Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 25
Filtrar
1.
PLoS Pathog ; 18(12): e1011022, 2022 12.
Artigo em Inglês | MEDLINE | ID: mdl-36480574

RESUMO

Rabies virus (RABV) transcription and replication take place within viral factories having liquid properties, called Negri bodies (NBs), that are formed by liquid-liquid phase separation (LLPS). The co-expression of RABV nucleoprotein (N) and phosphoprotein (P) in mammalian cells is sufficient to induce the formation of cytoplasmic biocondensates having properties that are like those of NBs. This cellular minimal system was previously used to identify P domains that are essential for biocondensates formation. Here, we constructed fluorescent versions of N and analyzed by FRAP their dynamics inside the biocondensates formed in this minimal system as well as in NBs of RABV-infected cells using FRAP. The behavior of N appears to be different of P as there was no fluorescence recovery of N proteins after photobleaching. We also identified arginine residues as well as two exposed loops of N involved in condensates formation. Corresponding N mutants exhibited distinct phenotypes in infected cells ranging from co-localization with NBs to exclusion from them associated with a dominant-negative effect on infection. We also demonstrated that in vitro, in crowded environments, purified P as well as purified N0-P complex (in which N is RNA-free) form liquid condensates. We identified P domains required for LLPS in this acellular system. P condensates were shown to associate with liposomes, concentrate RNA, and undergo a liquid-gel transition upon ageing. Conversely, N0-P droplets were disrupted upon incubation with RNA. Taken together, our data emphasize the central role of P in NBs formation and reveal some physicochemical features of P and N0-P droplets relevant for explaining NBs properties such as their envelopment by cellular membranes at late stages of infection and nucleocapsids ejections from the viral factories.


Assuntos
Vírus da Raiva , Raiva , Animais , Vírus da Raiva/genética , Vírus da Raiva/metabolismo , Nucleoproteínas/genética , Raiva/metabolismo , Nucleocapsídeo/metabolismo , Fosfoproteínas/genética , Fosfoproteínas/metabolismo , Replicação Viral , Mamíferos
2.
PLoS Pathog ; 16(3): e1008383, 2020 03.
Artigo em Inglês | MEDLINE | ID: mdl-32150590

RESUMO

Mokola virus (MOKV) belongs to the lyssavirus genus. As other genus members-including rabies virus (RABV)-it causes deadly encephalitis in mammals. MOKV entry into host cells is mediated by its transmembrane glycoprotein G. First, G binds cellular receptors, triggering virion endocytosis. Then, in the acidic endosomal environment, G undergoes a conformational change from its pre- toward its post-fusion state that catalyzes the merger of the viral and endosomal membranes. Here, we have determined the crystal structure of a soluble MOKV G ectodomain in which the hydrophobic fusion loops have been replaced by more hydrophilic sequences. The crystal structure corresponds to a monomer that is similar to the protomer of the trimeric post-fusion state of vesicular stomatitis virus (VSV) G. However, by electron microscopy, we show that, at low pH, at the surface of pseudotyped VSV, MOKV spikes adopt the trimeric post-fusion conformation and have a tendency to reorganize into regular arrays. Sequence alignment between MOKV G and RABV G allows a precise location of RABV G antigenic sites. Repositioning MOKV G domains on VSV G pre-fusion structure reveals that antigenic sites are located in the most exposed part of the molecule in its pre-fusion conformation and are therefore very accessible to antibodies. Furthermore, the structure allows the identification of pH-sensitive molecular switches. Specifically, the long helix, which constitutes the core of the post-fusion trimer for class III fusion glycoproteins, contains many acidic residues located at the trimeric interface. Several of them, aligned along the helix, point toward the trimer axis. They have to be protonated for the post-fusion trimer to be stable. At high pH, when they are negatively charged, they destabilize the interface, which explains the conformational change reversibility. Finally, the present structure will be of great help to perform rational mutagenesis on lyssavirus glycoproteins.


Assuntos
Lyssavirus/química , Multimerização Proteica , Proteínas Virais de Fusão/química , Cristalografia por Raios X , Estrutura Quaternária de Proteína , Estrutura Secundária de Proteína
3.
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
4.
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
5.
PLoS Pathog ; 8(2): e1002556, 2012 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-22383886

RESUMO

Entry of enveloped viruses requires fusion of viral and cellular membranes, driven by conformational changes of viral glycoproteins. Crystal structures provide static pictures of pre- and post-fusion conformations of these proteins but the transition pathway remains elusive. Here, using several biophysical techniques, including analytical ultracentrifugation, circular dichroïsm, electron microscopy and small angle X-ray scattering, we have characterized the low-pH-induced fusogenic structural transition of a soluble form of vesicular stomatitis virus (VSV) glycoprotein G ectodomain (G(th), aa residues 1-422, the fragment that was previously crystallized). While the post-fusion trimer is the major species detected at low pH, the pre-fusion trimer is not detected in solution. Rather, at high pH, G(th) is a flexible monomer that explores a large conformational space. The monomeric population exhibits a marked pH-dependence and adopts more elongated conformations when pH decreases. Furthermore, large relative movements of domains are detected in absence of significant secondary structure modification. Solution studies are complemented by electron micrographs of negatively stained viral particles in which monomeric ectodomains of G are observed at the viral surface at both pH 7.5 and pH 6.7. We propose that the monomers are intermediates during the conformational change and thus that VSV G trimers dissociate at the viral surface during the structural transition.


Assuntos
Glicoproteínas de Membrana/química , Vesiculovirus , Dicroísmo Circular , Concentração de Íons de Hidrogênio , Lipossomos/metabolismo , Glicoproteínas de Membrana/metabolismo , Modelos Moleculares , Conformação Proteica , Multimerização Proteica , Estrutura Secundária de Proteína/fisiologia , Vesiculovirus/química , Vesiculovirus/metabolismo , Proteínas do Envelope Viral/química , Proteínas do Envelope Viral/metabolismo , Proteínas Virais de Fusão/química , Proteínas Virais de Fusão/metabolismo , Vírion/metabolismo
6.
Microbes Infect ; : 105402, 2024 Aug 08.
Artigo em Inglês | MEDLINE | ID: mdl-39127089

RESUMO

During a viral infection, several membraneless compartments with liquid properties are formed. They can be of viral origin concentrating viral proteins and nucleic acids, and harboring essential stages of the viral cycle, or of cellular origin containing components involved in innate immunity. This is a paradigm shift in our understanding of viral replication and the interaction between viruses and innate cellular immunity.

7.
Mol Biol Evol ; 29(1): 381-90, 2012 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-21917725

RESUMO

Retroelements represent a considerable fraction of many eukaryotic genomes and are considered major drives for adaptive genetic innovations. Recent discoveries showed that despite not normally using DNA intermediates like retroviruses do, Mononegaviruses (i.e., viruses with nonsegmented, negative-sense RNA genomes) can integrate gene fragments into the genomes of their hosts. This was shown for Bornaviridae and Filoviridae, the sequences of which have been found integrated into the germ line cells of many vertebrate hosts. Here, we show that Rhabdoviridae sequences, the major Mononegavirales family, have integrated only into the genomes of arthropod species. We identified 185 integrated rhabdoviral elements (IREs) coding for nucleoproteins, glycoproteins, or RNA-dependent RNA polymerases; they were mostly found in the genomes of the mosquito Aedes aegypti and the blacklegged tick Ixodes scapularis. Phylogenetic analyses showed that most IREs in A. aegypti derived from multiple independent integration events. Since RNA viruses are submitted to much higher substitution rates as compared with their hosts, IREs thus represent fossil traces of the diversity of extinct Rhabdoviruses. Furthermore, analyses of orthologous IREs in A. aegypti field mosquitoes sampled worldwide identified an integrated polymerase IRE fragment that appeared under purifying selection within several million years, which supports a functional role in the host's biology. These results show that A. aegypti was subjected to repeated Rhabdovirus infectious episodes during its evolution history, which led to the accumulation of many integrated sequences. They also suggest that like retroviruses, integrated rhabdoviral sequences may participate actively in the evolution of their hosts.


Assuntos
Artrópodes/genética , Evolução Molecular , Genoma de Inseto , Rhabdoviridae/genética , Animais , Teorema de Bayes , Bases de Dados Genéticas , Transferência Genética Horizontal , Filogenia
8.
Artigo em Inglês | MEDLINE | ID: mdl-22949203

RESUMO

Fusion in members of the Rhabdoviridae virus family is mediated by the G glycoprotein. At low pH, the G glycoprotein catalyzes fusion between viral and endosomal membranes by undergoing a major conformational change from a pre-fusion trimer to a post-fusion trimer. The structure of the G glycoprotein from vesicular stomatitis virus (VSV G), the prototype of Vesiculovirus, has recently been solved in its trimeric pre-fusion and post-fusion conformations; however, little is known about the structural details of the transition. In this work, a soluble form of the ectodomain of Chandipura virus G glycoprotein (CHAV G(th)) was purified using limited proteolysis of purified virus; this soluble ectodomain was also crystallized. This protein shares 41% amino-acid identity with VSV G and thus its structure could provide further clues about the structural transition of rhabdoviral glycoproteins induced by low pH. Crystals of CHAV G(th) obtained at pH 7.5 diffracted X-rays to 3.1 Å resolution. These crystals belonged to the orthorhombic space group P2(1)2(1)2, with unit-cell parameters a = 150.3, b = 228.2, c = 78.8 Å. Preliminary analysis of the data based on the space group and the self-rotation function indicated that there was no trimeric association of the protomers. This unusual oligomeric status could result from the presence of fusion intermediates in the crystal.


Assuntos
Glicoproteínas/química , Vesiculovirus/química , Proteínas Virais/química , Cristalização , Cristalografia por Raios X
9.
Biochim Biophys Acta Mol Cell Res ; 1867(12): 118831, 2020 12.
Artigo em Inglês | MEDLINE | ID: mdl-32835749

RESUMO

Viruses reshape the organization of the cell interior to achieve different steps of their cellular cycle. Particularly, viral replication and assembly often take place in viral factories where specific viral and cellular proteins as well as nucleic acids concentrate. Viral factories can be either membrane-delimited or devoid of any cellular membranes. In the latter case, they are referred as membrane-less replication compartments. The most emblematic ones are the Negri bodies, which are inclusion bodies that constitute the hallmark of rabies virus infection. Interestingly, Negri bodies and several other viral replication compartments have been shown to arise from a liquid-liquid phase separation process and, thus, constitute a new class of liquid organelles. This is a paradigm shift in the field of virus replication. Here, we review the different aspects of membrane-less virus replication compartments with a focus on the Mononegavirales order and discuss their interactions with the host cell machineries and the cytoskeleton. We particularly examine the interplay between viral factories and the cellular innate immune response, of which several components also form membrane-less condensates in infected cells.


Assuntos
Corpos de Inclusão Viral/genética , Raiva/genética , Compartimentos de Replicação Viral , Replicação Viral/genética , Membrana Celular/genética , Corpos de Inclusão Viral/virologia , Raiva/virologia , Vírus da Raiva/genética , Vírus da Raiva/patogenicidade , Proteínas Virais/genética
10.
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
11.
Adv Virus Res ; 104: 147-183, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31439148

RESUMO

Rhabdoviruses are enveloped viruses with a negative-sense single strand RNA genome and are widespread among a great variety of organisms. In their membrane, they have a single glycoprotein (G) that mediates both virus attachment to cellular receptors and fusion between viral and endosomal membranes allowing viral genome release in the cytoplasm. We present structural and cellular aspects of Rhabdovirus entry into their host cell with a focus on vesicular stomatitis virus (VSV) and rabies virus (RABV) for which the early events of the viral cycle have been extensively studied. Recent data have shown that the only VSV receptors are the members of the LDL-R family. This is in contrast with RABV for which multiple receptors belonging to unrelated families have been identified. Despite having different receptors, after attachment, rhabdovirus internalization occurs through clathrin-mediated endocytosis (CME) in an actin-dependent manner. There are still debates about the exact endocytic pathway of VSV in the cell and on RABV transport in the neuronal axon. In any case, fusion is triggered in the endosomal vesicle via a low-pH induced structural rearrangement of G from its pre- to its postfusion conformation. Vesiculovirus G is one of the best characterized fusion glycoproteins as the previously reported crystal structures of the pre- and postfusion states have been recently completed by those of intermediates during the structural transition. Understanding the entry pathway of rhabdoviruses may have strong impact in biotechnologies as, for example, VSV G is used for pseudotyping lentiviruses to promote efficient transduction, and VSV is a promising oncolytic virus.


Assuntos
Interações Hospedeiro-Patógeno , Vírus da Raiva/fisiologia , Vesiculovirus/fisiologia , Ligação Viral , Internalização do Vírus , Endocitose , Glicoproteínas/metabolismo , Receptores Virais/metabolismo , Proteínas do Envelope Viral/metabolismo
12.
Virologie (Montrouge) ; 12(6): 407-418, 2008 Dec 01.
Artigo em Francês | MEDLINE | ID: mdl-36131396

RESUMO

Glycoprotein G of the rhabdoviruses is involved in both receptor recognition at the host cell surface and membrane fusion via a low pHinduced structural rearrangement. G is an atypical fusion protein as there is a pH-dependent equilibrium between the pre- and post-fusion conformations of the protein. The atomic structures of the pre- and post-fusion conformations reveal that G is homologous to both glycoprotein gB of herpesviruses and gp64 of baculovirus and also that it combines features of the previously characterized class I and class II fusion proteins. Comparison of the structures of G pre- and post-fusion states reveals an extensive structural reorganization of the molecule that resembles that of paramyxovirus fusion protein F. It also allows to localize the fusion loops and to identify conserved key residues that constitute pH-sensitive molecular switches. Finally, the fusion properties and the structures of G also reveal some particularities that invite us to reconsider a few dogmas concerning fusion proteins.

13.
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
15.
J Mol Biol ; 339(2): 301-12, 2004 May 28.
Artigo em Inglês | MEDLINE | ID: mdl-15136034

RESUMO

Recombinant measles virus nucleoprotein (N) was produced in insect cells where it bound to cellular RNA to form helical N-RNA structures. These structures were observed by electron microscopy but were too flexible for high-resolution image analysis. Removal of the C-terminal tail of N by trypsin treatment resulted in structures that were much more rigid and seemed more regular. Several methods of image analysis were employed in order to make a helical reconstruction of the digested N-RNA. During this analysis, it became clear that the apparently regular coils of digested N-RNA consisted of a series of closely related helical states. The iterative helical real space reconstruction method allowed the identification of two helical states for which a reconstruction could be calculated. The model with the highest resolution shows N monomers that consist of three domains and that are connected to their neighbours by two narrow connections, one close to the helical axis and another toward the middle of the monomers. There are no connections between N molecules in subsequent helical turns. After labelling the RNA in the structure with cis-platinum, the connection closest to the helical axis increased in density, suggesting the position of the RNA. The shapes of the monomers of the nucleoproteins of influenza virus, rabies virus (both determined before) and that of measles virus (determined here) are all similar, whereas the overall shapes of their respective N-RNAs (nucleocapsids) is very different. This is probably due to the position and number of the connections between the N subunits in the N-RNA, one for influenza virus allowing much flexibility, two for rabies virus at either end of the N molecules leading to ribbons and two for measles virus leading to the typical paramyxovirus helical nucleocapsid.


Assuntos
Vírus do Sarampo/genética , Conformação de Ácido Nucleico , Nucleoproteínas/genética , RNA Viral/química , Tripsina/administração & dosagem , Sequência de Aminoácidos , Animais , Microscopia Crioeletrônica , RNA Viral/ultraestrutura , Proteínas Recombinantes/genética , Recombinação Genética , Spodoptera
16.
Curr Opin Struct Biol ; 33: 52-60, 2015 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-26277251

RESUMO

Enveloped viruses enter the cell by fusing their envelope with a cellular membrane. Fusion is catalyzed by conformational changes of viral glycoproteins from pre-fusion to post-fusion states. Structural studies have defined three classes of viral fusion glycoproteins. Class III comprises the fusion glycoproteins from rhabdoviruses (G), herpesviruses (gB), and baculoviruses (GP64). Although sharing the same fold, those glycoproteins exhibit striking differences in their modes of activation and interaction with the target membrane. Furthermore, for gB and GP64, only the post-fusion structure is known and the extent of their conformational change is still an unresolved issue. Further structural studies are therefore required to get a detailed insight in the working of those fusion machines.


Assuntos
Proteínas Virais de Fusão/química , Proteínas Virais de Fusão/genética , Baculoviridae/genética , Membrana Celular/fisiologia , Herpesviridae/genética , Concentração de Íons de Hidrogênio , Conformação Proteica , Rhabdoviridae/genética
18.
Curr Opin Virol ; 3(2): 143-50, 2013 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-23562213

RESUMO

Entry of enveloped viruses into cells requires the fusion of viral and cellular membranes, driven by conformational changes in viral glycoproteins. Three different classes of viral fusion proteins have been hitherto identified based on common structural elements. Crystal structures have provided static pictures of pre-fusion and post-fusion conformations of these proteins and have revealed the dramatic reorganization of the molecules, but the transition pathway remains elusive. In this review, we will focus on recent data aiming to characterize intermediate structures during the conformational change. All these data support the existence of a pre-hairpin intermediate, but its oligomeric status is still a matter of debate.


Assuntos
Proteínas Virais de Fusão/química , Proteínas Virais de Fusão/metabolismo , Internalização do Vírus , Cristalografia por Raios X , Modelos Biológicos , Modelos Moleculares , Conformação Proteica
19.
Viruses ; 4(1): 117-39, 2012 01.
Artigo em Inglês | MEDLINE | ID: mdl-22355455

RESUMO

Rhabdoviruses enter the cell via the endocytic pathway and subsequently fuse with a cellular membrane within the acidic environment of the endosome. Both receptor recognition and membrane fusion are mediated by a single transmembrane viral glycoprotein (G). Fusion is triggered via a low-pH induced structural rearrangement. G is an atypical fusion protein as there is a pH-dependent equilibrium between its pre- and post-fusion conformations. The elucidation of the atomic structures of these two conformations for the vesicular stomatitis virus (VSV) G has revealed that it is different from the previously characterized class I and class II fusion proteins. In this review, the pre- and post-fusion VSV G structures are presented in detail demonstrating that G combines the features of the class I and class II fusion proteins. In addition to these similarities, these G structures also reveal some particularities that expand our understanding of the working of fusion machineries. Combined with data from recent studies that revealed the cellular aspects of the initial stages of rhabdovirus infection, all these data give an integrated view of the entry pathway of rhabdoviruses into their host cell.


Assuntos
Rhabdoviridae/fisiologia , Internalização do Vírus , Animais , Cristalografia por Raios X , Endocitose , Endossomos/virologia , Genoma Viral , Humanos , Concentração de Íons de Hidrogênio , Fusão de Membrana , Glicoproteínas de Membrana/química , Glicoproteínas de Membrana/fisiologia , Camundongos , Neurônios/virologia , Conformação Proteica , Receptores Virais/fisiologia , Rhabdoviridae/classificação , Rhabdoviridae/genética , Rhabdoviridae/ultraestrutura , Infecções por Rhabdoviridae/virologia , Proteínas do Envelope Viral/química , Proteínas do Envelope Viral/fisiologia , Proteínas Virais/genética , Proteínas Virais/fisiologia
20.
Adv Virus Res ; 79: 1-22, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21601039

RESUMO

Rabies virus (RABV) is a negative-stranded RNA virus. Its genome is tightly encapsidated by the viral nucleoprotein (N) and this RNA-N complex is the template for transcription and replication by the viral RNA-dependent RNA polymerase (L) and its cofactor, the phosphoprotein (P). We present molecular, structural, and cellular aspects of RABV transcription and replication. We first summarize the characteristics and molecular biology of both RNA synthesis processes. We then discuss biochemical and structural data on the viral proteins (N, P, and L) and their interactions with regard to their role in viral transcription and replication. Finally, we review evidence that rabies viral transcription and replication take place in cytoplasmic inclusion bodies formed in RABV-infected cells and discuss the role of this cellular compartmentalization.


Assuntos
RNA Viral/biossíntese , Vírus da Raiva/fisiologia , Transcrição Gênica , Replicação Viral , Animais , Humanos , Corpos de Inclusão Viral , Proteínas Virais/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA