Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 71
Filtrar
Mais filtros











Intervalo de ano de publicação
1.
Exp Cell Res ; 378(2): 171-181, 2019 05 15.
Artigo em Inglês | MEDLINE | ID: mdl-30880029

RESUMO

Paget's disease (PD) features abnormal osteoclasts (OC) which sharply increase in number and size and then intensely induce bone resorption. The purpose of this study was to determine the direct effects of canine distemper virus (CDV) and its fusion protein and hemagglutinin protein (F + H) on receptor activator of nuclear factor kappa-B ligand (RANKL) induced OC formation in vitro. Immunofluorescence assay, OC morphological and functional detection, intracellular signaling pathway detection, Real-time PCR analysis and ELISA were applied in this study. Immunofluorescence assay provided the conclusive proof that CDV can infect and replicate in RAW264.7 mouse monocyte cell line, primary human peripheral blood mononuclear cells (PBMC) and their further fused OC. Both CDV and F + H significantly promoted OC formation and bone resorption ability induced by RANKL. Meanwhile, intracellular signaling transduction analysis revealed CDV and F + H specifically upregulated the phosphorylation of nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB) and mitogen-activated protein kinase (MAPK) induced by RANKL, respectively. Furthermore, without RANKL stimulation, both CDV and F + H slightly induced OC-like cells formation in RAW264.7 cell line even in the presence of NF-κB inhibitor. F + H upregulate OC differentiation and activity through modulation of NF-κB signaling pathway, and induce OC precursor cells merging dependent on the function of glycoproteins themselves. These results meant that F and H proteins play a pivotal role in CDV supporting OC formation. Moreover, this work further provide a new research direction that F and H proteins in CDV should be considered as a trigger during the pathogenesis of PD.


Assuntos
Vírus da Cinomose Canina/fisiologia , Hemaglutininas Virais/fisiologia , Osteoclastos , Proteínas Virais de Fusão/fisiologia , Animais , Diferenciação Celular/genética , Fusão Celular , Chlorocebus aethiops , Citocinas/metabolismo , Humanos , Camundongos , Proteínas Quinases Ativadas por Mitógeno/metabolismo , NF-kappa B/metabolismo , Osteoclastos/virologia , Ligante RANK/metabolismo , Células RAW 264.7 , Células Vero
2.
Microb Pathog ; 107: 81-87, 2017 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-28330747

RESUMO

Peste des petits ruminants virus (PPRV), belonging to paramyxoviruses, has six structure proteins (such as matrix protein (M), nucleocapsid proteins (N), fusion protein (F) and hemagglutinin protein (H)) and could cause high morbidity and mortality in sheep and goats. Although a vaccine strain of PPRV has been rescued and co-expression of M and N could yield PPRV-like particles, the roles of structure proteins in virion assembly and release have not been investigated in detail. In this study, plasmids carrying PPRV cDNA sequences encoding the N, M, H, and F proteins were expressed in Vero cells. The co-expression of all four proteins resulted in the release of virus-like particles (VLPs) with similar release efficiency to that of authentic virions. Moreover, the co-expression of M together with F also resulted in efficient VLPs release. In the absence of M protein, the expression of no combination of the other proteins resulted in particle release. In summary, a VLPs production system for PPRV has been established and M protein is necessary for promoting the assembly and release of VLPs, of which the predominant protein is M protein. Further study will be focused on the immunogenicity of the VLPs.


Assuntos
Vírus da Peste dos Pequenos Ruminantes/metabolismo , Vírus da Peste dos Pequenos Ruminantes/fisiologia , Células Vero/metabolismo , Proteínas da Matriz Viral/metabolismo , Animais , Anticorpos Antivirais , Chlorocebus aethiops/metabolismo , Chlorocebus aethiops/fisiologia , DNA Complementar , DNA Viral , Hemaglutininas Virais/metabolismo , Hemaglutininas Virais/fisiologia , Camundongos , Proteínas do Nucleocapsídeo/metabolismo , Proteínas do Nucleocapsídeo/fisiologia , Vírus da Peste dos Pequenos Ruminantes/genética , Vírus da Peste dos Pequenos Ruminantes/imunologia , Proteínas Virais de Fusão/metabolismo , Proteínas Virais de Fusão/fisiologia
3.
Uirusu ; 67(1): 3-16, 2017.
Artigo em Japonês | MEDLINE | ID: mdl-29593149

RESUMO

Measles virus (MeV) is exceptionally contagious and still a major cause of death in child.However, recently significant progress towards the elimination of measles has been made through increased vaccination coverage of measles-containing vaccines. The hemagglutinin (H) protein of MeV interacts with a cellular receptor, and this interaction is the first step of infection. MeV uses two different receptors, signaling lymphocyte activation molecule (SLAM) and nectin-4 expressed on immune cells and epithelial cells, respectively. The interactions of MeV with these receptors nicely explain the immune suppressive and high contagious properties of MeV. Binding of the H protein to a receptor triggers conformational changes in the fusion (F) protein, inducing fusion between viral and host plasma membranes for entry. The stalk region of the H protein plays a key role in the F protein-triggering. Recent studies of the H protein epitopes have revealed that the receptor binding site of the H protein constitutes a major neutralizing epitope. The interaction with two proteinaceous receptors probably imposes strong functional constraints on this epitope for amino acid changes. This would be a reason why measles vaccines, which are derived from MV strains isolated more than 60 years ago, are still highly effective against all MV strains currently circulating.


Assuntos
Vacina contra Sarampo , Vírus do Sarampo , Animais , Moléculas de Adesão Celular/metabolismo , Epitopos , Hemaglutininas Virais/química , Hemaglutininas Virais/metabolismo , Hemaglutininas Virais/fisiologia , Humanos , Vírus do Sarampo/patogenicidade , Ligação Proteica , Estrutura Secundária de Proteína , Receptores Virais/metabolismo , Proteína Associada à Molécula de Sinalização da Ativação Linfocitária/metabolismo , Proteínas Virais de Fusão/química , Internalização do Vírus
4.
J Virol ; 88(11): 6158-67, 2014 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-24648460

RESUMO

UNLABELLED: The measles virus (MeV) membrane fusion apparatus consists of a fusion protein trimer and an attachment protein tetramer. To trigger membrane fusion, the heads of the MeV attachment protein, hemagglutinin (H), bind cellular receptors while the 96-residue-long H stalk transmits the triggering signal. Structural and functional studies of the triggering mechanism of other paramyxoviruses suggest that receptor binding to their hemagglutinin-neuraminidase (HN) results in signal transmission through the central segments of their stalks. To gain insight into H-stalk structure and function, we individually replaced its residues with cysteine. We then assessed how stable the mutant proteins are, how efficiently they can be cross-linked by disulfide bonds, whether cross-linking results in loss of function, and, in this case, whether disulfide bond reduction restores function. While many residues in the central segment of the stalk and in the spacer segment above it can be efficiently cross-linked by engineered disulfide bonds, we report here that residues 59 to 79 cannot, suggesting that the 20 membrane-proximal residues are not engaged in a tetrameric structure. Rescue-of-function studies by disulfide bond reduction resulted in the redefinition and extension of the central fusion-activation segment as covering residues 84 to 117. In particular, we identified four residues located between positions 92 and 99, the function of which cannot be restored by disulfide bond reduction after cysteine mutagenesis. These mutant H proteins reached the cell surface as complex oligomers but could not trigger membrane fusion. We discuss these observations in the context of the stalk exposure model of membrane fusion triggering by paramyxoviruses. IMPORTANCE: Measles virus, while being targeted for eradication, still causes significant morbidity and mortality. Here, we seek to understand how it enters cells by membrane fusion. Two viral integral membrane glycoproteins (hemagglutinin tetramers and fusion protein trimers) mediate the concerted receptor recognition and membrane fusion processes. Since previous studies have suggested that the hemagglutinin stalk transmits the triggering signal to the fusion protein trimer, we completed an analysis of its structure and function by systematic Cys mutagenesis. We report that while certain residues of the central stalk segment confer specificity to the interaction with the fusion protein trimer, others are necessary to allow folding of the H-oligomer in a standard conformation conducive to fusion triggering, and still other residues sustain the conformational change that transmits the fusion-triggering signal.


Assuntos
Hemaglutininas Virais/metabolismo , Vírus do Sarampo/fisiologia , Fusão de Membrana/fisiologia , Modelos Biológicos , Transdução de Sinais/fisiologia , Animais , Chlorocebus aethiops , Cisteína , Dissulfetos/metabolismo , Citometria de Fluxo , Células HEK293 , Hemaglutininas Virais/fisiologia , Humanos , Mutagênese , Estabilidade Proteica , Células Vero
5.
Can J Microbiol ; 59(12): 814-24, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-24313454

RESUMO

In this study, we investigated the potential for different components of the measles virus (MV) to induce apoptosis of HeLa cells and explored the apoptotic molecular mechanisms. After testing the 2 envelope glycoproteins hemagglutinin (H) and fusion (F), we found that MV H alone was sufficient to induce the apoptosis of HeLa cells, whereas MV F did not. MV F also had no influence on MV-H-mediated apoptosis. MV H could induce cellular apoptosis in HeLa cells through its interaction with the cellular receptor CD46 via both the TRAIL-mediated extrinsic pathway and the mitochondria-controlled intrinsic pathway, and that cross talk between these 2 pathways occurred during the process. These findings extend the functions of MV envelope glycoproteins in the pathogenesis of MV infection and suggest that MV H may be a potential therapeutic in the treatment of some cancers.


Assuntos
Apoptose , Hemaglutininas Virais/fisiologia , Vírus do Sarampo/patogenicidade , Sarampo/virologia , Proteínas do Envelope Viral/fisiologia , Antígenos CD/imunologia , Antígenos CD/metabolismo , Antígenos CD/fisiologia , Células HeLa , Hemaglutininas Virais/genética , Humanos , Sarampo/patologia , Proteína Cofatora de Membrana/imunologia , Proteína Cofatora de Membrana/metabolismo , Redes e Vias Metabólicas , Proteínas do Envelope Viral/genética
6.
J Virol ; 84(20): 10913-7, 2010 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-20702637

RESUMO

Measles virus (MV) entry requires at least 2 viral proteins, the hemagglutinin (H) and fusion (F) proteins. We describe the rescue and characterization of a measles virus with a specific mutation in the stalk region of H (I98A) that is able to bind normally to cells but infects at a lower rate than the wild type due to a reduction in fusion triggering. The mutant H protein binds to F more avidly than the parent H protein does, and the corresponding virus is more sensitive to inhibition by fusion-inhibitory peptide. We show that after binding of MV to its receptor, H-F dissociation is required for productive infection.


Assuntos
Hemaglutininas Virais/genética , Vírus do Sarampo/genética , Vírus do Sarampo/patogenicidade , Mutação de Sentido Incorreto , Proteínas Virais de Fusão/genética , Substituição de Aminoácidos , Animais , Linhagem Celular , Chlorocebus aethiops , Células Gigantes/virologia , Hemaglutininas Virais/química , Hemaglutininas Virais/fisiologia , Humanos , Vírus do Sarampo/fisiologia , Proteínas Mutantes/química , Proteínas Mutantes/genética , Proteínas Mutantes/fisiologia , Células Vero , Proteínas Virais de Fusão/química , Proteínas Virais de Fusão/fisiologia , Internalização do Vírus
7.
J Gen Virol ; 88(Pt 11): 3100-3111, 2007 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-17947536

RESUMO

Infectious salmon anemia virus (ISAV) is classified in the genus Isavirus of the family Orthomyxoviridae. Although virulence variation of ISAV can be demonstrated experimentally in fish, virus strain identification is ambiguous because the correlates of pathogenicity and/or antigenicity of ISAV are not well defined. Thirteen ISAV isolates characterized for their ability to kill fish were used to search for markers of virulence on the virus surface glycoprotein genes; haemagglutinin-esterase (HE) and fusion (F) protein genes. A single amino acid change N(164)D in the putative globular head of the HE protein, and a deletion/insertion of

Assuntos
Hemaglutininas Virais/fisiologia , Isavirus/genética , Isavirus/patogenicidade , Proteínas Virais de Fusão/fisiologia , Motivos de Aminoácidos/genética , Motivos de Aminoácidos/fisiologia , Substituição de Aminoácidos/genética , Animais , Sequência de Bases , Linhagem Celular , Efeito Citopatogênico Viral/genética , Evolução Molecular , Doenças dos Peixes/mortalidade , Doenças dos Peixes/virologia , Geografia , Hemaglutininas Virais/genética , Isavirus/classificação , Dados de Sequência Molecular , Oncorhynchus kisutch , Oncorhynchus mykiss , Infecções por Orthomyxoviridae/mortalidade , Infecções por Orthomyxoviridae/veterinária , Infecções por Orthomyxoviridae/virologia , Filogenia , Salmo salar , Análise de Sequência de DNA , Deleção de Sequência , Homologia de Sequência , Proteínas Virais de Fusão/genética , Virulência
8.
J Virol ; 81(4): 1848-57, 2007 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-17151094

RESUMO

To understand the pathogenesis and develop an animal model of severe acute respiratory syndrome (SARS)-associated coronavirus (SARS-CoV), the Frankfurt 1 SARS-CoV isolate was passaged serially in young F344 rats. Young rats were susceptible to SARS-CoV but cleared the virus rapidly within 3 to 5 days of intranasal inoculation. After 10 serial passages, replication and virulence of SARS-CoV were increased in the respiratory tract of young rats without clinical signs. By contrast, adult rats infected with the passaged virus showed respiratory symptoms and severe pathological lesions in the lung. Levels of inflammatory cytokines in sera and lung tissues were significantly higher in adult F344 rats than in young rats. During in vivo passage of SARS-CoV, a single amino acid substitution was introduced within the binding domain of the viral spike protein recognizing angiotensin-converting enzyme 2 (ACE2), which is known as a SARS-CoV receptor. The rat-passaged virus more efficiently infected CHO cells expressing rat ACE2 than did the original isolate. These results strongly indicate that host and virus factors such as advanced age and virus adaptation are critical for the development of SARS in rats.


Assuntos
Modelos Animais de Doenças , Ratos Endogâmicos F344 , Síndrome Respiratória Aguda Grave/virologia , Coronavírus Relacionado à Síndrome Respiratória Aguda Grave/patogenicidade , Fatores Etários , Substituição de Aminoácidos , Enzima de Conversão de Angiotensina 2 , Animais , Chlorocebus aethiops , Citocinas/biossíntese , Feminino , Hemaglutininas Virais/fisiologia , Glicoproteínas de Membrana/química , Glicoproteínas de Membrana/fisiologia , Modelos Moleculares , Peptidil Dipeptidase A/química , Peptidil Dipeptidase A/metabolismo , Estrutura Terciária de Proteína/fisiologia , Ratos , Receptores Virais/metabolismo , Coronavírus Relacionado à Síndrome Respiratória Aguda Grave/química , Coronavírus Relacionado à Síndrome Respiratória Aguda Grave/genética , Inoculações Seriadas , Glicoproteína da Espícula de Coronavírus , Células Vero , Proteínas do Envelope Viral/química , Proteínas do Envelope Viral/fisiologia , Virulência
9.
Virology ; 352(2): 390-9, 2006 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-16781760

RESUMO

Natural infection and vaccination with a live-attenuated measles virus (MV) induce CD8(+) T-cell-mediated immune responses that may play a central role in controlling MV infection. In this study, we show that newly identified human HLA-A2 epitopes from MV hemagglutinin (H) and fusion (F) proteins induced protective immunity in HLA-A2 transgenic mice challenged with recombinant vaccinia viruses expressing F or H protein. HLA-A2 epitopes were predicted and synthesized. Five and four peptides from H and F, respectively, bound to HLA-A2 molecules in a T2-binding assay, and four from H and two from F could induce peptide-specific CD8+ T cell responses in HLA-A2 transgenic mice. Further experiments proved that three peptides from H (H9-567, H10-250, and H10-516) and one from F protein (F9-57) were endogenously processed and presented on HLA-A2 molecules. All peptides tested in this study are common to 5 different strains of MV including Edmonston. In both A2K(b) and HHD-2 mice, the identified peptide epitopes induced protective immunity against recombinant vaccinia viruses expressing H or F. Because F and H proteins induce neutralizing antibodies, they are major components of new vaccine strategies, and therefore data from this study will contribute to the development of new vaccines against MV infection.


Assuntos
Antígeno HLA-A2/imunologia , Hemaglutininas Virais/imunologia , Vírus do Sarampo/imunologia , Proteínas Virais de Fusão/imunologia , Sequência de Aminoácidos , Animais , Apresentação de Antígeno , Antígenos Virais/genética , Antígenos Virais/metabolismo , Sítios de Ligação/genética , Linfócitos T CD8-Positivos/imunologia , Epitopos/genética , Epitopos/imunologia , Antígeno HLA-A2/genética , Hemaglutininas Virais/fisiologia , Humanos , Vacina contra Sarampo/genética , Vacina contra Sarampo/imunologia , Vírus do Sarampo/patogenicidade , Vírus do Sarampo/fisiologia , Camundongos , Camundongos Transgênicos , Proteínas Recombinantes/genética , Proteínas Recombinantes/imunologia , Vacinas Sintéticas/genética , Vacinas Sintéticas/imunologia , Vaccinia virus/genética , Vaccinia virus/imunologia , Proteínas Virais de Fusão/fisiologia
10.
J Virol ; 80(12): 6106-14, 2006 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-16731949

RESUMO

The cytoplasmic tail of the murine leukemia virus (MuLV) envelope (Env) protein is known to play an important role in regulating viral fusion activity. Upon removal of the C-terminal 16 amino acids, designated as the R peptide, the fusion activity of the Env protein is activated. To extend our understanding of the inhibitory effect of the R peptide and investigate the specificity of inhibition, we constructed chimeric influenza virus-MuLV hemagglutinin (HA) genes. The influenza virus HA protein is the best-studied membrane fusion model, and we investigated the fusion activities of the chimeric HA proteins. We compared constructs in which the coding sequence for the cytoplasmic tail of the influenza virus HA protein was replaced by that of the wild-type or mutant MuLV Env protein or in which the cytoplasmic tail sequence of the MuLV Env protein was added to the HA cytoplasmic domain. Enzyme-linked immunosorbent assays and Western blot analysis showed that all chimeric HA proteins were effectively expressed on the cell surface and cleaved by trypsin. In BHK21 cells, the wild-type HA protein had a significant ability after trypsin cleavage to induce syncytium formation at pH 5.1; however, neither the chimeric HA protein with the full-length cytoplasmic tail of MuLV Env nor the full-length HA protein followed by the R peptide showed any syncytium formation. When the R peptide was truncated or mutated, the fusion activity was partially recovered in the chimeric HA proteins. A low-pH conformational-change assay showed that similar conformational changes occurred for the wild-type and chimeric HA proteins. All chimeric HA proteins were capable of promoting hemifusion and small fusion pore formation, as shown by a dye redistribution assay. These results indicate that the R peptide of the MuLV Env protein has a sequence-dependent inhibitory effect on influenza virus HA protein-induced membrane fusion and that the inhibitory effect occurs at a late stage in fusion pore enlargement.


Assuntos
Glicoproteínas de Hemaglutininação de Vírus da Influenza/fisiologia , Hemaglutininas Virais/fisiologia , Vírus da Leucemia Murina/química , Fusão de Membrana/efeitos dos fármacos , Oligopeptídeos/fisiologia , Orthomyxoviridae/química , Animais , Produtos do Gene env/fisiologia , Glicoproteínas de Hemaglutininação de Vírus da Influenza/análise , Glicoproteínas de Hemaglutininação de Vírus da Influenza/genética , Hemaglutininas Virais/análise , Hemaglutininas Virais/genética , Concentração de Íons de Hidrogênio , Camundongos , Oligopeptídeos/análise , Oligopeptídeos/genética , Conformação Proteica , Proteínas Recombinantes de Fusão/química , Proteínas Recombinantes de Fusão/farmacologia , Tripsina/metabolismo
11.
Virus Res ; 117(2): 273-82, 2006 May.
Artigo em Inglês | MEDLINE | ID: mdl-16343677

RESUMO

A specific interaction between the F and H proteins is required to enable fusion of the virus and host cell membranes and in some cases these proteins are not interchangeable between related viruses of the family Paramyxoviridae. For example, the F and H proteins of two ruminant morbilliviruses, rinderpest virus (RPV) and Peste-des-petits-ruminants virus (PPRV), are not interchangeable since viable virus could not be rescued from cDNA constructs where an individual glycoprotein gene of RPV was replaced with that from PPRV. To investigate which domain of the H protein, extracellular or cytoplasmic/transmembrane, was most important for preventing this interaction, two chimeric H gene constructs were made where the normal H gene of RPV was substituted with variant H genes where the transmembrane/cytoplasmic tail region (pRPV2C-PPRTm) or the whole ectodomain (pRPV2C-PPRExt) were derived from PPRV. Chimeric viruses were rescued from both the constructs and, while RPV2C-PPRTm virus grew to as high titres as the parent virus, RPV2C-PPRExt virus was extremely debilitated with respect to growth in tissue culture. Thus the ectodomain of H is the most important region required for effective interactions of the two glycoproteins for the recovery of viable virus. Nevertheless, the transmembrane/cytoplasmic domain of RPV alone can allow a chimeric virus to be rescued, which was not possible when the complete H gene was derived from PPRV. Both versions of the H protein and also the F protein were found to be incorporated into the envelope of the budded virions.


Assuntos
Hemaglutininas Virais/química , Hemaglutininas Virais/fisiologia , Vírus da Peste Bovina/fisiologia , Replicação Viral/genética , Animais , Chlorocebus aethiops , Citoplasma , DNA Complementar , Expressão Gênica , Glicoproteínas/metabolismo , Hemaglutininas Virais/genética , Proteínas de Membrana , Microscopia de Fluorescência , Microscopia Imunoeletrônica , Vírus da Peste dos Pequenos Ruminantes/genética , Vírus da Peste dos Pequenos Ruminantes/fisiologia , Estrutura Terciária de Proteína/genética , RNA Mensageiro/análise , RNA Viral/análise , Proteínas Recombinantes de Fusão/metabolismo , Vírus da Peste Bovina/genética , Células Vero , Proteínas Virais de Fusão/metabolismo , Ensaio de Placa Viral , Proteínas Virais/análise
12.
Virology ; 347(1): 11-27, 2006 Mar 30.
Artigo em Inglês | MEDLINE | ID: mdl-16375939

RESUMO

The fusion (F) protein of simian virus 5 strain W3A induces syncytium formation independently of coexpression of the hemagglutinin-neuraminidase protein. This property can be transferred to the F protein of strain WR by replacing the leucine at position 22 with the W3A F counterpart, proline. The resulting mutant L22P has a conformation that is distinct from that of the WR F protein. Se-L22P is a cleavage site mutant of L22P that is cleavable only by addition of exogenous trypsin. We showed here that the cell surface-localized L22P was internalized with a t1/2 of 25 min and degraded in the cell, while the WR F protein was not. The cell surface-localized Se-L22P underwent a significant conformational change upon cleavage. Intriguingly, it disappeared from the cell surface due to its internalization, while inducing extensive syncytium formation. These results indicate that L22P may display an internalization signal during the course of fusion induction.


Assuntos
Vírus da Parainfluenza 5/genética , Vírus da Parainfluenza 5/fisiologia , Proteínas Virais de Fusão/genética , Proteínas Virais de Fusão/fisiologia , Substituição de Aminoácidos , Transporte Biológico Ativo , Células Gigantes/virologia , Células HeLa , Hemaglutininas Virais/fisiologia , Humanos , Peso Molecular , Neuraminidase/fisiologia , Vírus da Parainfluenza 5/patogenicidade , Mutação Puntual , Conformação Proteica , Sinais Direcionadores de Proteínas/genética , Sinais Direcionadores de Proteínas/fisiologia , Proteínas Virais de Fusão/química
13.
J Virol ; 79(16): 10155-63, 2005 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-16051808

RESUMO

To engineer a targeting envelope for gene and oncolytic vector delivery, we characterized and modified the envelope proteins of Tupaia paramyxovirus (TPMV), a relative of the morbilli- and henipaviruses that neither infects humans nor has cross-reactive relatives that infect humans. We completed the TPMV genomic sequence and noted that the predicted fusion (F) protein cleavage-activation site is not preceded by a canonical furin cleavage sequence. Coexpression of the TPMV F and hemagglutinin (H) proteins induced fusion of Tupaia baby fibroblasts but not of human cells, a finding consistent with the restricted TPMV host range. To identify the factors restricting fusion of non-Tupaia cells, we initially analyzed F protein cleavage. Even without an oligo- or monobasic protease cleavage sequence, TPMV F was cleaved in F1 and F2 subunits in human cells. Edman degradation of the F1 subunit yielded the sequence IFWGAIIA, placing the conserved phenylalanine in position 2, a novelty for paramyxoviruses but not the cause of fusion restriction. We then verified whether the lack of a TPMV H receptor limits fusion. Toward this end, we displayed a single-chain antibody (scFv) specific for the designated receptor human carcinoembryonic antigen on the TPMV H ectodomain. The H-scFv hybrid protein coexpressed with TPMV F mediated fusion of cells expressing the designated receptor, proving that the lack of a receptor limits fusion and that TPMV H can be retargeted. Targeting competence and the absence of antibodies in humans define the TPMV envelope as a module to be adapted for ferrying ribonucleocapsids of oncolytic viruses and gene delivery vectors.


Assuntos
Vetores Genéticos/fisiologia , Hemaglutininas Virais/fisiologia , Paramyxoviridae/fisiologia , Tupaia/virologia , Proteínas Virais de Fusão/metabolismo , Sequência de Aminoácidos , Animais , Genoma Viral , Humanos , Fusão de Membrana , Dados de Sequência Molecular , Paramyxoviridae/genética , Receptores Virais/fisiologia , Tropismo
14.
J Gen Virol ; 86(Pt 8): 2269-2274, 2005 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-16033974

RESUMO

Severe acute respiratory syndrome coronavirus (SARS-CoV) contains a single spike (S) protein, which binds to its receptor, angiotensin-converting enzyme 2 (ACE2), induces membrane fusion and serves as a neutralizing antigen. A SARS-CoV-S protein-bearing vesicular stomatitis virus (VSV) pseudotype using the VSVDeltaG* system was generated. Partial deletion of the SARS-CoV-S protein cytoplasmic domain allowed efficient incorporation into VSV particles and led to the generation of a pseudotype (VSV-SARS-St19) at high titre. Green fluorescent protein expression was demonstrated as early as 7 h after infection of Vero E6 cells with VSV-SARS-St19. VSV-SARS-St19 was neutralized by anti-SARS-CoV antibody and soluble ACE2, and its infection was blocked by treatment of Vero E6 cells with anti-ACE2 antibody. These results indicated that VSV-SARS-St19 infection is mediated by SARS-CoV-S protein in an ACE2-dependent manner. VSV-SARS-St19 will be useful for analysing the function of SARS-CoV-S protein and for developing rapid methods of detecting neutralizing antibodies specific for SARS-CoV infection.


Assuntos
Hemaglutininas Virais/fisiologia , Glicoproteínas de Membrana/fisiologia , Vírus Reordenados/fisiologia , Infecções por Rhabdoviridae/virologia , Coronavírus Relacionado à Síndrome Respiratória Aguda Grave/fisiologia , Vírus da Estomatite Vesicular Indiana/fisiologia , Proteínas do Envelope Viral/fisiologia , Enzima de Conversão de Angiotensina 2 , Inibidores da Enzima Conversora de Angiotensina/farmacologia , Animais , Carboxipeptidases/antagonistas & inibidores , Carboxipeptidases/metabolismo , Chlorocebus aethiops , Relação Dose-Resposta a Droga , Hemaglutininas Virais/metabolismo , Humanos , Glicoproteínas de Membrana/metabolismo , Peptidil Dipeptidase A , Vírus Reordenados/metabolismo , Coronavírus Relacionado à Síndrome Respiratória Aguda Grave/química , Glicoproteína da Espícula de Coronavírus , Células Vero , Vírus da Estomatite Vesicular Indiana/metabolismo , Proteínas do Envelope Viral/metabolismo , Replicação Viral
15.
Uirusu ; 55(2): 207-19, 2005 Dec.
Artigo em Japonês | MEDLINE | ID: mdl-16557006

RESUMO

The majority of viral fusion proteins can be divided into two classes. The influenza hemagglutinin (HA) belongs to the class I fusion proteins and undergoes a series of conformational changes at acidic pH, leading to membrane fusion. The crystal structures of the prefusion and the postfusion forms of HA have been revealed in 1981 and 1994, respectively. On the basis of these structures, a model for the mechanism of membrane fusion mediated by the conformational changes of HA has been proposed. The flavivirus E and alphavirus E1 proteins belong to the class II fusion proteins and mediate membrane fusion at acidic pH. Their prefusion structures are distinct from that of HA. Last year, however, it has become evident that the postfusion structures of these class I and class II fusion proteins are similar. The paramyxovirus F protein belongs to the class I fusion proteins. In contrast to HA, an interaction between F and its homologous attachment protein is required for F to undergo the conformational changes. Since F mediates fusion at neutral pH, the infected cells can fuse with neighboring uninfected cells. The crystal structures of F and the attachment protein HN have recently been clarified, which will facilitate studies of the molecular mechanism of F-mediated membrane fusion.


Assuntos
Fusão de Membrana , Paramyxoviridae/fisiologia , Proteínas Virais de Fusão , Cristalografia por Raios X , Proteína-1 Reguladora de Fusão/fisiologia , Hemaglutininas Virais/química , Hemaglutininas Virais/fisiologia , Concentração de Íons de Hidrogênio , Microdomínios da Membrana/fisiologia , Conformação Proteica , Proteínas Virais de Fusão/química , Proteínas Virais de Fusão/classificação , Proteínas Virais de Fusão/fisiologia , Proteína rhoA de Ligação ao GTP/fisiologia
16.
J Biol Chem ; 280(8): 6933-41, 2005 Feb 25.
Artigo em Inglês | MEDLINE | ID: mdl-15507445

RESUMO

Many viruses achieve reversible attachment to sialic acid (Sia) by encoding envelope glycoproteins with receptor-binding and receptor-destroying activities. Toroviruses and group 2 coronaviruses bind to O-acetylated Sias, presumably via their spike proteins (S), whereas other glycoproteins, the hemagglutinin-esterases (HE), destroy Sia receptors by de-O-acetylation. Here, we present a comprehensive study of these enzymes. Sialate-9-O-acetylesterases specific for 5-N-acetyl-9-O-acetylneuraminic acid, described for bovine and human coronaviruses, also occur in equine coronaviruses and in porcine toroviruses. Bovine toroviruses, however, express novel sialate-9-O-acetylesterases, which prefer the di-O-acetylated substrate 5-N-acetyl-7(8),9-di-O-acetylneuraminic acid. Whereas most rodent coronaviruses express sialate-4-O-acetylesterases, the HE of murine coronavirus DVIM cleaves 9-O-acetylated Sias. Under the premise that HE specificity reflects receptor usage, we propose that two types of Sias serve as initial attachment factors for coronaviruses in mice. There are striking parallels between orthomyxo- and nidovirus biology. Reminiscent of antigenic shifts in orthomyxoviruses, rodent coronaviruses exchanged S and HE sequences through recombination to extents not appreciated before. As for orthomyxovirus reassortants, the fitness of nidovirus recombinant offspring probably depends both on antigenic properties and on compatibility of receptor-binding and receptor-destroying activities.


Assuntos
Acetiltransferases/fisiologia , Evolução Molecular , Nidovirales/enzimologia , Animais , Sequência de Bases , Coronaviridae/enzimologia , Coronaviridae/genética , Coronaviridae/patogenicidade , Hemaglutininas Virais/fisiologia , Humanos , Dados de Sequência Molecular , Nidovirales/genética , Nidovirales/patogenicidade , Receptores Virais/metabolismo , Especificidade da Espécie , Especificidade por Substrato , Torovirus/enzimologia , Torovirus/genética , Torovirus/patogenicidade , Proteínas Virais de Fusão/fisiologia
17.
Proc Natl Acad Sci U S A ; 99(23): 15124-9, 2002 Nov 12.
Artigo em Inglês | MEDLINE | ID: mdl-12417744

RESUMO

In the first stage of engineering a herpes simplex virus (HSV)-1 that specifically targets human malignant glioma cells, we constructed a recombinant virus designated R5111 in which we have ablated the binding sites for sulfated proteoglycans in glycoproteins B and C, replaced the amino-terminal 148 aa in glycoprotein C by IL-13 flanked at its amino terminus with a signal peptide, and inserted a second copy of IL-13 after the amino acid 24 of glycoprotein D. In the process, the binding site for HveA, a viral entry receptor, was disrupted. We have also transformed a cell line (J1.1) lacking HSV-1 receptors to express IL13Ralpha2 receptor (J13R cells). We report the following: the R5111 recombinant virus replicates as well as wild-type virus in a variety of cell lines including cell lines derived from brain tumors. R5111 failed to replicate in the parent J1.1 cell line but multiplied to titers similar to those obtained in other cell lines in the J13R cell line. On the basis of the evidence that R5111 can use the IL13Ralpha2 receptor for entry, we conclude that HSV-1 can use receptors other than HveA or nectins, provided it can bind to them. The domains of gD that interact with HveA and nectin receptors are independent of each other. Lastly, the fusogenic activities of the glycoproteins in the viral envelope are not dependent on a set of unique interactions between glycoprotein D and its receptor. The construction of R5111 opens the way for construction of viruses totally dependent on selected receptors for entry or imaging of targeted cells.


Assuntos
Herpesvirus Humano 1/fisiologia , Receptores de Interleucina/fisiologia , Receptores Virais/fisiologia , Proteínas do Envelope Viral/fisiologia , Sequência de Bases , Linhagem Celular , Primers do DNA , Genoma Viral , Hemaglutininas Virais/fisiologia , Herpesvirus Humano 1/genética , Humanos , Subunidade alfa1 de Receptor de Interleucina-13 , Dados de Sequência Molecular , Fases de Leitura Aberta , Receptores de Interleucina-13 , Recombinação Genética , Transfecção , Replicação Viral
18.
J Virol ; 76(14): 7174-86, 2002 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-12072517

RESUMO

The generation of replication-competent measles virus (MV) depends on the incorporation of biologically active, fusogenic glycoprotein complexes, which are required for attachment and penetration into susceptible host cells and for direct virus spread by cell-to-cell fusion. Whereas multiple studies have analyzed the importance of the ectodomains of the MV glycoproteins hemagglutinin (H) and fusion protein (F), we have investigated the role of the cytoplasmic tails of the F and H proteins for the formation of fusogenic complexes. Deletions in the cytoplasmic tails of transiently expressed MV glycoproteins were found to have varying effects on receptor binding, fusion, or fusion promotion activity. F tail truncation to only three amino acids did not affect fusion capacity. In contrast, truncation of the H cytoplasmic tail was limited. H protein mutants with cytoplasmic tails of <14 residues no longer supported F-mediated cell fusion, predominantly due to a decrease in surface expression and receptor binding. This indicates that a minimal length of the H protein tail of 14 amino acids is required to ensure a threshold local density to have sufficient accumulation of fusogenic H-F complexes. By using reverse genetics, a recombinant MV with an F tail of three amino acids (rMV-FcDelta30), as well as an MV with an H tail of 14 residues (rMV-HcDelta20), could be rescued, whereas generation of viruses with shorter H tails failed. Thus, glycoprotein truncation does not interfere with the successful generation of recombinant MV if fusion competence is maintained.


Assuntos
Hemaglutininas Virais/química , Vírus do Sarampo/patogenicidade , Fusão de Membrana , Recombinação Genética , Proteínas Virais de Fusão/química , Sequência de Aminoácidos , Animais , Fusão Celular , Linhagem Celular , Chlorocebus aethiops , Células Gigantes , Hemaglutininas Virais/genética , Hemaglutininas Virais/fisiologia , Humanos , Vírus do Sarampo/química , Vírus do Sarampo/genética , Dados de Sequência Molecular , Mutagênese Sítio-Dirigida , Células Vero , Proteínas Virais de Fusão/genética , Proteínas Virais de Fusão/fisiologia , Replicação Viral
19.
J Neurovirol ; 7(5): 391-9, 2001 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-11582511

RESUMO

Although CNS complications occurring early and late after acute measles are a serious problem and often fatal, the transient immunosuppression lasting for several weeks after the rash is the major cause of measles-related morbidity and mortality worldwide. This review is focused on the interactions of measles virus (MV) with cellular receptors on neural and lymphoid cells which are important elements in viral pathogenesis. First, the cognate MV receptors, CD46 and CD150, are important components of viral tropism by mediating binding and entry. Second, however, additional unknown cellular surface molecules may (independently of viral uptake) after interaction with the MV glycoprotein complex act as signaling molecules and thereby modulate cellular survival, proliferation, and specific functions.


Assuntos
Antígenos CD/fisiologia , Glicoproteínas/fisiologia , Imunoglobulinas/fisiologia , Linfócitos/virologia , Vírus do Sarampo/fisiologia , Glicoproteínas de Membrana/fisiologia , Neurônios/virologia , Receptores Virais/fisiologia , Antígenos de Superfície/fisiologia , Encéfalo/patologia , Encéfalo/virologia , Citocinas/fisiologia , Encefalite Viral/virologia , Hemaglutininas Virais/fisiologia , Humanos , Hospedeiro Imunocomprometido , Ativação Linfocitária , Linfócitos/imunologia , Linfócitos/metabolismo , Linfopenia/complicações , Linfopenia/imunologia , Linfopenia/virologia , Vírus do Sarampo/patogenicidade , Proteína Cofatora de Membrana , Fusão de Membrana/fisiologia , Modelos Biológicos , Neuroglia/virologia , Neurônios/metabolismo , Infecções Oportunistas/etiologia , Pneumonia Viral/virologia , Receptores de Superfície Celular , Fase de Repouso do Ciclo Celular , Transdução de Sinais , Membro 1 da Família de Moléculas de Sinalização da Ativação Linfocitária , Panencefalite Esclerosante Subaguda/imunologia , Panencefalite Esclerosante Subaguda/virologia , Subpopulações de Linfócitos T/imunologia , Subpopulações de Linfócitos T/patologia , Subpopulações de Linfócitos T/virologia , Proteínas Virais de Fusão/fisiologia , Proteínas da Matriz Viral/fisiologia , Viremia/virologia , Virulência , Replicação Viral
20.
Virology ; 285(1): 153-64, 2001 Jun 20.
Artigo em Inglês | MEDLINE | ID: mdl-11414815

RESUMO

The hemagglutinin-esterase (HE) glycoprotein of influenza C virus is composed of three domains: a stem domain active in membrane fusion (F), an acetylesterase domain (E), and a receptor-binding domain (R). The protein contains eight N-linked glycosylation sites, four (positions 26, 395, 552, and 603) in the F domain, three (positions 61, 131, and 144) in the E domain, and one (position 189) in the R domain. Here, we investigated the role of the individual oligosaccharide chains in antigenic properties, intracellular transport, and biological activities of the HE protein by eliminating each of the glycosylation sites by site-specific mutagenesis. Comparison of electrophoretic mobility between the wild-type and the mutant proteins showed that while seven of the glycosylation sites are used, one (position 131) is not. Analysis of reactivity of the mutants with anti-HE monoclonal antibodies demonstrated that glycosylation at position 144 is essential for the formation of conformation-dependent epitopes. It was also evident that glycosylation at the two sites in the F domain (positions 26 and 603), in addition to that in the E domain (position 144), is required for the HE molecule to be transported from the endoplasmic reticulum and that mutant HEs lacking one of these three sites failed to undergo the trimer assembly. Removal of an oligosaccharide chain at position 144 or 189 resulted in a decrease in the esterase activity. By contrast, two mutants lacking an oligosaccharide chain at position 26 or 603, which were defective not only in cell surface expression but in trimerization, possessed full-enzyme activity, suggesting that the HE monomers present within the cell have acetylesterase activity. Fusion activity of cells expressing each of mutant HEs was found to be comparable with the ability of the protein to be transported to the cell surface, suggesting that there is no specific oligosaccharide chain that plays a critical role in promoting membrane fusion.


Assuntos
Gammainfluenzavirus/fisiologia , Hemaglutininas Virais/fisiologia , Oligossacarídeos/metabolismo , Proteínas Virais de Fusão/fisiologia , Acetilesterase/metabolismo , Animais , Anticorpos Monoclonais/imunologia , Anticorpos Antivirais/imunologia , Sítios de Ligação , Células COS , Fusão Celular , Embrião de Galinha , Epitopos/imunologia , Glicosilação , Complexo de Golgi/metabolismo , Testes de Hemaglutinação , Hemaglutininas Virais/genética , Hemaglutininas Virais/metabolismo , Humanos , Gammainfluenzavirus/imunologia , Gammainfluenzavirus/metabolismo , Mutagênese Sítio-Dirigida , Conformação Proteica , Processamento de Proteína Pós-Traducional , Transfecção , Proteínas Virais de Fusão/genética , Proteínas Virais de Fusão/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA