, binding to echovirus 1 and Coxsackievirus A9, and to a lesser extent to Coxsackie virus B3 was verified by using a gold nanocluster labeled probe by TEM analysis. Molecular modelling suggested that the probe fits the hydrophobic pockets of EV1 and CVA9, but not of CVB3 as expected, correlating well with the variations in the infectivity and stability of the virus particles. EV1 conjugated to the fluorescent dye labeled probe was efficiently internalized into the cells. The virus-fluorescent probe conjugate accumulated in the cytoplasmic endosomes and caused infection starting from 6 hours onwards. Remarkably, before and during the time of replication, the fluorescent probe was seen to leak from the virus-positive endosomes and thus separate from the capsid proteins that were left in the endosomes. These results suggest that, like the physiological hydrophobic content, the probe may be released upon virus uncoating. Our results collectively thus show that the gold and fluorescently labeled probes may be used to track and visualize the studied enteroviruses during the early phases of infection opening new avenues to follow virus uncoating in cells.


Assuntos
Enterovirus Humano B/química , Corantes Fluorescentes/química , Oxidiazóis/química , Linhagem Celular Tumoral , Infecções por Coxsackievirus/metabolismo , Infecções por Coxsackievirus/patologia , Enterovirus Humano B/metabolismo , Enterovirus Humano B/ultraestrutura , Ouro/química , Humanos , Interações Hidrofóbicas e Hidrofílicas , Oxazóis
4.
J Virol ; 88(10): 5755-65, 2014 May.
Artigo em Inglês | MEDLINE | ID: mdl-24623425

RESUMO

UNLABELLED: The coxsackievirus and adenovirus receptor (CAR) has been identified as the cellular receptor for group B coxsackieviruses, including serotype 3 (CVB3). CAR mediates infection by binding to CVB3 and catalyzing conformational changes in the virus that result in formation of the altered, noninfectious A-particle. Kinetic analyses show that the apparent first-order rate constant for the inactivation of CVB3 by soluble CAR (sCAR) at physiological temperatures varies nonlinearly with sCAR concentration. Cryo-electron microscopy (cryo-EM) reconstruction of the CVB3-CAR complex resulted in a 9.0-Šresolution map that was interpreted with the four available crystal structures of CAR, providing a consensus footprint for the receptor binding site. The analysis of the cryo-EM structure identifies important virus-receptor interactions that are conserved across picornavirus species. These conserved interactions map to variable antigenic sites or structurally conserved regions, suggesting a combination of evolutionary mechanisms for receptor site preservation. The CAR-catalyzed A-particle structure was solved to a 6.6-Šresolution and shows significant rearrangement of internal features and symmetric interactions with the RNA genome. IMPORTANCE: This report presents new information about receptor use by picornaviruses and highlights the importance of attaining at least an ∼9-Šresolution for the interpretation of cryo-EM complex maps. The analysis of receptor binding elucidates two complementary mechanisms for preservation of the low-affinity (initial) interaction of the receptor and defines the kinetics of receptor-catalyzed conformational change to the A-particle.


Assuntos
Proteína de Membrana Semelhante a Receptor de Coxsackie e Adenovirus/metabolismo , Enterovirus Humano B/fisiologia , Enterovirus Humano B/ultraestrutura , Ligação Viral , Sequência de Aminoácidos , Sítios de Ligação , Microscopia Crioeletrônica , Cinética , Modelos Moleculares , Dados de Sequência Molecular , Ligação Proteica , Conformação Proteica , Vírion/metabolismo , Vírion/ultraestrutura , Inativação de Vírus
5.
J Virol ; 86(23): 12571-81, 2012 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-22973031

RESUMO

The coxsackievirus-adenovirus receptor (CAR) and decay-accelerating factor (DAF) have been identified as cellular receptors for coxsackievirus B3 (CVB3). The first described DAF-binding isolate was obtained during passage of the prototype strain, Nancy, on rhabdomyosarcoma (RD) cells, which express DAF but very little CAR. Here, the structure of the resulting variant, CVB3-RD, has been solved by X-ray crystallography to 2.74 Å, and a cryo-electron microscopy reconstruction of CVB3-RD complexed with DAF has been refined to 9.0 Å. This new high-resolution structure permits us to correct an error in our previous view of DAF-virus interactions, providing a new footprint of DAF that bridges two adjacent protomers. The contact sites between the virus and DAF clearly encompass CVB3-RD residues recently shown to be required for binding to DAF; these residues interact with DAF short consensus repeat 2 (SCR2), which is known to be essential for virus binding. Based on the new structure, the mode of the DAF interaction with CVB3 differs significantly from the mode reported previously for DAF binding to echoviruses.


Assuntos
Antígenos CD55/química , Enterovirus Humano B/ultraestrutura , Modelos Moleculares , Conformação Proteica , Receptores Virais/química , Antígenos CD55/metabolismo , Linhagem Celular Tumoral , Microscopia Crioeletrônica , Cristalização , Cristalografia por Raios X , Humanos , Receptores Virais/metabolismo
6.
Curr Top Microbiol Immunol ; 323: 67-87, 2008.
Artigo em Inglês | MEDLINE | ID: mdl-18357766

RESUMO

The coxsackievirus and adenovirus receptor (CAR) has been studied extensively since its identification and isolation in 1997. The CAR is an immunoglobulin superfamily protein with two extracellular Ig-like domains, a single membrane-spanning sequence, and a significant cytoplasmic domain. It is structurally and functionally similar to the junctional adhesion molecules. The amino terminal domain, distal from the membrane, has been structurally characterized alone, bound to the adenovirus fiber knob, and, in full-length CAR, docked in the canyon structure of the coxsackievirus capsid. Although the past decade has produced a burst of new knowledge about CAR, significant questions concerning its function in normal physiology and coxsackievirus-related pathology remain unanswered.


Assuntos
Infecções por Coxsackievirus/virologia , Enterovirus Humano B/metabolismo , Proteínas de Membrana/metabolismo , Receptores Virais/química , Receptores Virais/metabolismo , Animais , Proteína de Membrana Semelhante a Receptor de Coxsackie e Adenovirus , Enterovirus Humano B/ultraestrutura , Humanos , Conformação Proteica , Estrutura Terciária de Proteína , Receptores Virais/genética
7.
J Virol ; 81(23): 12927-35, 2007 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-17804498

RESUMO

Many entero-, parecho-, and rhinoviruses use immunoglobulin (Ig)-like receptors that bind into the viral canyon and are required to initiate viral uncoating during infection. However, some of these viruses use an alternative or additional receptor that binds outside the canyon. Both the coxsackievirus-adenovirus receptor (CAR), an Ig-like molecule that binds into the viral canyon, and decay-accelerating factor (DAF) have been identified as cellular receptors for coxsackievirus B3 (CVB3). A cryoelectron microscopy reconstruction of a variant of CVB3 complexed with DAF shows full occupancy of the DAF receptor in each of 60 binding sites. The DAF molecule bridges the canyon, blocking the CAR binding site and causing the two receptors to compete with one another. The binding site of DAF on CVB3 differs from the binding site of DAF on the surface of echoviruses, suggesting independent evolutionary processes.


Assuntos
Antígenos CD55/metabolismo , Enterovirus Humano B/metabolismo , Receptores Virais/metabolismo , Ligação Viral , Antígenos CD55/química , Antígenos CD55/ultraestrutura , Microscopia Crioeletrônica , Enterovirus Humano B/química , Enterovirus Humano B/ultraestrutura , Modelos Moleculares , Ligação Proteica , Receptores Virais/química , Receptores Virais/ultraestrutura
8.
J Virol ; 77(9): 5475-86, 2003 May.
Artigo em Inglês | MEDLINE | ID: mdl-12692248

RESUMO

Swine vesicular disease virus (SVDV) is an Enterovirus of the family Picornaviridae that causes symptoms indistinguishable from those of foot-and-mouth disease virus. Phylogenetic studies suggest that it is a recently evolved genetic sublineage of the important human pathogen coxsackievirus B5 (CBV5), and in agreement with this, it has been shown to utilize the coxsackie and adenovirus receptor (CAR) for cell entry. The 3.0-A crystal structure of strain UK/27/72 SVDV (highly virulent) reveals the expected similarity in core structure to those of other picornaviruses, showing most similarity to the closest available structure to CBV5, that of coxsackievirus B3 (CBV3). Features that help to cement together and rigidify the protein subunits are extended in this virus, perhaps explaining its extreme tolerance of environmental factors. Using the large number of capsid sequences available for both SVDV and CBV5, we have mapped the amino acid substitutions that may have occurred during the supposed adaptation of SVDV to a new host onto the structure of SVDV and a model of the SVDV/CAR complex generated by reference to the cryo-electron microscopy-visualized complex of CBV3 and CAR. The changes fall into three clusters as follows: one lines the fivefold pore, a second maps to the CAR-binding site and partially overlaps the site for decay accelerating factor (DAF) to bind to echovirus 7 (ECHO7), and the third lies close to the fivefold axis, where the low-density lipoprotein receptor binds to the minor group of rhinoviruses. Later changes in SVDV (post-1971) map to the first two clusters and may, by optimizing recognition of a pig CAR and/or DAF homologue, have improved the adaptation of the virus to pigs.


