Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
1.
EMBO J ; 36(5): 679-692, 2017 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-28188244

RESUMO

Vesiculoviruses enter cells by membrane fusion, driven by a large, low-pH-induced, conformational change in the fusion glycoprotein G that involves transition from a trimeric pre-fusion toward a trimeric post-fusion state via monomeric intermediates. Here, we present the structure of the G fusion protein at intermediate pH for two vesiculoviruses, vesicular stomatitis virus (VSV) and Chandipura virus (CHAV), which is responsible for deadly encephalopathies. First, a CHAV G crystal structure shows two intermediate conformations forming a flat dimer of heterodimers. On virions, electron microscopy (EM) and tomography reveal monomeric spikes similar to one of the crystal conformations. In solution, mass spectrometry shows dimers of G. Finally, mutations at a dimer interface, involving fusion domains associated in an antiparallel manner to form an intermolecular ß-sheet, affect G fusion properties. The location of the compensatory mutations restoring fusion activity strongly suggests that this interface is functionally relevant. This work reveals the range of G structural changes and suggests that G monomers can re-associate, through antiparallel interactions between fusion domains, into dimers that play a role at some early stage of the fusion process.


Assuntos
Glicoproteínas/metabolismo , Vesiculovirus/fisiologia , Proteínas do Envelope Viral/metabolismo , Internalização do Vírus , Cristalografia por Raios X , Concentração de Íons de Hidrogênio , Espectrometria de Massas , Microscopia Eletrônica , Modelos Biológicos , Modelos Moleculares , Conformação Proteica , Multimerização Proteica , Tomografia
2.
PLoS Pathog ; 11(3): e1004756, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-25803715

RESUMO

Chandipura virus (CHAV), a member of the vesiculovirus genus, is an emerging human pathogen. As for other rhabdoviruses, CHAV entry into susceptible cells is mediated by its single envelope glycoprotein G which is both involved in receptor recognition and fusion of viral and cellular membranes. Here, we have characterized the fusion properties of CHAV-G. As for vesicular stomatitis virus (VSV, the prototype of the genus) G, fusion is triggered at low pH below 6.5. We have also analyzed the biochemical properties of a soluble form of CHAV-G ectodomain (CHAV-Gth, generated by thermolysin limited-proteolysis of recombinant VSV particles in which the G gene was replaced by that of CHAV). The overall behavior of CHAV-Gth is similar to that previously reported for VSV-Gth. Particularly, CHAV-Gth pre-fusion trimer is not stable in solution and low-pH-induced membrane association of CHAV-Gth is reversible. Furthermore, CHAV-Gth was crystallized in its low pH post-fusion conformation and its structure was determined at 3.6Å resolution. An overall comparison of this structure with the previously reported VSV-Gth post-fusion conformation, shows a high structural similarity as expected from the comparison of primary structure. Among the three domains of G, the pleckstrin homology domain (PHD) appears to be the most divergent and the largest differences are confined to the secondary structure of the major antigenic site of rhabdoviruses. Finally, local differences indicate that CHAV has evolved alternate structural solutions in hinge regions between PH and fusion domains but also distinct pH sensitive switches. Globally the comparison between the post fusion conformation of CHAV and VSV-G highlights several features essential for the protein's function. It also reveals the remarkable plasticity of G in terms of local structures.


Assuntos
Evolução Molecular , Nucleocapsídeo/química , Vesiculovirus/química , Proteínas Virais de Fusão/química , Humanos , Concentração de Íons de Hidrogênio , Nucleocapsídeo/genética , Nucleocapsídeo/metabolismo , Estrutura Terciária de Proteína , Vesiculovirus/genética , Vesiculovirus/metabolismo , Proteínas Virais de Fusão/genética , Proteínas Virais de Fusão/metabolismo
3.
J Virol ; 89(21): 11019-29, 2015 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-26311888

RESUMO

UNLABELLED: The matrix protein (M) of vesicular stomatitis virus (VSV) is involved in virus assembly, budding, gene regulation, and cellular pathogenesis. Using a yeast two-hybrid system, the M globular domain was shown to interact with LMP2, a catalytic subunit of the immunoproteasome (which replaces the standard proteasome catalytic subunit PSMB6). The interaction was validated by coimmunoprecipitation of M and LMP2 in VSV-infected cells. The sites of interaction were characterized. A single mutation of M (I96A) which significantly impairs the interaction between M and LMP2 was identified. We also show that M preferentially binds to the inactive precursor of LMP2 (bearing an N-terminal propeptide which is cleaved upon LMP2 maturation). Furthermore, taking advantage of a sequence alignment between LMP2 and its proteasome homolog, PSMB6 (which does not bind to M), we identified a mutation (L45R) in the S1 pocket where the protein substrate binds prior to cleavage and a second one (D17A) of a conserved residue essential for the catalytic activity, resulting in a reduction of the level of binding to M. The combination of both mutations abolishes the interaction. Taken together, our data indicate that M binds to LMP2 before its incorporation into the immunoproteasome. As the immunoproteasome promotes the generation of major histocompatibility complex (MHC) class I-compatible peptides, a feature which favors the recognition and the elimination of infected cells by CD8 T cells, we suggest that M, by interfering with the immunoproteasome assembly, has evolved a mechanism that allows infected cells to escape detection and elimination by the immune system. IMPORTANCE: The immunoproteasome promotes the generation of MHC class I-compatible peptides, a feature which favors the recognition and the elimination of infected cells by CD8 T cells. Here, we report on the association of vesicular stomatitis virus (VSV) matrix protein (M) with LMP2, one of the immunoproteasome-specific catalytic subunits. M preferentially binds to the LMP2 inactive precursor. The M-binding site on LMP2 is facing inwards in the immunoproteasome and is therefore not accessible to M after its assembly. Hence, M binds to LMP2 before its incorporation into the immunoproteasome. We suggest that VSV M, by interfering with the immunoproteasome assembly, has evolved a mechanism that allows infected cells to escape detection and elimination by the immune system. Modulating this M-induced immunoproteasome impairment might be relevant in order to optimize VSV for oncolytic virotherapy.


Assuntos
Cisteína Endopeptidases/metabolismo , Vesiculovirus/metabolismo , Proteínas da Matriz Viral/metabolismo , Sequência de Bases , Western Blotting , Cisteína Endopeptidases/genética , Células HeLa , Humanos , Imunoprecipitação , Dados de Sequência Molecular , Mutação/genética , Ligação Proteica , Alinhamento de Sequência , Análise de Sequência de DNA , Técnicas do Sistema de Duplo-Híbrido , Proteínas da Matriz Viral/genética
4.
J Virol ; 88(22): 13396-409, 2014 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-25210175

RESUMO

UNLABELLED: Vesicular stomatitis virus (VSV; the prototype rhabdovirus) fusion is triggered at low pH and mediated by glycoprotein G, which undergoes a low-pH-induced structural transition. A unique feature of rhabdovirus G is that its conformational change is reversible. This allows G to recover its native prefusion state at the viral surface after its transport through the acidic Golgi compartments. The crystal structures of G pre- and postfusion states have been elucidated, leading to the identification of several acidic amino acid residues, clustered in the postfusion trimer, as potential pH-sensitive switches controlling the transition back toward the prefusion state. We mutated these residues and produced a panel of single and double mutants whose fusion properties, conformational change characteristics, and ability to pseudotype a virus lacking the glycoprotein gene were assayed. Some of these mutations were also introduced in the genome of recombinant viruses which were further characterized. We show that D268, located in the segment consisting of residues 264 to 273, which refolds into postfusion helix F during G structural transition, is the major pH sensor while D274, D395, and D393 have additional contributions. Furthermore, a single passage of recombinant virus bearing the mutation D268L (which was demonstrated to stabilize the G postfusion state) resulted in a pseudorevertant with a compensatory second mutation, L271P. This revealed that the propensity of the segment of residues 264 to 273 to refold into helix F has to be finely tuned since either an increase (mutation D268L alone) or a decrease (mutation L271P alone) of this propensity is detrimental to the virus. IMPORTANCE: Vesicular stomatitis virus enters cells via endocytosis. Endosome acidification induces a structural transition of its unique glycoprotein (G), which mediates fusion between viral and endosomal membranes. G conformational change is reversible upon increases in pH. This allows G to recover its native prefusion state at the viral surface after its transport through the acidic Golgi compartments. We mutated five acidic residues, proposed to be pH-sensitive switches controlling the structural transition back toward the prefusion state. Our results indicate that residue D268 is the major pH sensor, while other acidic residues have additional contributions, and reveal that the propensity of the segment consisting of residues 264 to 273 to adopt a helical conformation is finely regulated. This segment might be a good target for antiviral compounds.


Assuntos
Glicoproteínas de Membrana/química , Vesiculovirus/fisiologia , Proteínas do Envelope Viral/química , Internalização do Vírus , Animais , Linhagem Celular , Cristalografia por Raios X , Análise Mutacional de DNA , Humanos , Concentração de Íons de Hidrogênio , Glicoproteínas de Membrana/efeitos dos fármacos , Glicoproteínas de Membrana/genética , Modelos Moleculares , Proteínas Mutantes/química , Proteínas Mutantes/efeitos dos fármacos , Proteínas Mutantes/genética , Conformação Proteica , Proteínas do Envelope Viral/efeitos dos fármacos , Proteínas do Envelope Viral/genética
5.
Philos Trans R Soc Lond B Biol Sci ; 379(1911): 20230154, 2024 Oct 07.
Artigo em Inglês | MEDLINE | ID: mdl-39155719

RESUMO

A fundamental component of human cognition is the ability to intuitively reason about behaviours of objects and systems in the physical world without resorting to explicit scientific knowledge. This skill was traditionally considered a symbolic process. However, in the last decades, there has been a shift towards ideas of embodiment, suggesting that accessing physical knowledge and predicting physical outcomes is grounded in bodily interactions with the environment. Infants and children, who learn mainly through their embodied experiences, serve as a model to probe the link between reasoning and physical concepts. Here, we tested school-aged children (5- to 15-year-olds) in online reasoning games that involve different physical action concepts such as supporting, launching and clearing. We assessed changes in children's performance and strategies over development and their relationships with the different action concepts. Children reasoned more accurately in problems that involved supporting actions compared to launching or clearing actions. Moreover, when children failed, they were more strategic in subsequent attempts when problems involved support rather than launching or clearing. Children improved with age, but improvements differed across action concepts. Our findings suggest that accessing physical knowledge and predicting physical events are affected by action concepts, and those effects change over development. This article is part of the theme issue 'Minds in movement: embodied cognition in the age of artificial intelligence'.


Assuntos
Desenvolvimento Infantil , Cognição , Humanos , Criança , Adolescente , Masculino , Feminino , Pré-Escolar , Desenvolvimento Infantil/fisiologia , Formação de Conceito , Pensamento/fisiologia , Resolução de Problemas
6.
J Gen Virol ; 93(Pt 4): 857-865, 2012 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-22190013

RESUMO

Vesicular stomatitis virus (VSV) matrix protein (M) has a flexible amino-terminal part that recruits cellular partners. It contains a dynamin-binding site that is required for efficient virus assembly, and two motifs, (24)PPPY(27) and (37)PSAP(40), that constitute potential late domains. Late domains are present in proteins of several enveloped viruses and are involved in the ultimate step of the budding process (i.e. fission between viral and cellular membranes). In baby hamster kidney (BHK)-21 cells, it has been demonstrated that the (24)PPPY(27) motif binds the Nedd4 (neuronal precursor cell-expressed developmentally downregulated 4) E3 ubiquitin ligase for efficient virus budding and that the (37)PSAP(40) motif, although conserved among M proteins of vesiculoviruses, does not possess late-domain activity. In this study, we have re-examined the contribution of the PSAP motif to VSV budding. First, we demonstrate that VSV M indeed binds TSG101 [tumour susceptibility gene 101; a component of the ESCRT1 (endosomal sorting complex required for transport 1)] through its PSAP motif. Second, we analysed the phenotype of several recombinant mutants. We show that a double mutant with point mutations in both the PSAP and the PPPY motifs is impaired compared with a single mutant in the PPPY motif, indicating that the PSAP motif partially compensates for the lack of the PPPY motif. Mutants' phenotypes depend on cell lines: in CERA (chicken embryo-related, Alger clone) cells, a recombinant virus with a single mutation in the PSAP motif was impaired compared with the wild type, and a mutant with a single mutation in the dynamin-binding motif was much less impaired in Vero cells than in BSR (clones of BHK-21) cells. These results have implications for the VSV budding pathway that will be discussed.


Assuntos
Vesiculovirus/genética , Proteínas da Matriz Viral/genética , Animais , Linhagem Celular , Cricetinae , Dinaminas/metabolismo , Complexos Endossomais de Distribuição Requeridos para Transporte/metabolismo , Humanos , Microscopia Eletrônica , Mutagênese Sítio-Dirigida , Mutação/genética , Mutação/fisiologia , Ubiquitina-Proteína Ligases Nedd4 , Fenótipo , Ubiquitina-Proteína Ligases/metabolismo , Estomatite Vesicular/metabolismo , Estomatite Vesicular/virologia , Vesiculovirus/fisiologia , Proteínas da Matriz Viral/fisiologia , Montagem de Vírus/genética , Montagem de Vírus/fisiologia
7.
J Virol ; 84(24): 12609-18, 2010 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-20943988

RESUMO

Matrix proteins (M) direct the process of assembly and budding of viruses belonging to the Mononegavirales order. Using the two-hybrid system, the amino-terminal part of vesicular stomatitis virus (VSV) M was shown to interact with dynamin pleckstrin homology domain. This interaction was confirmed by coimmunoprecipitation of both proteins in cells transfected by a plasmid encoding a c-myc-tagged dynamin and infected by VSV. A role for dynamin in the viral cycle (in addition to its role in virion endocytosis) was suggested by the fact that a late stage of the viral cycle was sensitive to dynasore. By alanine scanning, we identified a single mutation of M protein that abolished this interaction and reduced virus yield. The adaptation of mutant virus (M.L4A) occurred rapidly, allowing the isolation of revertants, among which the M protein, despite having an amino acid sequence distinct from that of the wild type, recovered a significant level of interaction with dynamin. This proved that the mutant phenotype was due to the loss of interaction between M and dynamin. The infectious cycle of the mutant virus M.L4A was blocked at a late stage, resulting in a quasi-absence of bullet-shaped viruses in the process of budding at the cell membrane. This was associated with an accumulation of nucleocapsids at the periphery of the cell and a different pattern of VSV glycoprotein localization. Finally, we showed that M-dynamin interaction affects clathrin-dependent endocytosis. Our study suggests that hijacking the endocytic pathway might be an important feature for enveloped virus assembly and budding at the plasma membrane.


Assuntos
Dinaminas/metabolismo , Nucleocapsídeo/metabolismo , Vesiculovirus/fisiologia , Proteínas da Matriz Viral/metabolismo , Montagem de Vírus/fisiologia , Células Cultivadas , Clatrina/metabolismo , Dinaminas/genética , Endocitose/fisiologia , Endossomos/metabolismo , Imunofluorescência , Humanos , Imunoprecipitação , Rim/virologia , Mutação/genética , Técnicas do Sistema de Duplo-Híbrido , Proteínas da Matriz Viral/genética , Liberação de Vírus/fisiologia
8.
Cell Rep ; 32(7): 108042, 2020 08 18.
Artigo em Inglês | MEDLINE | ID: mdl-32814045

RESUMO

VSV fusion machinery, like that of many other enveloped viruses, is triggered at low pH in endosomes after virion endocytosis. It was suggested that some histidines could play the role of pH-sensitive switches. By mutating histidine residues H22, H60, H132, H162, H389, H397, H407, and H409, we demonstrate that residues H389 and D280, facing each other in the six-helix bundle of the post-fusion state, and more prominently H407, located at the interface between the C-terminal part of the ectodomain and the fusion domain, are crucial for fusion. Passages of recombinant viruses bearing mutant G resulted in the selection of compensatory mutations. Thus, the H407A mutation in G resulted in two independent compensatory mutants, L396I and S422I. Together with a crystal structure of G, presented here, which extends our knowledge of G pre-fusion structure, this indicates that the conformational transition is initiated by refolding of the C-terminal part of the G ectodomain.


Assuntos
Vírus da Estomatite Vesicular Indiana/genética , Proteínas do Envelope Viral/genética , Proteínas Virais de Fusão/genética , Estrutura Molecular , Transfecção
9.
Nat Commun ; 9(1): 1029, 2018 03 12.
Artigo em Inglês | MEDLINE | ID: mdl-29531262

RESUMO

Vesicular stomatitis virus (VSV) is an oncolytic rhabdovirus and its glycoprotein G is widely used to pseudotype other viruses for gene therapy. Low-density lipoprotein receptor (LDL-R) serves as a major entry receptor for VSV. Here we report two crystal structures of VSV G in complex with two distinct cysteine-rich domains (CR2 and CR3) of LDL-R, showing that their binding sites on G are identical. We identify two basic residues on G, which are essential for its interaction with CR2 and CR3. Mutating these residues abolishes VSV infectivity even though VSV can use alternative receptors, indicating that all VSV receptors are members of the LDL-R family. Collectively, our data suggest that VSV G has specifically evolved to interact with receptor CR domains. These structural insights into the interaction between VSV G and host cell receptors provide a basis for the design of recombinant viruses with an altered tropism.


Assuntos
Glicoproteínas de Membrana/metabolismo , Receptores de LDL/química , Receptores de LDL/metabolismo , Receptores Virais/química , Receptores Virais/metabolismo , Estomatite Vesicular/metabolismo , Vírus da Estomatite Vesicular Indiana/metabolismo , Proteínas do Envelope Viral/metabolismo , Humanos , Glicoproteínas de Membrana/química , Glicoproteínas de Membrana/genética , Família Multigênica , Ligação Proteica , Domínios Proteicos , Receptores de LDL/genética , Receptores Virais/genética , Estomatite Vesicular/genética , Estomatite Vesicular/virologia , Vírus da Estomatite Vesicular Indiana/química , Vírus da Estomatite Vesicular Indiana/genética , Proteínas do Envelope Viral/química , Proteínas do Envelope Viral/genética
10.
J Cell Biol ; 191(1): 199-210, 2010 Oct 04.
Artigo em Inglês | MEDLINE | ID: mdl-20921141

RESUMO

The entry of enveloped viruses into cells requires the fusion of viral and cellular membranes, driven by conformational changes in viral glycoproteins. Many studies have shown that fusion involves the cooperative action of a large number of these glycoproteins, but the underlying mechanisms are unknown. We used electron microscopy and tomography to study the low pH-induced fusion reaction catalyzed by vesicular stomatitis virus glycoprotein (G). Pre- and post-fusion crystal structures were observed on virions at high and low pH, respectively. Individual fusion events with liposomes were also visualized. Fusion appears to be driven by two successive structural rearrangements of G at different sites on the virion. Fusion is initiated at the flat base of the particle. Glycoproteins located outside the contact zone between virions and liposomes then reorganize into regular arrays. We suggest that the formation of these arrays, which have been shown to be an intrinsic property of the G ectodomain, induces membrane constraints, achieving the fusion reaction.


Assuntos
Fusão de Membrana/fisiologia , Glicoproteínas de Membrana/fisiologia , Vírus da Estomatite Vesicular Indiana/patogenicidade , Proteínas Virais de Fusão/fisiologia , Proteínas Virais/fisiologia , Internalização do Vírus , Cristalografia por Raios X , Concentração de Íons de Hidrogênio , Lipossomos/ultraestrutura , Glicoproteínas de Membrana/química , Estrutura Terciária de Proteína , Vírus da Estomatite Vesicular Indiana/metabolismo , Vírus da Estomatite Vesicular Indiana/ultraestrutura , Proteínas Virais de Fusão/química , Proteínas Virais/química , Vírion/metabolismo , Vírion/patogenicidade , Vírion/ultraestrutura
11.
Virology ; 334(2): 284-93, 2005 Apr 10.
Artigo em Inglês | MEDLINE | ID: mdl-15780878

RESUMO

Rabies virus P protein is a co-factor of the viral RNA polymerase. It has been shown previously that P mRNA directs the synthesis of four N-terminally truncated P products P2, P3, P4, and P5 due to translational initiation by a leaky scanning mechanism at internal Met codons. Whereas P and P2 are located in the cytoplasm, P3, P4, and P5 are found in the nucleus. Here, we have analyzed the molecular basis of the subcellular localization of these proteins. Using deletion mutants fused to GFP protein, we show the presence of a nuclear localization signal (NLS) in the C-terminal part of P (172-297). This domain contains a short lysine-rich stretch ((211)KKYK(214)) located in close proximity with arginine 260 as revealed by the crystal structure of P. We demonstrate the critical role of lysine 214 and arginine 260 in NLS activity. In the presence of Leptomycin B, P is retained in the nucleus indicating that it contains a CRM1-dependent nuclear export signal (NES). The subcellular distribution of P deletion mutants indicates that the domain responsible for export is the amino-terminal part of the protein. The use of fusion proteins that have amino terminal fragments of P fused to beta-galactosidase containing the NLS of SV40 T antigen allows us to identify a NES between residues 49 and 58. The localization of NLS and NES determines the cellular distribution of the P gene products.


Assuntos
Núcleo Celular/metabolismo , Carioferinas/metabolismo , Sinais de Localização Nuclear/metabolismo , Fosfoproteínas/metabolismo , Vírus da Raiva/metabolismo , Receptores Citoplasmáticos e Nucleares/metabolismo , Proteínas Estruturais Virais/metabolismo , Transporte Ativo do Núcleo Celular , Sequência de Aminoácidos , Animais , Linhagem Celular , Cricetinae , Chaperonas Moleculares , Dados de Sequência Molecular , Mutagênese Sítio-Dirigida , Sinais de Localização Nuclear/química , Fosfoproteínas/química , Fosfoproteínas/genética , Vírus da Raiva/patogenicidade , Proteínas Estruturais Virais/química , Proteínas Estruturais Virais/genética , Proteína Exportina 1
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA