Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 46
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Viruses ; 14(4)2022 03 28.
Artigo em Inglês | MEDLINE | ID: mdl-35458429

RESUMO

Focusing on the transmembrane domains (TMDs) of viral fusion and channel-forming proteins (VCPs), experimentally available and newly generated peptides in an ideal conformation of the S and E proteins of severe acute respiratory syndrome coronavirus type 2 (SARS-CoV-2) and SARS-CoV, gp41 and Vpu, both of human immunodeficiency virus type 1 (HIV-1), haemagglutinin and M2 of influenza A, as well as gB of herpes simplex virus (HSV), are embedded in a fully hydrated lipid bilayer and used in multi-nanosecond molecular dynamics simulations. It is aimed to identify differences in the dynamics of the individual TMDs of the two types of viral membrane proteins. The assumption is made that the dynamics of the individual TMDs are decoupled from their extra-membrane domains, and that the mechanics of the TMDs are distinct from each other due to the different mechanism of function of the two types of proteins. The diffusivity coefficient (DC) of the translational and rotational diffusion is decreased in the oligomeric state of the TMDs compared to those values when calculated from simulations in their monomeric state. When comparing the calculations for two different lengths of the TMD, a longer full peptide and a shorter purely TMD stretch, (i) the difference of the calculated DCs begins to level out when the difference exceeds approximately 15 amino acids per peptide chain, and (ii) the channel protein rotational DC is the most affected diffusion parameter. The rotational dynamics of the individual amino acids within the middle section of the TMDs of the fusion peptides remain high upon oligomerization, but decrease for the channel peptides, with an increasing number of monomers forming the oligomeric state, suggesting an entropic penalty on oligomerization for the latter.


Assuntos
COVID-19 , Canais Iônicos , Simulação de Dinâmica Molecular , Proteínas Virais de Fusão , Aminoácidos , Humanos , Canais Iônicos/ultraestrutura , Peptídeos/química , SARS-CoV-2 , Proteínas Virais de Fusão/ultraestrutura
2.
MAbs ; 13(1): 1955812, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34420474

RESUMO

Respiratory syncytial virus (RSV) is the most common cause of acute lower respiratory tract infections resulting in medical intervention and hospitalizations during infancy and early childhood, and vaccination against RSV remains a public health priority. The RSV F glycoprotein is a major target of neutralizing antibodies, and the prefusion stabilized form of F (DS-Cav1) is under investigation as a vaccine antigen. AM14 is a human monoclonal antibody with the exclusive capacity of binding an epitope on prefusion F (PreF), which spans two F protomers. The quality of recognizing a trimer-specific epitope makes AM14 valuable for probing PreF-based immunogen conformation and functionality during vaccine production. Currently, only a low-resolution (5.5 Å) X-ray structure is available of the PreF-AM14 complex, revealing few reliable details of the interface. Here, we perform complementary structural studies using X-ray crystallography and cryo-electron microscopy (cryo-EM) to provide improved resolution structures at 3.6 Å and 3.4 Å resolutions, respectively. Both X-ray and cryo-EM structures provide clear side-chain densities, which allow for accurate mapping of the AM14 epitope on DS-Cav1. The structures help rationalize the molecular basis for AM14 loss of binding to RSV F monoclonal antibody-resistant mutants and reveal flexibility for the side chain of a key antigenic residue on PreF. This work provides the basis for a comprehensive understanding of RSV F trimer specificity with implications in vaccine design and quality assessment of PreF-based immunogens.


Assuntos
Anticorpos Monoclonais/ultraestrutura , Fragmentos Fab das Imunoglobulinas/ultraestrutura , Infecções por Vírus Respiratório Sincicial/prevenção & controle , Vacinas contra Vírus Sincicial Respiratório/química , Vírus Sincicial Respiratório Humano/imunologia , Proteínas Virais de Fusão/imunologia , Proteínas Virais de Fusão/ultraestrutura , Animais , Anticorpos Monoclonais/imunologia , Anticorpos Monoclonais/metabolismo , Sítios de Ligação de Anticorpos , Células CHO , Cricetulus , Microscopia Crioeletrônica , Cristalografia por Raios X , Epitopos , Fragmentos Fab das Imunoglobulinas/imunologia , Modelos Moleculares , Mutação , Conformação Proteica , Infecções por Vírus Respiratório Sincicial/imunologia , Infecções por Vírus Respiratório Sincicial/virologia , Vacinas contra Vírus Sincicial Respiratório/genética , Vacinas contra Vírus Sincicial Respiratório/imunologia , Vírus Sincicial Respiratório Humano/genética , Vírus Sincicial Respiratório Humano/patogenicidade , Relação Estrutura-Atividade , Desenvolvimento de Vacinas , Proteínas Virais de Fusão/genética
3.
Nat Commun ; 11(1): 4646, 2020 09 16.
Artigo em Inglês | MEDLINE | ID: mdl-32938911

RESUMO

The human betacoronaviruses HKU1 and OC43 (subgenus Embecovirus) arose from separate zoonotic introductions, OC43 relatively recently and HKU1 apparently much longer ago. Embecovirus particles contain two surface projections called spike (S) and haemagglutinin-esterase (HE), with S mediating receptor binding and membrane fusion, and HE acting as a receptor-destroying enzyme. Together, they promote dynamic virion attachment to glycan-based receptors, specifically 9-O-acetylated sialic acid. Here we present the cryo-EM structure of the ~80 kDa, heavily glycosylated HKU1 HE at 3.4 Å resolution. Comparison with existing HE structures reveals a drastically truncated lectin domain, incompatible with sialic acid binding, but with the structure and function of the esterase domain left intact. Cryo-EM and mass spectrometry analysis reveals a putative glycan shield on the now redundant lectin domain. The findings further our insight into the evolution and host adaptation of human embecoviruses, and demonstrate the utility of cryo-EM for studying small, heavily glycosylated proteins.


Assuntos
Betacoronavirus/química , Betacoronavirus/fisiologia , Infecções por Coronavirus/virologia , Hemaglutininas Virais/química , Proteínas Virais de Fusão/química , Betacoronavirus/classificação , Sítios de Ligação , Domínio Catalítico , Microscopia Crioeletrônica , Glicosilação , Células HEK293 , Hemaglutininas Virais/metabolismo , Hemaglutininas Virais/ultraestrutura , Humanos , Lectinas/química , Lectinas/metabolismo , Espectrometria de Massas , Modelos Moleculares , Ácido N-Acetilneuramínico/metabolismo , Polissacarídeos/química , Domínios Proteicos , Proteínas Virais de Fusão/metabolismo , Proteínas Virais de Fusão/ultraestrutura
4.
Nat Commun ; 11(1): 2688, 2020 05 27.
Artigo em Inglês | MEDLINE | ID: mdl-32461612

RESUMO

Severe acute respiratory syndrome (SARS) and Middle East respiratory syndrome (MERS) coronaviruses (CoVs) are zoonotic pathogens with high fatality rates and pandemic potential. Vaccine development focuses on the principal target of the neutralizing humoral immune response, the spike (S) glycoprotein. Coronavirus S proteins are extensively glycosylated, encoding around 66-87 N-linked glycosylation sites per trimeric spike. Here, we reveal a specific area of high glycan density on MERS S that results in the formation of oligomannose-type glycan clusters, which were absent on SARS and HKU1 CoVs. We provide a comparison of the global glycan density of coronavirus spikes with other viral proteins including HIV-1 envelope, Lassa virus glycoprotein complex, and influenza hemagglutinin, where glycosylation plays a known role in shielding immunogenic epitopes. Overall, our data reveal how organisation of glycosylation across class I viral fusion proteins influence not only individual glycan compositions but also the immunological pressure across the protein surface.


Assuntos
Glicoproteínas/imunologia , Coronavírus da Síndrome Respiratória do Oriente Médio , Polissacarídeos , Glicoproteína da Espícula de Coronavírus/imunologia , Proteínas Virais de Fusão/imunologia , Infecções por Coronavirus/imunologia , Infecções por Coronavirus/virologia , Microscopia Crioeletrônica , Epitopos/química , Epitopos/imunologia , Epitopos/metabolismo , Glicoproteínas/química , Glicoproteínas/ultraestrutura , Glicosilação , Células HEK293 , HIV-1/imunologia , HIV-1/metabolismo , Humanos , Evasão da Resposta Imune/fisiologia , Vírus Lassa/imunologia , Vírus Lassa/metabolismo , Coronavírus da Síndrome Respiratória do Oriente Médio/imunologia , Coronavírus da Síndrome Respiratória do Oriente Médio/metabolismo , Orthomyxoviridae/imunologia , Orthomyxoviridae/metabolismo , Polissacarídeos/química , Polissacarídeos/imunologia , Glicoproteína da Espícula de Coronavírus/química , Glicoproteína da Espícula de Coronavírus/ultraestrutura , Proteínas Virais de Fusão/química , Proteínas Virais de Fusão/ultraestrutura , Proteínas Virais/química , Proteínas Virais/imunologia , Proteínas Virais/ultraestrutura
5.
Nat Commun ; 9(1): 3050, 2018 08 03.
Artigo em Inglês | MEDLINE | ID: mdl-30076303

RESUMO

Understanding virus assembly mechanisms is important for developing therapeutic interventions. Nipah virus (NiV) is of interest because of its high mortality rate and efficient human-human transmissions. The current model for most enveloped viruses suggests that matrix proteins (M) recruit attachment glycoproteins (G) and fusion glycoproteins (F) to the assembly site at the plasma membrane. Here we report an assembly model that differs in many aspects from the current one. Examining NiV proteins on the cell plasma membrane using super-resolution microscopy reveals that clusters of F and G are randomly distributed on the plasma membrane regardless of the presence or absence of M. Our data suggests a model in which the M molecules assemble at the plasma membrane to form virus-like particles (VLPs), while the incorporation of F and G into the nascent VLPs is stochastic.


Assuntos
Microscopia/métodos , Vírus Nipah/fisiologia , Vírus Nipah/ultraestrutura , Montagem de Vírus , Animais , Linhagem Celular , Membrana Celular/virologia , Glicoproteínas/metabolismo , Células HeLa , Humanos , Transfecção , Proteínas do Envelope Viral/metabolismo , Proteínas Virais de Fusão/metabolismo , Proteínas Virais de Fusão/ultraestrutura , Proteínas da Matriz Viral/metabolismo , Proteínas da Matriz Viral/ultraestrutura , Vírion/metabolismo
6.
Nat Commun ; 9(1): 1736, 2018 04 30.
Artigo em Inglês | MEDLINE | ID: mdl-29712906

RESUMO

Measles virus (MeV) remains a major human pathogen, but there are presently no licensed antivirals to treat MeV or other paramyxoviruses. Here, we use cryo-electron tomography (cryo-ET) to elucidate the principles governing paramyxovirus assembly in MeV-infected human cells. The three-dimensional (3D) arrangement of the MeV structural proteins including the surface glycoproteins (F and H), matrix protein (M), and the ribonucleoprotein complex (RNP) are characterized at stages of virus assembly and budding, and in released virus particles. The M protein is observed as an organized two-dimensional (2D) paracrystalline array associated with the membrane. A two-layered F-M lattice is revealed suggesting that interactions between F and M may coordinate processes essential for MeV assembly. The RNP complex remains associated with and in close proximity to the M lattice. In this model, the M lattice facilitates the well-ordered incorporation and concentration of the surface glycoproteins and the RNP at sites of virus assembly.


Assuntos
Hemaglutininas Virais/ultraestrutura , Vírus do Sarampo/ultraestrutura , Ribonucleoproteínas/ultraestrutura , Proteínas Virais de Fusão/ultraestrutura , Proteínas da Matriz Viral/ultraestrutura , Vírion/ultraestrutura , Linhagem Celular , Microscopia Crioeletrônica , Fibroblastos/ultraestrutura , Fibroblastos/virologia , Células HeLa , Hemaglutininas Virais/metabolismo , Humanos , Vírus do Sarampo/metabolismo , Ribonucleoproteínas/metabolismo , Proteínas Virais de Fusão/metabolismo , Proteínas da Matriz Viral/metabolismo , Vírion/metabolismo , Montagem de Vírus , Liberação de Vírus
7.
Cell Host Microbe ; 23(1): 101-109.e4, 2018 Jan 10.
Artigo em Inglês | MEDLINE | ID: mdl-29324225

RESUMO

Since their first identification 50 years ago, marburgviruses have emerged several times, with 83%-90% lethality in the largest outbreaks. Although no vaccines or therapeutics are available for human use, the human antibody MR191 provides complete protection in non-human primates when delivered several days after inoculation of a lethal marburgvirus dose. The detailed neutralization mechanism of MR191 remains outstanding. Here we present a 3.2 Å crystal structure of MR191 complexed with a trimeric marburgvirus surface glycoprotein (GP). MR191 neutralizes by occupying the conserved receptor-binding site and competing with the host receptor Niemann-Pick C1. The structure illuminates previously disordered regions of GP including the stalk, fusion loop, CX6CC switch, and an N-terminal region of GP2 that wraps about the outside of GP1 to anchor a marburgvirus-specific "wing" antibody epitope. Virus escape mutations mapped far outside the MR191 receptor-binding site footprint suggest a role for these other regions in the GP quaternary structure.


Assuntos
Anticorpos Monoclonais/imunologia , Anticorpos Neutralizantes/imunologia , Anticorpos Antivirais/imunologia , Marburgvirus/imunologia , Receptores Virais/imunologia , Receptores Virais/ultraestrutura , Proteínas Virais de Fusão/imunologia , Proteínas Virais de Fusão/ultraestrutura , Agrobacterium tumefaciens , Animais , Anticorpos Monoclonais/ultraestrutura , Sítios de Ligação/imunologia , Proteínas de Transporte/imunologia , Linhagem Celular , Chlorocebus aethiops , Cristalografia por Raios X , Drosophila melanogaster , Humanos , Peptídeos e Proteínas de Sinalização Intracelular , Marburgvirus/metabolismo , Glicoproteínas de Membrana/imunologia , Proteína C1 de Niemann-Pick , Nicotiana , Células Vero , Proteínas do Envelope Viral/genética , Proteínas do Envelope Viral/imunologia , Ligação Viral
8.
Science ; 358(6363): 663-667, 2017 11 03.
Artigo em Inglês | MEDLINE | ID: mdl-29097548

RESUMO

The Rift Valley fever virus (RVFV) is transmitted by infected mosquitoes, causing severe disease in humans and livestock across Africa. We determined the x-ray structure of the RVFV class II fusion protein Gc in its postfusion form and in complex with a glycerophospholipid (GPL) bound in a conserved cavity next to the fusion loop. Site-directed mutagenesis and molecular dynamics simulations further revealed a built-in motif allowing en bloc insertion of the fusion loop into membranes, making few nonpolar side-chain interactions with the aliphatic moiety and multiple polar interactions with lipid head groups upon membrane restructuring. The GPL head-group recognition pocket is conserved in the fusion proteins of other arthropod-borne viruses, such as Zika and chikungunya viruses, which have recently caused major epidemics worldwide.


Assuntos
Membrana Celular/virologia , Glicerofosfolipídeos/química , Vírus da Febre do Vale do Rift/química , Proteínas Virais de Fusão/química , Sequência de Aminoácidos , Animais , Vírus Chikungunya/química , Vírus Chikungunya/ultraestrutura , Colesterol/química , Sequência Conservada , Cristalografia por Raios X , Humanos , Gado/virologia , Simulação de Dinâmica Molecular , Mutagênese Sítio-Dirigida , Conformação Proteica , Vírus da Febre do Vale do Rift/genética , Vírus da Febre do Vale do Rift/ultraestrutura , Proteínas Virais de Fusão/genética , Proteínas Virais de Fusão/ultraestrutura , Zika virus/química , Zika virus/ultraestrutura
9.
Microsc Microanal ; 23(1): 56-68, 2017 02.
Artigo em Inglês | MEDLINE | ID: mdl-28112080

RESUMO

The bulk of the major core protein VP7 in African horse sickness virus (AHSV) self-assembles into flat, hexagonal crystalline particles in a process appearing unrelated to viral replication. Why this unique characteristic of AHSV VP7 is genetically conserved, and whether VP7 aggregation and particle formation have an effect on cellular biology or the viral life cycle, is unknown. Here we investigated how different small peptide and enhanced green fluorescent protein (eGFP) insertions into the VP7 top domain affected VP7 localization, aggregation, and particle formation. This was done using a dual laser scanning confocal and transmission electron microscopy approach in conjunction with analyses of the solubility, aggregation, and fluorescence profiles of the proteins. VP7 top domain modifications did not prevent trimerization, or intracellular trafficking, to one or two discrete sites in the cell. However, modifications that resulted in a misfolded and insoluble VP7-eGFP component blocked trafficking, and precluded protein accumulation at a single cellular site, perhaps by interfering with normal trimer-trimer interactions. Furthermore, the modifications disrupted the stable layering of the trimers into characteristic AHSV VP7 crystalline particles. It was concluded that VP7 trafficking is driven by a balance between VP7 solubility, trimer forming ability, and trimer-trimer interactions.


Assuntos
Vírus da Doença Equina Africana/metabolismo , Microscopia Confocal/métodos , Microscopia Eletrônica de Transmissão/métodos , Proteínas do Core Viral/fisiologia , Proteínas do Core Viral/ultraestrutura , Vírus da Doença Equina Africana/genética , Animais , Baculoviridae/genética , Regulação Viral da Expressão Gênica , Vetores Genéticos , Proteínas de Fluorescência Verde , Estágios do Ciclo de Vida , Transporte Proteico , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/ultraestrutura , Células Sf9 , Proteínas do Core Viral/química , Proteínas do Core Viral/genética , Proteínas Virais de Fusão/fisiologia , Proteínas Virais de Fusão/ultraestrutura , Replicação Viral
10.
Nat Commun ; 6: 8176, 2015 Sep 14.
Artigo em Inglês | MEDLINE | ID: mdl-26365435

RESUMO

Human cytomegalovirus (HCMV) poses a significant threat to immunocompromised individuals and neonates infected in utero. Glycoprotein B (gB), the herpesvirus fusion protein, is a target for neutralizing antibodies and a vaccine candidate due to its indispensable role in infection. Here we show the crystal structure of the HCMV gB ectodomain bound to the Fab fragment of 1G2, a neutralizing human monoclonal antibody isolated from a seropositive subject. The gB/1G2 interaction is dominated by aromatic residues in the 1G2 heavy chain CDR3 protruding into a hydrophobic cleft in the gB antigenic domain 5 (AD-5). Structural analysis and comparison with HSV gB suggest the location of additional neutralizing antibody binding sites on HCMV gB. Finally, immunoprecipitation experiments reveal that 1G2 can bind to HCMV virion gB suggesting that its epitope is exposed and accessible on the virus surface. Our data will support the development of vaccines and therapeutic antibodies against HCMV infection.


Assuntos
Anticorpos Neutralizantes/metabolismo , Anticorpos Antivirais/metabolismo , Antígenos Virais/metabolismo , Fragmentos Fab das Imunoglobulinas/metabolismo , Proteínas do Envelope Viral/metabolismo , Proteínas Virais de Fusão/metabolismo , Anticorpos Neutralizantes/química , Anticorpos Neutralizantes/ultraestrutura , Anticorpos Antivirais/química , Anticorpos Antivirais/ultraestrutura , Antígenos Virais/química , Antígenos Virais/ultraestrutura , Cristalização , Cristalografia por Raios X , Citomegalovirus/imunologia , Humanos , Fragmentos Fab das Imunoglobulinas/química , Fragmentos Fab das Imunoglobulinas/ultraestrutura , Imunoprecipitação , Microscopia Eletrônica , Mutagênese Sítio-Dirigida , Conformação Proteica , Proteínas do Envelope Viral/química , Proteínas do Envelope Viral/ultraestrutura , Proteínas Virais de Fusão/química , Proteínas Virais de Fusão/ultraestrutura
11.
mBio ; 6(1): e02393-14, 2015 Feb 17.
Artigo em Inglês | MEDLINE | ID: mdl-25691596

RESUMO

UNLABELLED: In order to deliver their genetic material to host cells during infection, enveloped viruses use specialized proteins on their surfaces that bind cellular receptors and induce fusion of the viral and host membranes. In paramyxoviruses, a diverse family of single-stranded RNA (ssRNA) viruses, including several important respiratory pathogens, such as parainfluenza viruses, the attachment and fusion machinery is composed of two separate proteins: a receptor binding protein (hemagglutinin-neuraminidase [HN]) and a fusion (F) protein that interact to effect membrane fusion. Here we used negative-stain and cryo-electron tomography to image the 3-dimensional ultrastructure of human parainfluenza virus 3 (HPIV3) virions in the absence of receptor engagement. We observed that HN exists in at least two organizations. The first were arrays of tetrameric HN that lacked closely associated F proteins: in these purely HN arrays, HN adopted a "heads-down" configuration. In addition, we observed regions of complex surface density that contained HN in an apparently extended "heads-up" form, colocalized with prefusion F trimers. This colocalization with prefusion F prior to receptor engagement supports a model for fusion in which HN in its heads-up state and F may interact prior to receptor engagement without activating F, and that interaction with HN in this configuration is not sufficient to activate F. Only upon receptor engagement by HN's globular head does HN transmit its activating signal to F. IMPORTANCE: Human parainfluenza virus 3 (HPIV3) is an enveloped, ssRNA virus that can cause serious respiratory illness, especially in children. HPIV3, like most other paramyxoviruses, uses two specialized proteins to mediate cell entry: the fusion protein (F) and the receptor binding protein, hemagglutinin-neuraminidase (HN). F becomes activated to mediate fusion during entry when it is triggered by a signal from HN. Here we used electron tomography to reconstruct the 3-dimensional ultrastructure of HPIV3. From these structures, we could discern the distribution and, in some cases, conformation of HN and F proteins, which provided an understanding of their interrelationship on virions. HN is found in arrays alone in one conformation and interspersed with prefusion F trimers in another. The data support a model of paramyxovirus membrane fusion in which HN associates with F before receptor engagement, and receptor engagement by the globular head of HN switches the HN-F interaction into one of fusion activation.


Assuntos
Tomografia com Microscopia Eletrônica , Proteína HN/metabolismo , Proteína HN/ultraestrutura , Vírus da Parainfluenza 3 Humana/química , Vírus da Parainfluenza 3 Humana/ultraestrutura , Proteínas Virais de Fusão/metabolismo , Proteínas Virais de Fusão/ultraestrutura , Microscopia Crioeletrônica , Glicoproteínas de Membrana/metabolismo , Glicoproteínas de Membrana/ultraestrutura , Ligação Proteica , Receptores de Superfície Celular/metabolismo , Coloração e Rotulagem
12.
Proc Natl Acad Sci U S A ; 110(33): 13362-7, 2013 Aug 13.
Artigo em Inglês | MEDLINE | ID: mdl-23898184

RESUMO

The sequential steps in the alphavirus membrane fusion pathway have been postulated based on the prefusion and postfusion crystal structures of the viral fusion protein E1 in conjunction with biochemical studies. However, the molecular structures of the hypothesized fusion intermediates have remained obscure due to difficulties inherent in the dynamic nature of the process. We developed an experimental system that uses liposomes as the target membrane to capture Sindbis virus, a prototypical alphavirus, in its membrane-binding form at pH 6.4. Cryoelectron micrograph analyses and 3D reconstructions showed that the virus retains its overall icosahedral structure at this mildly acidic pH, except in the membrane-binding region, where monomeric E1 associates with the target membrane and the E2 glycoprotein retains its original trimeric organization. The remaining E2 trimers may hinder E1 homotrimerization and are a potential target for antiviral drugs.


Assuntos
Lipossomos/química , Modelos Moleculares , Sindbis virus/química , Sindbis virus/ultraestrutura , Proteínas Virais de Fusão/química , Microscopia Crioeletrônica/métodos , Concentração de Íons de Hidrogênio , Lipossomos/metabolismo , Lipossomos/ultraestrutura , Proteínas Virais de Fusão/metabolismo , Proteínas Virais de Fusão/ultraestrutura
13.
Proc Natl Acad Sci U S A ; 110(27): 11133-8, 2013 Jul 02.
Artigo em Inglês | MEDLINE | ID: mdl-23776214

RESUMO

Human respiratory syncytial virus is a human pathogen that causes severe infection of the respiratory tract. Current information about the structure of the virus and its interaction with host cells is limited. We carried out an electron cryotomographic characterization of cell culture-grown human respiratory syncytial virus to determine the architecture of the virion. The particles ranged from 100 nm to 1,000 nm in diameter and were spherical, filamentous, or a combination of the two. The filamentous morphology correlated with the presence of a cylindrical matrix protein layer linked to the inner leaflet of the viral envelope and with local ordering of the glycoprotein spikes. Recombinant viruses with only the fusion protein in their envelope showed that these glycoproteins were predominantly in the postfusion conformation, but some were also in the prefusion form. The ribonucleocapsids were left-handed, randomly oriented, and curved inside the virions. In filamentous particles, they were often adjacent to an intermediate layer of protein assigned to M2-1 (an envelope-associated protein known to mediate association of ribonucleocapsids with the matrix protein). Our results indicate important differences in structure between the Paramyxovirinae and Pneumovirinae subfamilies within the Paramyxoviridae, and provide fresh insights into host cell exit of a serious pathogen.


Assuntos
Vírus Sincicial Respiratório Humano/ultraestrutura , Linhagem Celular , Microscopia Crioeletrônica , Tomografia com Microscopia Eletrônica , Humanos , Conformação Proteica , Vírus Sincicial Respiratório Humano/química , Ribonucleoproteínas/química , Ribonucleoproteínas/ultraestrutura , Proteínas Virais de Fusão/química , Proteínas Virais de Fusão/ultraestrutura
14.
J Virol ; 86(21): 11457-71, 2012 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-22915804

RESUMO

The henipaviruses, Hendra virus (HeV) and Nipah virus (NiV), are paramyxoviruses discovered in the mid- to late 1990s that possess a broad host tropism and are known to cause severe and often fatal disease in both humans and animals. HeV and NiV infect cells by a pH-independent membrane fusion mechanism facilitated by their attachment (G) and fusion (F) glycoproteins. Here, several soluble forms of henipavirus F (sF) were engineered and characterized. Recombinant sF was produced by deleting the transmembrane (TM) and cytoplasmic tail (CT) domains and appending a glycosylphosphatidylinositol (GPI) anchor signal sequence followed by GPI-phospholipase D digestion, appending a trimeric coiled-coil (GCNt) domain (sF(GCNt)), or deleting the TM, CT, and fusion peptide domain. These sF glycoproteins were produced as F(0) precursors, and all were apparent stable trimers recognized by NiV-specific antisera. Surprisingly, however, only the GCNt-appended constructs (sF(GCNt)) could elicit cross-reactive henipavirus-neutralizing antibody in mice. In addition, sF(GCNt) constructs could be triggered in vitro by protease cleavage and heat to transition from an apparent prefusion to postfusion conformation, transitioning through an intermediate that could be captured by a peptide corresponding to the C-terminal heptad repeat domain of F. The pre- and postfusion structures of sF(GCNt) and non-GCNt-appended sF could be revealed by electron microscopy and were distinguishable by F-specific monoclonal antibodies. These data suggest that only certain sF constructs could serve as potential subunit vaccine immunogens against henipaviruses and also establish important tools for further structural, functional, and diagnostic studies on these important emerging viruses.


Assuntos
Henipavirus/imunologia , Henipavirus/ultraestrutura , Proteínas Virais de Fusão/imunologia , Proteínas Virais de Fusão/ultraestrutura , Animais , Anticorpos Monoclonais/imunologia , Anticorpos Neutralizantes/sangue , Anticorpos Antivirais/sangue , Reações Cruzadas , Células HeLa , Henipavirus/genética , Humanos , Camundongos , Microscopia Eletrônica , Conformação Proteica , Proteínas Recombinantes/genética , Proteínas Recombinantes/imunologia , Proteínas Recombinantes/metabolismo , Proteínas Recombinantes/ultraestrutura , Proteínas Virais de Fusão/genética , Proteínas Virais de Fusão/metabolismo
15.
Proc Natl Acad Sci U S A ; 109(8): 3089-94, 2012 Feb 21.
Artigo em Inglês | MEDLINE | ID: mdl-22323598

RESUMO

Human respiratory syncytial virus (hRSV) is the most important viral agent of pediatric respiratory infections worldwide. The only specific treatment available today is a humanized monoclonal antibody (Palivizumab) directed against the F glycoprotein, administered prophylactically to children at very high risk of severe hRSV infections. Palivizumab, as most anti-F antibodies so far described, recognizes an epitope that is shared by the two conformations in which hRSV_F can fold, the metastable prefusion form and the highly stable postfusion conformation. We now describe a unique class of antibodies specific for the prefusion form of this protein that account for most of the neutralizing activity of either a rabbit serum raised against a vaccinia virus recombinant expressing hRSV_F or a human Ig preparation (Respigam), which was used for prophylaxis before Palivizumab. These antibodies therefore offer unique possibilities for immune intervention against hRSV, and their production should be assessed in trials of hRSV vaccines.


Assuntos
Anticorpos Neutralizantes/imunologia , Anticorpos Antivirais/imunologia , Infecções por Vírus Respiratório Sincicial/terapia , Vírus Sincicial Respiratório Humano/imunologia , Proteínas Virais de Fusão/imunologia , Sequência de Aminoácidos , Animais , Humanos , Imunização , Dados de Sequência Molecular , Estabilidade Proteica , Coelhos , Proteínas Recombinantes/imunologia , Infecções por Vírus Respiratório Sincicial/imunologia , Infecções por Vírus Respiratório Sincicial/virologia , Vaccinia virus/imunologia , Proteínas Virais de Fusão/química , Proteínas Virais de Fusão/ultraestrutura
16.
Proc Natl Acad Sci U S A ; 108(23): 9619-24, 2011 Jun 07.
Artigo em Inglês | MEDLINE | ID: mdl-21586636

RESUMO

Respiratory syncytial virus (RSV), the main cause of infant bronchiolitis, remains a major unmet vaccine need despite more than 40 years of vaccine research. Vaccine candidates based on a chief RSV neutralization antigen, the fusion (F) glycoprotein, have foundered due to problems with stability, purity, reproducibility, and potency. Crystal structures of related parainfluenza F glycoproteins have revealed a large conformational change between the prefusion and postfusion states, suggesting that postfusion F antigens might not efficiently elicit neutralizing antibodies. We have generated a homogeneous, stable, and reproducible postfusion RSV F immunogen that elicits high titers of neutralizing antibodies in immunized animals. The 3.2-Å X-ray crystal structure of this substantially complete RSV F reveals important differences from homology-based structural models. Specifically, the RSV F crystal structure demonstrates the exposure of key neutralizing antibody binding sites on the surface of the postfusion RSV F trimer. This unanticipated structural feature explains the engineered RSV F antigen's efficiency as an immunogen. This work illustrates how structural-based antigen design can guide the rational optimization of candidate vaccine antigens.


Assuntos
Anticorpos Antivirais/imunologia , Infecções por Vírus Respiratório Sincicial/imunologia , Vírus Sinciciais Respiratórios/imunologia , Proteínas Virais de Fusão/imunologia , Sequência de Aminoácidos , Animais , Anticorpos Monoclonais/imunologia , Anticorpos Monoclonais/metabolismo , Anticorpos Monoclonais Humanizados , Anticorpos Neutralizantes/imunologia , Anticorpos Neutralizantes/metabolismo , Anticorpos Antivirais/metabolismo , Sítios de Ligação de Anticorpos , Dicroísmo Circular , Cristalografia por Raios X , Humanos , Imunização , Lactente , Camundongos , Camundongos Endogâmicos BALB C , Microscopia Eletrônica , Modelos Moleculares , Dados de Sequência Molecular , Palivizumab , Multimerização Proteica , Estrutura Secundária de Proteína , Estrutura Terciária de Proteína , Infecções por Vírus Respiratório Sincicial/virologia , Vírus Sinciciais Respiratórios/genética , Vírus Sinciciais Respiratórios/metabolismo , Homologia de Sequência de Aminoácidos , Sigmodontinae , Proteínas Virais de Fusão/química , Proteínas Virais de Fusão/ultraestrutura
17.
Virology ; 402(2): 372-9, 2010 Jul 05.
Artigo em Inglês | MEDLINE | ID: mdl-20439109

RESUMO

The paramyxovirus F protein is a class I viral membrane fusion protein which undergoes a significant refolding transition during virus entry. Previous studies of the Newcastle disease virus, human parainfluenza virus 3 and parainfluenza virus 5 F proteins revealed differences in the pre- and post-fusion structures. The NDV Queensland (Q) F structure lacked structural elements observed in the other two structures, which are key to the refolding and fusogenic activity of F. Here we present the NDV Australia-Victoria (AV) F protein post-fusion structure and provide EM evidence for its folding to a pre-fusion form. The NDV AV F structure contains heptad repeat elements missing in the previous NDV Q F structure, forming a post-fusion six-helix bundle (6HB) similar to the post-fusion hPIV3 F structure. Electrostatic and temperature factor analysis of the F structures points to regions of these proteins that may be functionally important in their membrane fusion activity.


Assuntos
Vírus da Doença de Newcastle/química , Dobramento de Proteína , Proteínas Virais de Fusão/química , Proteínas Virais de Fusão/metabolismo , Cristalografia por Raios X , Microscopia Eletrônica , Modelos Moleculares , Vírus da Doença de Newcastle/ultraestrutura , Conformação Proteica , Estrutura Terciária de Proteína , Proteínas Virais de Fusão/ultraestrutura
18.
Proc Natl Acad Sci U S A ; 107(14): 6292-7, 2010 Apr 06.
Artigo em Inglês | MEDLINE | ID: mdl-20332209

RESUMO

Bluetongue virus (BTV) is transmitted by blood-feeding insects (Culicoides sp.) and causes hemorrhagic diseases in livestock. BTV is a nonenveloped, double-stranded RNA (dsRNA) virus with two capsids: a well-studied, stable core enclosing the dsRNA genome and a highly unstable, poorly studied coat responsible for host cell attachment and entry. Here, based on cryo-electron microscopy (cryoEM), we report a 7-A resolution structure of the infectious BTV virion, including the coat proteins. We show that unlike other dsRNA viruses, the VP2 attachment trimer has a triskelion shape composed of three tip domains branching from a central hub domain. We identify three putative sialic acid-binding pockets in the hub and present supporting biochemical data indicating sugar moiety binding is important for BTV infection. Despite being a nonenveloped virus, the putative VP5 membrane penetration trimer, located slightly inward of the VP2 attachment trimer, has a central coiled-coil alpha-helical bundle, similar to the fusion proteins of many enveloped viruses (e.g., HIV, herpesviruses, vesicular stomatitis virus, and influenza virus). Moreover, mapping of the amino acid sequence of VP5 to the secondary structural elements identified by cryoEM locates 15 amphipathic alpha-helical regions on the external surface of each VP5 trimer. The cryoEM density map also reveals few, weak interactions between the VP5 trimer and both the outer-coat VP2 trimer and the underlying core VP7 trimer, suggesting that the surface of VP5 could unfurl like an umbrella during penetration and shedding of the coat to release the transcriptionally active core particle.


Assuntos
Vírus Bluetongue/química , Proteínas do Capsídeo/química , Ácido N-Acetilneuramínico/química , Multimerização Proteica , Proteínas Virais de Fusão/química , Sequência de Aminoácidos , Animais , Sítios de Ligação , Vírus Bluetongue/metabolismo , Proteínas do Capsídeo/metabolismo , Proteínas do Capsídeo/ultraestrutura , Linhagem Celular , Cricetinae , Microscopia Crioeletrônica , Humanos , Modelos Moleculares , Dados de Sequência Molecular , Ácido N-Acetilneuramínico/metabolismo , Estrutura Quaternária de Proteína , Estrutura Secundária de Proteína , Estrutura Terciária de Proteína , Proteínas Virais de Fusão/metabolismo , Proteínas Virais de Fusão/ultraestrutura , Replicação Viral
19.
Mol Gen Mikrobiol Virusol ; (2): 3-11, 2009.
Artigo em Russo | MEDLINE | ID: mdl-19522065

RESUMO

This review discusses the available data on molecular mechanism of penetration of baculoviruses into cell and structural features of proteins involved in this process. These data are discussed in terms of basic postulates of contemporary model of fusion of biological membranes and other large DNA-containing viruses.


Assuntos
Baculoviridae/fisiologia , Insetos/virologia , Internalização do Vírus , Animais , Membrana Celular/metabolismo , Membrana Celular/virologia , Herpesviridae/fisiologia , Poxviridae/fisiologia , Proteínas Virais de Fusão/metabolismo , Proteínas Virais de Fusão/ultraestrutura
20.
Cell Host Microbe ; 4(6): 600-8, 2008 Dec 11.
Artigo em Inglês | MEDLINE | ID: mdl-19064260

RESUMO

Alphaviruses infect cells via a low-pH-triggered membrane fusion reaction mediated by the class II virus fusion protein E1, an elongated molecule with three extramembrane domains (DI-III). E1 drives fusion by inserting its fusion peptide loop into the target membrane and refolding to a hairpin-like trimer in which DIII moves toward the target membrane and packs against the central trimer. Three-dimensional structures provide static pictures of prefusion and postfusion E1 but do not explain this transition. Using truncated forms of E1, we reconstituted a low-pH-dependent intermediate composed of trimers of DI/II. Unexpectedly, DI/II trimers were stable in the absence of DIII. Once formed at a low pH, DI/II trimers efficiently and specifically bound recombinant DIII through a pH-independent reaction. Even in the absence of DIII, DI/II trimers interacted to form hexagonal lattices and to cause membrane deformation and tubulation. These studies identify a prefusion intermediate in class II membrane fusion.


Assuntos
Alphavirus/fisiologia , Proteínas Virais de Fusão/metabolismo , Internalização do Vírus , Concentração de Íons de Hidrogênio , Substâncias Macromoleculares , Microscopia Eletrônica de Transmissão , Modelos Moleculares , Ligação Proteica , Proteínas Virais de Fusão/genética , Proteínas Virais de Fusão/ultraestrutura
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...