Assuntos
Adaptação Fisiológica , Sequência de Aminoácidos , Substituição de Aminoácidos , Animais , Sítios de Ligação , Proteínas do Capsídeo/química , Proteínas do Capsídeo/genética , Proteína de Membrana Semelhante a Receptor de Coxsackie e Adenovirus , Microscopia Crioeletrônica , Cristalografia por Raios X , Enterovirus Humano B/química , Enterovirus Humano B/patogenicidade , Enterovirus Humano B/ultraestrutura , Humanos , Modelos Moleculares , Dados de Sequência Molecular , Receptores Virais/metabolismo , Suínos , Doença Vesicular Suína/virologia
9.
J Virol ; 76(4): 1856-65, 2002 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-11799180

RESUMO

Echovirus 1 (EV1) is a human pathogen which belongs to the Picornaviridae family of RNA viruses. We have analyzed the early events of infection after EV1 binding to its receptor alpha 2 beta 1 integrin and elucidated the route by which EV1 gains access to the host cell. EV1 binding onto the cell surface and subsequent entry resulted in conformational changes of the viral capsid as demonstrated by sucrose gradient sedimentation analysis. After 15 min to 2 h postinfection (p.i.) EV1 capsid proteins were seen in vesicular structures that were negative for markers of the clathrin-dependent endocytic pathway. In contrast, immunofluorescence confocal microscopy showed that EV1, alpha 2 beta 1 integrin, and caveolin-1 were internalized together in vesicular structures to the perinuclear area. Electron microscopy showed the presence of EV1 particles inside caveolae. Furthermore, infective EV1 could be isolated with anti-caveolin-1 beads 15 min p.i., confirming a close association with caveolin-1. Finally, the expression of dominant negative caveolin in cells markedly inhibited EV1 infection, indicating the importance of caveolae for the viral replication cycle of EV1.


Assuntos
Cavéolas/virologia , Enterovirus Humano B/patogenicidade , Infecções por Enterovirus/virologia , Animais , Capsídeo/metabolismo , Cavéolas/ultraestrutura , Caveolina 1 , Caveolinas/metabolismo , Clatrina/metabolismo , Enterovirus Humano B/ultraestrutura , Humanos , Integrinas/metabolismo , Microscopia Confocal , Microscopia Eletrônica , Coelhos , Receptores de Colágeno , Células Tumorais Cultivadas/ultraestrutura , Células Tumorais Cultivadas/virologia , Microglobulina beta-2/metabolismo
10.
Diabetologia ; 40(1): 53-61, 1997 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-9028718

RESUMO

Techniques were developed to look for evidence of viral infection in formalin-fixed paraffin-embedded autopsy pancreatic tissues from patients who had died of recent-onset insulin-dependent diabetes mellitus. DNA extracted from 47 pancreases in which good DNA preservation was confirmed was analysed by a polymerase chain reaction for Epstein-Barr virus and by a nested polymerase chain reaction for cytomegalovirus. Histological sections from 29 pancreases in which there was good RNA preservation were tested for the presence of enterovirus and Epstein-Barr virus using in situ hybridization techniques. Seventy-five pancreases were analysed immunohistochemically for the presence of mumps virus. None of these viruses could be detected in any of the diabetic pancreases studied. Control studies suggested that the techniques employed were as sensitive as culture done at the time of autopsy. Pancreas was available for study in 9 infants who had died of myocarditis; enterovirus was demonstrable in islets in 5 of these cases. An acute or persisting infection in the pancreas at the time of clinical onset of insulin-dependent diabetes by any of the 4 virus included in this study seems unlikely.


Assuntos
Citomegalovirus/isolamento & purificação , Diabetes Mellitus Tipo 1/virologia , Enterovirus Humano B/isolamento & purificação , Herpesvirus Humano 4/isolamento & purificação , Vírus da Caxumba/isolamento & purificação , Pâncreas/virologia , Adolescente , Adulto , Autopsia , Sequência de Bases , Criança , Pré-Escolar , Citomegalovirus/genética , Citomegalovirus/ultraestrutura , Primers do DNA/química , DNA Viral/análise , DNA Viral/genética , Diabetes Mellitus Tipo 1/patologia , Eletroforese em Gel de Ágar , Enterovirus Humano B/genética , Enterovirus Humano B/ultraestrutura , Feminino , Coração/virologia , Herpesvirus Humano 4/genética , Herpesvirus Humano 4/ultraestrutura , Humanos , Lactente , Recém-Nascido , Pulmão/patologia , Pulmão/ultraestrutura , Pulmão/virologia , Masculino , Vírus da Caxumba/genética , Vírus da Caxumba/ultraestrutura , Miocárdio/patologia , Miocárdio/ultraestrutura , Pâncreas/patologia , Pâncreas/ultraestrutura , Reação em Cadeia da Polimerase , Glândulas Salivares/patologia , Glândulas Salivares/ultraestrutura , Glândulas Salivares/virologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA