Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Glycobiology ; 29(1): 2-21, 2019 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-29878112

RESUMEN

Glycosylation is a biologically important protein modification process by which a carbohydrate chain is enzymatically added to a protein at a specific amino acid residue. This process plays roles in many cellular functions, including intracellular trafficking, cell-cell signaling, protein folding and receptor binding. While glycosylation is a common host cell process, it is utilized by many pathogens as well. Protein glycosylation is widely employed by viruses for both host invasion and evasion of host immune responses. Thus better understanding of viral glycosylation functions has potential applications for improved antiviral therapeutic and vaccine development. Here, we summarize our current knowledge on the broad biological functions of glycans for the Mononegavirales, an order of enveloped negative-sense single-stranded RNA viruses of high medical importance that includes Ebola, rabies, measles and Nipah viruses. We discuss glycobiological findings by genera in alphabetical order within each of eight Mononegavirales families, namely, the bornaviruses, filoviruses, mymonaviruses, nyamiviruses, paramyxoviruses, pneumoviruses, rhabdoviruses and sunviruses.


Asunto(s)
Glicoproteínas/metabolismo , Mononegavirales/metabolismo , Polisacáridos/metabolismo , Proteínas Virales/metabolismo , Animales , Glicoproteínas/genética , Glicosilación , Humanos , Mononegavirales/genética , Polisacáridos/genética , Proteínas Virales/genética
2.
J Virol ; 88(8): 4338-52, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24478423

RESUMEN

UNLABELLED: Human metapneumovirus (hMPV) is a relatively recently identified paramyxovirus that causes acute upper and lower respiratory tract infection. Entry of hMPV is unusual among the paramyxoviruses, in that fusion is accomplished by the fusion (F) protein without the attachment glycoprotein (G protein). It has been suggested that hMPV F protein utilizes integrin αvß1 as a cellular receptor. Consistent with this, the F proteins of all known hMPV strains possess an integrin-binding motif ((329)RGD(331)). The role of this motif in viral entry, infectivity, and pathogenesis is poorly understood. Here, we show that α5ß1 and αv integrins are essential for cell-cell fusion and hMPV infection. Mutational analysis found that residues R329 and G330 in the (329)RGD(331) motif are essential for cell-cell fusion, whereas mutations at D331 did not significantly impact fusion activity. Furthermore, fusion-defective RGD mutations were either lethal to the virus or resulted in recombinant hMPVs that had defects in viral replication in cell culture. In cotton rats, recombinant hMPV with the R329K mutation in the F protein (rhMPV-R329K) and rhMPV-D331A exhibited significant defects in viral replication in nasal turbinates and lungs. Importantly, inoculation of cotton rats with these mutants triggered a high level of neutralizing antibodies and protected against hMPV challenge. Taken together, our data indicate that (i) α5ß1 and αv integrins are essential for cell-cell fusion and viral replication, (ii) the first two residues in the RGD motif are essential for fusion activity, and (iii) inhibition of the interaction of the integrin-RGD motif may serve as a new target to rationally attenuate hMPV for the development of live attenuated vaccines. IMPORTANCE: Human metapneumovirus (hMPV) is one of the major causative agents of acute respiratory disease in humans. Currently, there is no vaccine or antiviral drug for hMPV. hMPV enters host cells via a unique mechanism, in that viral fusion (F) protein mediates both attachment and fusion activity. Recently, it was suggested that hMPV F protein utilizes integrins as receptors for entry via a poorly understood mechanism. Here, we show that α5ß1 and αv integrins are essential for hMPV infectivity and F protein-mediated cell-cell fusion and that the integrin-binding motif in the F protein plays a crucial role in these functions. Our results also identify the integrin-binding motif to be a new, attenuating target for the development of a live vaccine for hMPV. These findings not only will facilitate the development of antiviral drugs targeting viral entry steps but also will lead to the development new live attenuated vaccine candidates for hMPV.


Asunto(s)
Integrina alfa5beta1/metabolismo , Integrina alfaV/metabolismo , Metapneumovirus/fisiología , Metapneumovirus/patogenicidad , Infecciones por Paramyxoviridae/metabolismo , Proteínas Virales de Fusión/química , Proteínas Virales de Fusión/metabolismo , Internalización del Virus , Secuencias de Aminoácidos , Animales , Femenino , Humanos , Integrina alfa5beta1/genética , Integrina alfaV/genética , Metapneumovirus/genética , Mutación Missense , Infecciones por Paramyxoviridae/genética , Infecciones por Paramyxoviridae/virología , Unión Proteica , Ratas , Sigmodontinae , Proteínas Virales de Fusión/genética , Virulencia
3.
PLoS Pathog ; 9(11): e1003770, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24278018

RESUMEN

Membrane fusion is essential for entry of the biomedically-important paramyxoviruses into their host cells (viral-cell fusion), and for syncytia formation (cell-cell fusion), often induced by paramyxoviral infections [e.g. those of the deadly Nipah virus (NiV)]. For most paramyxoviruses, membrane fusion requires two viral glycoproteins. Upon receptor binding, the attachment glycoprotein (HN/H/G) triggers the fusion glycoprotein (F) to undergo conformational changes that merge viral and/or cell membranes. However, a significant knowledge gap remains on how HN/H/G couples cell receptor binding to F-triggering. Via interdisciplinary approaches we report the first comprehensive mechanism of NiV membrane fusion triggering, involving three spatiotemporally sequential cell receptor-induced conformational steps in NiV-G: two in the head and one in the stalk. Interestingly, a headless NiV-G mutant was able to trigger NiV-F, and the two head conformational steps were required for the exposure of the stalk domain. Moreover, the headless NiV-G prematurely triggered NiV-F on virions, indicating that the NiV-G head prevents premature triggering of NiV-F on virions by concealing a F-triggering stalk domain until the correct time and place: receptor-binding. Based on these and recent paramyxovirus findings, we present a comprehensive and fundamentally conserved mechanistic model of paramyxovirus membrane fusion triggering and cell entry.


Asunto(s)
Glicoproteínas/metabolismo , Proteínas de la Fusión de la Membrana/metabolismo , Virus Nipah/fisiología , Receptores Virales/metabolismo , Proteínas Virales/metabolismo , Internalización del Virus , Animales , Células CHO , Cricetinae , Cricetulus , Glicoproteínas/genética , Infecciones por Henipavirus/genética , Infecciones por Henipavirus/metabolismo , Proteínas de la Fusión de la Membrana/genética , Receptores Virales/genética , Proteínas Virales/genética
4.
J Virol ; 87(15): 8813-5, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23740987

RESUMEN

Newcastle disease virus (NDV)-induced membrane fusion requires formation of a complex between the hemagglutinin-neuraminidase (HN) and fusion (F) proteins. Substitutions for NDV HN stalk residues A89, L90, and L94 block fusion by modulating formation of the HN-F complex. Here, we demonstrate that a nearby L97A substitution, though previously shown to block fusion, allows efficient HN-F complex formation and likely acts by preventing changes in the HN stalk required for triggering of the bound F protein.


Asunto(s)
Proteína HN/metabolismo , Virus de la Enfermedad de Newcastle/fisiología , Multimerización de Proteína , Proteínas Virales de Fusión/metabolismo , Internalización del Virus , Sustitución de Aminoácidos , Proteína HN/genética , Proteínas Mutantes/genética , Proteínas Mutantes/metabolismo , Virus de la Enfermedad de Newcastle/genética , Unión Proteica
5.
J Virol ; 87(6): 3119-29, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-23283956

RESUMEN

The promotion of membrane fusion by most paramyxoviruses requires an interaction between the viral attachment and fusion (F) proteins to enable receptor binding by the former to trigger the activation of the latter for fusion. Numerous studies demonstrate that the F-interactive sites on the Newcastle disease virus (NDV) hemagglutinin-neuraminidase (HN) and measles virus (MV) hemagglutinin (H) proteins reside entirely within the stalk regions of those proteins. Indeed, stalk residues of NDV HN and MV H that likely mediate the F interaction have been identified. However, despite extensive efforts, the F-interactive site(s) on the Nipah virus (NiV) G attachment glycoprotein has not been identified. In this study, we have introduced individual N-linked glycosylation sites at several positions spaced at intervals along the stalk of the NiV G protein. Five of the seven introduced sites are utilized as established by a retardation of electrophoretic mobility. Despite surface expression, ephrinB2 binding, and oligomerization comparable to those of the wild-type protein, four of the five added N-glycans completely eliminate the ability of the G protein to complement the homologous F protein in the promotion of fusion. The most membrane-proximal added N-glycan reduces fusion by 80%. However, unlike similar NDV HN and MV H mutants, the NiV G glycosylation stalk mutants retain the ability to bind F, indicating that the fusion deficiency of these mutants is not due to prevention of the G-F interaction. These findings suggest that the G-F interaction is not mediated entirely by the stalk domain of G and may be more complex than that of HN/H-F.


Asunto(s)
Virus Nipah/fisiología , Polisacáridos/metabolismo , Proteínas del Envoltorio Viral/química , Proteínas del Envoltorio Viral/metabolismo , Internalización del Virus , Ensayo de Cambio de Movilidad Electroforética , Virus Nipah/química , Unión Proteica , Mapeo de Interacción de Proteínas
6.
J Virol ; 86(21): 11800-14, 2012 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-22915815

RESUMEN

The genus Metapneumovirus within the subfamily Pneumovirinae of the family Paramyxoviridae includes two members, human metapneumovirus (hMPV) and avian metapneumovirus (aMPV), causing respiratory tract infections in humans and birds, respectively. Paramyxoviruses enter host cells by fusing the viral envelope with a host cell membrane. Membrane fusion of hMPV appears to be unique, in that fusion of some hMPV strains requires low pH. Here, we show that the fusion (F) proteins of aMPV promote fusion in the absence of the attachment protein and low pH is not required. Furthermore, there are notable differences in cell-cell fusion among aMPV subtypes. Trypsin was required for cell-cell fusion induced by subtype B but not subtypes A and C. The F protein of aMPV subtype A was highly fusogenic, whereas those from subtypes B and C were not. By construction and evaluation of chimeric F proteins composed of domains from the F proteins of subtypes A and B, we localized a region composed of amino acid residues 170 to 338 in the F protein that is responsible for the hyperfusogenic phenotype of the F from subtype A. Further mutagenesis analysis revealed that residues R295, G297, and K323 in this region collectively contributed to the hyperfusogenicity. Taken together, we have identified a region in the aMPV F protein that modulates the extent of membrane fusion. A model for fusion consistent with these data is presented.


Asunto(s)
Fusión Celular , Metapneumovirus/patogenicidad , Proteínas Virales de Fusión/metabolismo , Animales , Línea Celular , Chlorocebus aethiops , Cricetinae , Análisis Mutacional de ADN , Genotipo , Datos de Secuencia Molecular , ARN Viral/genética , Recombinación Genética , Análisis de Secuencia de ADN , Proteínas Virales de Fusión/genética
7.
Curr Top Microbiol Immunol ; 359: 79-94, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22427111

RESUMEN

Nipah (NiV) and Hendra (HeV) viruses cause cell-cell fusion (syncytia) in brain, lung, heart, and kidney tissues, leading to encephalitis, pneumonia, and often death. Membrane fusion is essential to both viral entry and virus-induced cell-cell fusion, a hallmark of henipavirus infections. Elucidiation of the mechanism(s) of membrane fusion is critical to understanding henipavirus pathobiology and has the potential to identify novel strategies for the development of antiviral therapeutic agents. Henipavirus membrane fusion requires the coordinated actions of the viral attachment (G) and fusion (F) glycoproteins. Current henipavirus fusion models posit that attachment of NiV or HeV G to its cell surface receptors releases F from its metastable pre-fusion conformation to mediate membrane fusion. The identification of ephrinB2 and ephrinB3 as henipavirus receptors has paved the way for recent advances in our understanding of henipavirus membrane fusion. These advances highlight mechanistic similarities and differences between membrane fusion for the henipavirus and other genera within the Paramyxoviridae family. Here, we review these mechanisms and the current gaps in our knowledge in the field.


Asunto(s)
Efrina-B2/metabolismo , Efrina-B3/metabolismo , Virus Hendra/fisiología , Virus Nipah/fisiología , Receptores Virales/metabolismo , Proteínas Virales de Fusión/metabolismo , Animales , Encéfalo/patología , Encéfalo/virología , Efrina-B2/química , Efrina-B3/química , Virus Hendra/patogenicidad , Infecciones por Henipavirus/patología , Infecciones por Henipavirus/virología , Humanos , Riñón/patología , Riñón/virología , Pulmón/patología , Pulmón/virología , Fusión de Membrana , Virus Nipah/patogenicidad , Receptores Virales/química , Proteínas Virales de Fusión/química , Internalización del Virus
8.
J Virol ; 85(22): 12079-82, 2011 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-21880745

RESUMEN

Newcastle disease virus (NDV)-induced membrane fusion requires an interaction between the hemagglutinin-neuraminidase (HN) attachment and the fusion (F) proteins, triggered by HN's binding to receptors. NDV HN has two sialic acid binding sites: site I, which also mediates neuraminidase activity, and site II, which straddles the membrane-distal end of the dimer interface. By characterizing the effect on receptor binding avidity and F-interactive capability of HN dimer interface mutations, we present evidence consistent with (i) receptor engagement by site I triggering the interaction with F and (ii) site II functioning to maintain high-avidity receptor binding during the fusion process.


Asunto(s)
Proteína HN/metabolismo , Ácido N-Acetilneuramínico/metabolismo , Virus de la Enfermedad de Newcastle/fisiología , Proteínas Virales de Fusión/metabolismo , Internalización del Virus , Sitios de Unión , Unión Proteica
9.
PLoS Pathog ; 7(6): e1002058, 2011 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-21655106

RESUMEN

Measles virus (MeV), a member of the paramyxovirus family of enveloped RNA viruses and one of the most infectious viral pathogens identified, accounts for major pediatric morbidity and mortality worldwide although coordinated efforts to achieve global measles control are in place. Target cell entry is mediated by two viral envelope glycoproteins, the attachment (H) and fusion (F) proteins, which form a complex that achieves merger of the envelope with target cell membranes. Despite continually expanding knowledge of the entry strategies employed by enveloped viruses, our molecular insight into the organization of functional paramyxovirus fusion complexes and the mechanisms by which the receptor binding by the attachment protein triggers the required conformational rearrangements of the fusion protein remain incomplete. Recently reported crystal structures of the MeV attachment protein in complex with its cellular receptors CD46 or SLAM and newly developed functional assays have now illuminated some of the fundamental principles that govern cell entry by this archetype member of the paramyxovirus family. Here, we review these advances in our molecular understanding of MeV entry in the context of diverse entry strategies employed by other members of the paramyxovirus family.


Asunto(s)
Sarampión/virología , Paramyxovirinae/fisiología , Proteínas Virales de Fusión/metabolismo , Internalización del Virus , Membrana Celular/virología , Niño , Humanos , Virus del Sarampión/fisiología , Fusión de Membrana/fisiología , Receptores Virales/metabolismo , Acoplamiento Viral
10.
J Biol Chem ; 286(20): 17851-60, 2011 May 20.
Artículo en Inglés | MEDLINE | ID: mdl-21460213

RESUMEN

The fusion (F) proteins of Newcastle disease virus (NDV) and Nipah virus (NiV) are both triggered by binding to receptors, mediated in both viruses by a second protein, the attachment protein. However, the hemagglutinin-neuraminidase (HN) attachment protein of NDV recognizes sialic acid receptors, whereas the NiV G attachment protein recognizes ephrinB2/B3 as receptors. Chimeric proteins composed of domains from the two attachment proteins have been evaluated for fusion-promoting activity with each F protein. Chimeras having NiV G-derived globular domains and NDV HN-derived stalks, transmembranes, and cytoplasmic tails are efficiently expressed, bind ephrinB2, and trigger NDV F to promote fusion in Vero cells. Thus, the NDV F protein can be triggered by binding to the NiV receptor, indicating that an aspect of the triggering cascade induced by the binding of HN to sialic acid is conserved in the binding of NiV G to ephrinB2. However, the fusion cascade for triggering NiV F by the G protein and that of triggering NDV F by the chimeras can be distinguished by differential exposure of a receptor-induced conformational epitope. The enhanced exposure of this epitope marks the triggering of NiV F by NiV G but not the triggering of NDV F by the chimeras. Thus, the triggering cascade for NiV G-F fusion may be more complex than that of NDV HN and F. This is consistent with the finding that reciprocal chimeras having NDV HN-derived heads and NiV G-derived stalks, transmembranes, and tails do not trigger either F protein for fusion, despite efficient cell surface expression and receptor binding.


Asunto(s)
Virus de la Enfermedad de Newcastle/metabolismo , Virus Nipah/metabolismo , Proteínas Recombinantes de Fusión/metabolismo , Proteínas del Envoltorio Viral/metabolismo , Animales , Chlorocebus aethiops , Efrina-B2/genética , Efrina-B2/metabolismo , Cobayas , Virus de la Enfermedad de Newcastle/genética , Virus Nipah/genética , Unión Proteica , Proteínas Recombinantes de Fusión/genética , Células Vero , Proteínas del Envoltorio Viral/genética
11.
Virus Res ; 147(1): 153-7, 2010 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-19896994

RESUMEN

Newcastle disease virus (NDV) is an avian paramyxovirus that exists as hundreds of strains with widely different virulence properties. The NDV V protein exhibits interferon (IFN) antagonistic activity, which contributes to the virulence of the virus. The IFN-antagonistic activities of the V proteins from the avirulent strain La Sota and the moderately virulent strain Beaudette C (BC) were compared in an assay for the rescue of a recombinant NDV expressing the green fluorescent protein (NDV-GFP). Consistent with the virulence properties of the two viruses, the BC V protein exhibits a 4-fold greater ability to rescue replication of NDV-GFP than the La Sota V protein. Four amino acid differences in the C-terminal region of V, as well as the N-terminal region, contribute to the difference in IFN-antagonistic activity between the two V proteins.


Asunto(s)
Interferones/antagonistas & inhibidores , Virus de la Enfermedad de Newcastle/inmunología , Virus de la Enfermedad de Newcastle/patogenicidad , Proteínas Virales/fisiología , Factores de Virulencia/fisiología , Sustitución de Aminoácidos , Animales , Línea Celular , Pollos
12.
Virology ; 383(1): 1-5, 2009 Jan 05.
Artículo en Inglés | MEDLINE | ID: mdl-19013625

RESUMEN

Fusion promotion by measles virus (MV) depends on an interaction between the hemagglutinin (H) and fusion (F) glycoproteins. Amino acid substitutions in MV H that drastically reduce hemagglutinating activity result in an increase in the amount of H (primarily the 74 kDa isoform) detectable in a complex with F at the cell surface. This is in direct contrast to the loss of the ability to detect a complex between the fusion protein of Newcastle disease virus and most attachment proteins that lack receptor binding activity. These opposing results provide support for the existence of different mechanisms for the regulation of fusion by these two paramyxoviruses.


Asunto(s)
Hemaglutininas Virales/genética , Hemaglutininas Virales/metabolismo , Virus del Sarampión/fisiología , Mutación Missense , Proteínas Virales de Fusión/metabolismo , Acoplamiento Viral , Sustitución de Aminoácidos/genética , Proteína Cofactora de Membrana/metabolismo , Mutagénesis Sitio-Dirigida , Proteínas Mutantes/genética , Proteínas Mutantes/metabolismo , Receptores Virales/metabolismo
13.
Future Virol ; 4(4): 335-351, 2009 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-20161127

RESUMEN

The Paramyxoviridae are enveloped, negative-stranded RNA viruses, some of which recognize sialic acid-containing receptors, while others recognize specific proteinaceous receptors. The major cytopathic effect of paramyxovirus infection is membrane fusion-induced syncytium formation. Paramyxoviruses are unusual in that the receptor-binding and fusion-promoting activities reside on two different spike structures, the attachment and fusion glycoproteins, respectively. For most paramyxoviruses, this distribution of functions requires a mechanism by which the two processes can be linked for the promotion of fusion. This is accomplished by a virus-specific interaction between the two proteins. An increasing body of evidence supports the notion that members of this family of viruses utilize this glycoprotein interaction in different ways in order to mediate the regulation of the fusion protein activation, depending on the type of receptor utilized by the virus.

14.
J Virol ; 82(21): 10386-96, 2008 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-18753211

RESUMEN

The promotion of membrane fusion by Newcastle disease virus (NDV) requires an interaction between the viral hemagglutinin-neuraminidase (HN) and fusion (F) proteins, although the mechanism by which this interaction regulates fusion is not clear. The NDV HN protein exists as a tetramer composed of a pair of dimers. Based on X-ray crystallographic studies of the NDV HN globular domain (S. Crennell et al., Nat. Struct. Biol. 7:1068-1074, 2000), it was proposed that the protein undergoes a significant conformational change from an initial structure having minimal intermonomeric contacts to a structure with a much more extensive dimer interface. This conformational change was predicted to be integral to fusion promotion with the minimal interface form required to maintain F in its prefusion state until HN binds receptors. However, no evidence for such a conformational change exists for any other paramyxovirus attachment protein. To test the NDV model, we have engineered a pair of intermonomeric disulfide bonds across the dimer interface in the globular domain of an otherwise non-disulfide-linked NDV HN protein by the introduction of cysteine substitutions for residues T216 and D230. The disulfide-linked dimer is formed both intracellularly and in the absence of receptor binding and is efficiently expressed at the cell surface. The disulfide bonds preclude formation of the minimal interface form of the protein and yet enhance both receptor-binding activity at 37 degrees C and fusion promotion. These results confirm that neither the minimal interface form of HN nor the proposed drastic conformational change in the protein is required for fusion.


Asunto(s)
Proteína HN/metabolismo , Virus de la Enfermedad de Newcastle/fisiología , Proteínas Virales/metabolismo , Internalización del Virus , Sustitución de Aminoácidos/genética , Animales , Línea Celular , Cricetinae , Cisteína/genética , Cisteína/metabolismo , Disulfuros/metabolismo , Proteína HN/genética , Modelos Moleculares , Mutagénesis Sitio-Dirigida , Proteínas Virales/genética , Acoplamiento Viral
15.
Trends Microbiol ; 16(4): 135-7, 2008 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-18346895

RESUMEN

Paramyxovirus-mediated membrane fusion usually requires an interaction between the viral-attachment and -fusion proteins. The mechanism by which this interaction regulates fusion differs between paramyxoviruses that bind to sialic acid-containing receptors and those that recognize specific proteins. The recently solved structure of the globular head of the measles virus hemagglutinin suggests that this difference might be related to the location of the receptor-binding sites on the attachment proteins of the two classes of paramyxoviruses.


Asunto(s)
Sitios de Unión/genética , Virus del Sarampión/patogenicidad , Fusión de Membrana , Virus de la Enfermedad de Newcastle/patogenicidad , Receptores Virales/metabolismo , Animales , Proteína HN/química , Proteína HN/metabolismo , Hemaglutininas Virales/química , Hemaglutininas Virales/metabolismo , Humanos , Virus del Sarampión/metabolismo , Virus de la Enfermedad de Newcastle/metabolismo , Receptores Virales/química , Proteínas Virales/química , Proteínas Virales/metabolismo
16.
J Virol ; 81(18): 9900-10, 2007 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-17626104

RESUMEN

The hemagglutinin (H) protein of measles virus (MV) mediates attachment to cellular receptors. The ectodomain of the H spike is thought to consist of a membrane-proximal stalk and terminal globular head, in which resides the receptor-binding activity. Like other paramyxovirus attachment proteins, MV H also plays a role in fusion promotion, which is mediated through an interaction with the viral fusion (F) protein. The stalk of the hemagglutinin-neuraminidase (HN) protein of several paramyxoviruses determines specificity for the homologous F protein. In addition, mutations in a conserved domain in the Newcastle disease virus (NDV) HN stalk result in a sharp decrease in fusion and an impaired ability to interact with NDV F in a cell surface coimmunoprecipitation (co-IP) assay. The region of MV H that determines specificity for the F protein has not been identified. Here, we have adapted the co-IP assay to detect the MV H-F complex at the surface of transfected HeLa cells. We have also identified mutations in a domain in the MV H stalk, similar to the one in the NDV HN stalk, that also drastically reduce fusion yet do not block complex formation with MV F. These results indicate that this domain in the MV H stalk is required for fusion but suggest either that mutation of it indirectly affects the H-dependent activation of F or that the MV H-F interaction is mediated by more than one domain in H. This points to an apparent difference in the way the MV and NDV glycoproteins interact to regulate fusion.


Asunto(s)
Hemaglutininas Virales/metabolismo , Virus del Sarampión/metabolismo , Mutación , Virus de la Enfermedad de Newcastle/metabolismo , Proteínas Virales de Fusión/metabolismo , Internalización del Virus , Células HeLa , Hemaglutininas Virales/genética , Humanos , Virus del Sarampión/genética , Virus de la Enfermedad de Newcastle/genética , Unión Proteica/genética , Estructura Terciaria de Proteína/genética , Homología de Secuencia de Aminoácido , Proteínas Virales de Fusión/genética
17.
J Virol ; 80(2): 623-33, 2006 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-16378965

RESUMEN

Most paramyxovirus fusion (F) proteins require the coexpression of the homologous attachment (HN) protein to promote membrane fusion, consistent with the existence of a virus-specific interaction between the two proteins. Analysis of the fusion activities of chimeric HN proteins indicates that the stalk region of the HN spike determines its F protein specificity, and analysis of a panel of site-directed mutants indicates that the F-interactive site resides in this region. Here, we use the addition of oligosaccharides to further explore the role of the HN stalk in the interaction with F. N-glycans were individually added at several positions in the stalk to determine their effects on the activities of HN, as well as its structure. N-glycan addition at positions 69 and 77 in the stalk specifically blocks fusion and the HN-F interaction without affecting either HN structure or its other activities. N-glycans added at other positions in the stalk modulate activities that reside in the globular head of HN. This correlates with an alteration of the tetrameric structure of the protein, as indicated by sucrose gradient sedimentation analyses. Finally, N-glycan addition in another region of HN (residues 124 to 152), predicted by a peptide-based analysis to mediate the interaction with F, does not significantly reduce the level of fusion, arguing strongly against this site being part of the F-interactive domain in HN. Our data support the idea that the F-interactive site on HN is defined by the stalk region of the protein.


Asunto(s)
Proteína HN/metabolismo , Virus de la Enfermedad de Newcastle/fisiología , Polisacáridos/metabolismo , Proteínas Virales de Fusión/metabolismo , Secuencia de Aminoácidos , Animales , Línea Celular , Proteína HN/química , Proteína HN/genética , Datos de Secuencia Molecular , Estructura Terciaria de Proteína , Replicación Viral
18.
J Clin Microbiol ; 43(8): 4229-33, 2005 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-16081986

RESUMEN

Newcastle disease virus (NDV) strains are classified as having high (velogenic), intermediate (mesogenic), or low (lentogenic) pathogenesis and virulence in chickens. Recent studies have established that the hemagglutinin-neuraminidase (HN) protein plays an important role in viral tropism and virulence. A monoclonal antibody (AVS-I) has previously been shown to be specific for lentogenic strains of NDV (Srinivasappa et al., Avian Dis. 30:562-567, 1986) and is routinely used to identify these strains. We have used competition antibody binding assays with a previously characterized panel of monoclonal antibodies, binding to chimeric HN proteins, and the characterization of an escape mutant to localize the binding site of AVS-I to the extreme carboxy terminus of the protein. In addition, we have shown that AVS-I does recognize at least one mesogenic strain and one velogenic strain of the virus, calling into question the potential of this antibody as a diagnostic reagent for avirulent NDV strains.


Asunto(s)
Anticuerpos Monoclonales/inmunología , Proteína HN/inmunología , Virus de la Enfermedad de Newcastle/aislamiento & purificación , Sitios de Unión de Anticuerpos , Epítopos , Proteína HN/química , Proteína HN/toxicidad , Pruebas de Inhibición de Hemaglutinación , Pruebas de Neutralización , Virus de la Enfermedad de Newcastle/inmunología , Virus de la Enfermedad de Newcastle/patogenicidad , Conformación Proteica , Relación Estructura-Actividad , Virulencia
19.
J Virol ; 79(2): 1180-90, 2005 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-15613345

RESUMEN

It has been shown that the L289A-mutated Newcastle disease virus (NDV) fusion (F) protein gains the ability to promote fusion of Cos-7 cells independent of the viral hemagglutinin-neuraminidase (HN) protein and exhibits a 50% enhancement in HN-dependent fusion over wild-type (wt) F protein. Here, we show that HN-independent fusion by L289A-F is not exhibited in BHK cells or in several other cell lines. However, similar to the results in Cos-7 cells, the mutated protein plus HN does promote 50 to 70% more fusion above wt levels in all of the cell lines tested. L289A-F protein exhibits the same specificity as the wt F protein for the homologous HN protein, as well as NDV-human parainfluenza virus 3 HN chimeras. The mutated F protein promotes fusion more effectively than the wt when it is coexpressed with either the chimeras or HN proteins deficient in receptor recognition activity. In addition, its fusogenic activity is significantly more resistant to removal of sialic acid on target cells. These findings are consistent with the demonstration that L289A-F interacts more efficiently with wt and mutated HN proteins than does wt F by a cell surface coimmunoprecipitation assay. Taken together, these findings indicate that L289A-F promotes fusion by a mechanism analogous to that of the wt protein with respect to the HN-F interaction but is less dependent on the attachment activity of HN. The phenotype of the mutated F protein correlates with a conformational change in the protein detectable by two different monoclonal antibodies. This conformational change may reflect a destabilization of F structure induced by the L289A substitution, which may in turn indicate a lower energy requirement for fusion activation.


Asunto(s)
Anticuerpos Monoclonales/inmunología , Proteína HN/fisiología , Fusión de Membrana , Virus de la Enfermedad de Newcastle/fisiología , Receptores Virales/fisiología , Proteínas Virales de Fusión/fisiología , Animales , Secuencia de Bases , Células COS , Cricetinae , Dimerización , Proteína HN/química , Datos de Secuencia Molecular , Ácido N-Acetilneuramínico/fisiología , Conformación Proteica , Proteínas Virales de Fusión/química
20.
J Virol ; 78(23): 13053-61, 2004 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-15542657

RESUMEN

The hemagglutinin-neuraminidase (HN) protein of Newcastle disease virus mediates attachment to sialic acid receptors, as well as cleavage of the same moiety. HN also interacts with the other viral glycoprotein, the fusion (F) protein, to promote membrane fusion. The ectodomain of the HN spike consists of a stalk and a terminal globular head. The most conserved part of the stalk consists of two heptad repeats separated by a nonhelical intervening region (residues 89 to 95). Several amino acid substitutions for a completely conserved proline residue in this region not only impair fusion and the HN-F interaction but also decrease neuraminidase activity in the globular domain, suggesting that the substitutions may alter HN structure. Substitutions for L94 also interfere with fusion and the HN-F interaction but have no significant effect on any other HN function. Amino acid substitutions at other positions in the intervening region also modulate only fusion. In all cases, diminished fusion correlates with a decreased ability of the mutated HN protein to interact with F at the cell surface. These findings indicate that the intervening region is critical to the role of HN in the promotion of fusion and may be directly involved in its interaction with the homologous F protein.


Asunto(s)
Proteína HN/química , Fusión de Membrana , Virus de la Enfermedad de Newcastle/fisiología , Proteínas Virales de Fusión/fisiología , Secuencia de Aminoácidos , Sustitución de Aminoácidos , Animales , Cricetinae , Proteína HN/fisiología , Datos de Secuencia Molecular , Neuraminidasa/metabolismo , Virus de la Enfermedad de Newcastle/química , Receptores Virales/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...