Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
J Avian Med Surg ; 38(1): 7-14, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-38686883

RESUMO

Avian polyomavirus (APV) infection causes various health problems in psittacine species, including death. The present study was conducted to investigate the prevalence of APV among psittacine birds in Iran. We also aimed to evaluate the impact of age, sex, species, season, and origin of the birds on the prevalence of APV. This study investigated the presence of APV among 1050 individual birds from 7 psittacine species over a 1-year period in Iran, namely, green-cheeked parakeets (Pyrrhura molinae), rosy-faced lovebirds (Agapornis roseicollis), monk parakeets (Myiopsitta monachus), sun conures (Aratinga solstitialis), Senegal parrots (Poicephalus senegalus), cockatiels (Nymphicus hollandicus), and grey parrots (Psittacus erithacus). The overall prevalence of APV in all studied species was 25% (263/1050, 95% confidence interval [CI]: 22.5-27.8). Results of the study showed that age and the season of the year were 2 important determinant factors in the prevalence of APV in psittacine birds. Young psittacine birds <6 months old were 2.94 (95% CI: 1.19-7.27) times more likely to be infected with APV than birds >1 year old, and there was a significant interaction between season and species in the multivariate analysis. In the winter season, rosy-faced lovebirds and green-cheeked parakeets were 15.6 (95% CI: 4.20-57.95) and 4.76 (95% CI: 1.4-16.21) times more likely to be infected with APV than in other seasons, respectively. This is the first report on the detection rate of APV in psittacine birds in Iran.


Assuntos
Doenças das Aves , Infecções por Polyomavirus , Polyomavirus , Psittaciformes , Animais , Irã (Geográfico)/epidemiologia , Doenças das Aves/epidemiologia , Doenças das Aves/virologia , Infecções por Polyomavirus/veterinária , Infecções por Polyomavirus/epidemiologia , Infecções por Polyomavirus/virologia , Fatores de Risco , Masculino , Feminino , Polyomavirus/isolamento & purificação , Prevalência , Estações do Ano , Infecções Tumorais por Vírus/veterinária , Infecções Tumorais por Vírus/epidemiologia , Infecções Tumorais por Vírus/virologia
2.
Virus Res ; 318: 198841, 2022 09.
Artigo em Inglês | MEDLINE | ID: mdl-35649483

RESUMO

The envelope attachment (H)-protein of canine distemper virus (CDV) mediates receptor engagement and fusion protein-triggering; two key functions in viral cell entry and spread. Signaling lymphocyte activation molecule (SLAM) and Nectin-4 (N4) act as morbilliviral entry receptors in immune and epithelial cells, respectively, which defines very similar pathogeneses. High incidence of brain disorders is however unique to CDV. The wild-type CDV-A75/17 strain (A75) preferentially infects glial cells and spreads from astrocyte-to-astrocyte without inducing massive fusion events, despite the fact that SLAM and N4 expressions remained below detection levels. To investigate whether an A75 H-microdomain required to interact with SLAM may additionally contribute to promote viral spread between astrocytes, we initially engineered a novel A75 H-protein variant (546-SYT/RNR-548) that lost SLAM-binding property and, consequently, lacked fusion protein-triggering activity specifically in SLAM-expressing cells. Collectively, this approach provides the molecular tool to decipher the role of the selected H-microdomain in supporting A75-spread in glial cells.


Assuntos
Vírus da Cinomose Canina , Cinomose , Animais , Antígenos CD/genética , Antígenos CD/metabolismo , Moléculas de Adesão Celular/genética , Moléculas de Adesão Celular/metabolismo , Chlorocebus aethiops , Vírus da Cinomose Canina/genética , Cães , Receptores de Superfície Celular/genética , Receptores de Superfície Celular/metabolismo , Células Vero
3.
J Virol ; 90(3): 1622-37, 2016 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-26608324

RESUMO

UNLABELLED: Measles virus (MeV) and canine distemper virus (CDV) possess tetrameric attachment proteins (H) and trimeric fusion proteins, which cooperate with either SLAM or nectin 4 receptors to trigger membrane fusion for cell entry. While the MeV H-SLAM cocrystal structure revealed the binding interface, two distinct oligomeric H assemblies were also determined. In one of the conformations, two SLAM units were sandwiched between two discrete H head domains, thus spotlighting two binding interfaces ("front" and "back"). Here, we investigated the functional relevance of both interfaces in activating the CDV membrane fusion machinery. While alanine-scanning mutagenesis identified five critical regulatory residues in the front H-binding site of SLAM, the replacement of a conserved glutamate residue (E at position 123, replaced with A [E123A]) led to the most pronounced impact on fusion promotion. Intriguingly, while determination of the interaction of H with the receptor using soluble constructs revealed reduced binding for the identified SLAM mutants, no effect was recorded when physical interaction was investigated with the full-length counterparts of both molecules. Conversely, although mutagenesis of three strategically selected residues within the back H-binding site of SLAM did not substantially affect fusion triggering, nevertheless, the mutants weakened the H-SLAM interaction recorded with the membrane-anchored protein constructs. Collectively, our findings support a mode of binding between the attachment protein and the V domain of SLAM that is common to all morbilliviruses and suggest a major role of the SLAM residue E123, located at the front H-binding site, in triggering the fusion machinery. However, our data additionally support the hypothesis that other microdomain(s) of both glycoproteins (including the back H-binding site) might be required to achieve fully productive H-SLAM interactions. IMPORTANCE: A complete understanding of the measles virus and canine distemper virus (CDV) cell entry molecular framework is still lacking, thus impeding the rational design of antivirals. Both viruses share many biological features that partially rely on the use of analogous Ig-like host cell receptors, namely, SLAM and nectin 4, for entering immune and epithelial cells, respectively. Here, we provide evidence that the mode of binding between the membrane-distal V domain of SLAM and the attachment protein (H) of morbilliviruses is very likely conserved. Moreover, although structural information revealed two discrete conformational states of H, one of the structures displayed two H-SLAM binding interfaces ("front" and "back"). Our data not only spotlight the front H-binding site of SLAM as the main determinant of membrane fusion promotion but suggest that the triggering efficiency of the viral entry machinery may rely on a local conformational change within the front H-SLAM interactive site rather than the binding affinity.


Assuntos
Antígenos CD/metabolismo , Vírus da Cinomose Canina/fisiologia , Interações Hospedeiro-Patógeno , Receptores de Superfície Celular/metabolismo , Internalização do Vírus , Animais , Antígenos CD/genética , Sítios de Ligação , Linhagem Celular , Análise Mutacional de DNA , Humanos , Proteínas de Fusão de Membrana/metabolismo , Proteínas Mutantes/genética , Proteínas Mutantes/metabolismo , Ligação Proteica , Receptores de Superfície Celular/genética , Membro 1 da Família de Moléculas de Sinalização da Ativação Linfocitária
4.
PLoS Pathog ; 11(5): e1004880, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25946112

RESUMO

Despite large vaccination campaigns, measles virus (MeV) and canine distemper virus (CDV) cause major morbidity and mortality in humans and animals, respectively. The MeV and CDV cell entry system relies on two interacting envelope glycoproteins: the attachment protein (H), consisting of stalk and head domains, co-operates with the fusion protein (F) to mediate membrane fusion. However, how receptor-binding by the H-protein leads to F-triggering is not fully understood. Here, we report that an anti-CDV-H monoclonal antibody (mAb-1347), which targets the linear H-stalk segment 126-133, potently inhibits membrane fusion without interfering with H receptor-binding or F-interaction. Rather, mAb-1347 blocked the F-triggering function of H-proteins regardless of the presence or absence of the head domains. Remarkably, mAb-1347 binding to headless CDV H, as well as standard and engineered bioactive stalk-elongated CDV H-constructs treated with cells expressing the SLAM receptor, was enhanced. Despite proper cell surface expression, fusion promotion by most H-stalk mutants harboring alanine substitutions in the 126-138 "spacer" section was substantially impaired, consistent with deficient receptor-induced mAb-1347 binding enhancement. However, a previously reported F-triggering defective H-I98A variant still exhibited the receptor-induced "head-stalk" rearrangement. Collectively, our data spotlight a distinct mechanism for morbillivirus membrane fusion activation: prior to receptor contact, at least one of the morbillivirus H-head domains interacts with the membrane-distal "spacer" domain in the H-stalk, leaving the F-binding site located further membrane-proximal in the stalk fully accessible. This "head-to-spacer" interaction conformationally stabilizes H in an auto-repressed state, which enables intracellular H-stalk/F engagement while preventing the inherent H-stalk's bioactivity that may prematurely activate F. Receptor-contact disrupts the "head-to-spacer" interaction, which subsequently "unlocks" the stalk, allowing it to rearrange and trigger F. Overall, our study reveals essential mechanistic requirements governing the activation of the morbillivirus membrane fusion cascade and spotlights the H-stalk "spacer" microdomain as a possible drug target for antiviral therapy.


Assuntos
Antígenos CD/metabolismo , Moléculas de Adesão Celular/metabolismo , Modelos Moleculares , Morbillivirus/fisiologia , Receptores de Superfície Celular/metabolismo , Proteínas Virais/metabolismo , Internalização do Vírus , Substituição de Aminoácidos , Animais , Anticorpos Monoclonais/farmacologia , Antígenos CD/química , Antígenos CD/genética , Moléculas de Adesão Celular/química , Moléculas de Adesão Celular/genética , Chlorocebus aethiops , Vírus da Cinomose Canina/metabolismo , Cães , Células HEK293 , Humanos , Fusão de Membrana/efeitos dos fármacos , Morbillivirus/efeitos dos fármacos , Mutação , Conformação Proteica , Dobramento de Proteína/efeitos dos fármacos , Domínios e Motivos de Interação entre Proteínas , Estabilidade Proteica/efeitos dos fármacos , Receptores de Superfície Celular/química , Receptores de Superfície Celular/genética , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo , Membro 1 da Família de Moléculas de Sinalização da Ativação Linfocitária , Células Vero , Proteínas Virais/antagonistas & inibidores , Proteínas Virais/química , Proteínas Virais/genética , Ligação Viral/efeitos dos fármacos , Internalização do Vírus/efeitos dos fármacos
5.
J Virol ; 89(10): 5724-33, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25787275

RESUMO

UNLABELLED: Measles and canine distemper viruses (MeV and CDV, respectively) first replicate in lymphatic and epithelial tissues by using SLAM and nectin-4 as entry receptors, respectively. The viruses may also invade the brain to establish persistent infections, triggering fatal complications, such as subacute sclerosis pan-encephalitis (SSPE) in MeV infection or chronic, multiple sclerosis-like, multifocal demyelinating lesions in the case of CDV infection. In both diseases, persistence is mediated by viral nucleocapsids that do not require packaging into particles for infectivity but are directly transmitted from cell to cell (neurons in SSPE or astrocytes in distemper encephalitis), presumably by relying on restricted microfusion events. Indeed, although morphological evidence of fusion remained undetectable, viral fusion machineries and, thus, a putative cellular receptor, were shown to contribute to persistent infections. Here, we first showed that nectin-4-dependent cell-cell fusion in Vero cells, triggered by a demyelinating CDV strain, remained extremely limited, thereby supporting a potential role of nectin-4 in mediating persistent infections in astrocytes. However, nectin-4 could not be detected in either primary cultured astrocytes or the white matter of tissue sections. In addition, a bioengineered "nectin-4-blind" recombinant CDV retained full cell-to-cell transmission efficacy in primary astrocytes. Combined with our previous report demonstrating the absence of SLAM expression in astrocytes, these findings are suggestive for the existence of a hitherto unrecognized third CDV receptor expressed by glial cells that contributes to the induction of noncytolytic cell-to-cell viral transmission in astrocytes. IMPORTANCE: While persistent measles virus (MeV) infection induces SSPE in humans, persistent canine distemper virus (CDV) infection causes chronic progressive or relapsing demyelination in carnivores. Common to both central nervous system (CNS) infections is that persistence is based on noncytolytic cell-to-cell spread, which, in the case of CDV, was demonstrated to rely on functional membrane fusion machinery complexes. This inferred a mechanism where nucleocapsids are transmitted through macroscopically invisible microfusion events between infected and target cells. Here, we provide evidence that CDV induces such microfusions in a SLAM- and nectin-4-independent manner, thereby strongly suggesting the existence of a third receptor expressed in glial cells (referred to as GliaR). We propose that GliaR governs intercellular transfer of nucleocapsids and hence contributes to viral persistence in the brain and ensuing demyelinating lesions.


Assuntos
Antígenos CD/metabolismo , Astrócitos/virologia , Moléculas de Adesão Celular/metabolismo , Vírus da Cinomose Canina/fisiologia , Vírus da Cinomose Canina/patogenicidade , Receptores de Superfície Celular/metabolismo , Substituição de Aminoácidos , Animais , Antígenos CD/genética , Encéfalo/metabolismo , Encéfalo/virologia , Moléculas de Adesão Celular/genética , Células Cultivadas , Chlorocebus aethiops , Cinomose/metabolismo , Cinomose/transmissão , Cinomose/virologia , Vírus da Cinomose Canina/genética , Cães , Genes Virais , Interações Hospedeiro-Patógeno , Humanos , Vírus do Sarampo/patogenicidade , Nectinas , Receptores de Superfície Celular/genética , Receptores Virais/genética , Receptores Virais/metabolismo , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Membro 1 da Família de Moléculas de Sinalização da Ativação Linfocitária , Panencefalite Esclerosante Subaguda/etiologia , Células Vero , Proteínas Virais/genética , Proteínas Virais/metabolismo , Internalização do Vírus
6.
J Virol ; 89(2): 1445-51, 2015 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-25355896

RESUMO

Membrane fusion for morbillivirus cell entry relies on critical interactions between the viral fusion (F) and attachment (H) envelope glycoproteins. Through extensive mutagenesis of an F cavity recently proposed to contribute to F's interaction with the H protein, we identified two neighboring hydrophobic residues responsible for severe F-to-H binding and fusion-triggering deficiencies when they were mutated in combination. Since both residues reside on one side of the F cavity, the data suggest that H binds the F globular head domain sideways.


Assuntos
Vírus da Cinomose Canina/fisiologia , Multimerização Proteica , Proteínas do Envelope Viral/metabolismo , Internalização do Vírus , Animais , Análise Mutacional de DNA , Vírus da Cinomose Canina/genética , Modelos Moleculares , Ligação Proteica , Conformação Proteica , Proteínas do Envelope Viral/genética
7.
J Virol ; 88(14): 8057-64, 2014 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-24807725

RESUMO

The hemagglutinin (H) gene of canine distemper virus (CDV) encodes the receptor-binding protein. This protein, together with the fusion (F) protein, is pivotal for infectivity since it contributes to the fusion of the viral envelope with the host cell membrane. Of the two receptors currently known for CDV (nectin-4 and the signaling lymphocyte activation molecule [SLAM]), SLAM is considered the most relevant for host susceptibility. To investigate how evolution might have impacted the host-CDV interaction, we examined the functional properties of a series of missense single nucleotide polymorphisms (SNPs) naturally accumulating within the H-gene sequences during the transition between two distinct but related strains. The two strains, a wild-type strain and a consensus strain, were part of a single continental outbreak in European wildlife and occurred in distinct geographical areas 2 years apart. The deduced amino acid sequence of the two H genes differed at 5 residues. A panel of mutants carrying all the combinations of the SNPs was obtained by site-directed mutagenesis. The selected mutant, wild type, and consensus H proteins were functionally evaluated according to their surface expression, SLAM binding, fusion protein interaction, and cell fusion efficiencies. The results highlight that the most detrimental functional effects are associated with specific sets of SNPs. Strikingly, an efficient compensational system driven by additional SNPs appears to come into play, virtually neutralizing the negative functional effects. This system seems to contribute to the maintenance of the tightly regulated function of the H-gene-encoded attachment protein. Importance: To investigate how evolution might have impacted the host-canine distemper virus (CDV) interaction, we examined the functional properties of naturally occurring single nucleotide polymorphisms (SNPs) in the hemagglutinin gene of two related but distinct strains of CDV. The hemagglutinin gene encodes the attachment protein, which is pivotal for infection. Our results show that few SNPs have a relevant detrimental impact and they generally appear in specific combinations (molecular signatures). These drastic negative changes are neutralized by compensatory mutations, which contribute to maintenance of an overall constant bioactivity of the attachment protein. This compensational mechanism might reflect the reaction of the CDV machinery to the changes occurring in the virus following antigenic variations critical for virulence.


Assuntos
Substituição de Aminoácidos , Vírus da Cinomose Canina/genética , Vírus da Cinomose Canina/fisiologia , Hemaglutininas Virais/genética , Hemaglutininas Virais/metabolismo , Mutação de Sentido Incorreto , Ligação Viral , Animais , Animais Selvagens , Antígenos CD/metabolismo , Análise Mutacional de DNA , Cinomose/epidemiologia , Cinomose/virologia , Vírus da Cinomose Canina/isolamento & purificação , Europa (Continente)/epidemiologia , Evolução Molecular , Mutagênese Sítio-Dirigida , Proteínas Mutantes/genética , Proteínas Mutantes/metabolismo , Ligação Proteica , Receptores de Superfície Celular/metabolismo , Receptores Virais/metabolismo , Membro 1 da Família de Moléculas de Sinalização da Ativação Linfocitária , Supressão Genética , Proteínas Virais de Fusão/metabolismo
8.
J Virol ; 88(5): 2951-66, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24371057

RESUMO

UNLABELLED: The morbillivirus cell entry machinery consists of a fusion (F) protein trimer that refolds to mediate membrane fusion following receptor-induced conformational changes in its binding partner, the tetrameric attachment (H) protein. To identify molecular determinants that control F refolding, we generated F chimeras between measles virus (MeV) and canine distemper virus (CDV). We located a central pocket in the globular head domain of CDV F that regulates the stability of the metastable, prefusion conformational state of the F trimer. Most mutations introduced into this "pocket'" appeared to mediate a destabilizing effect, a phenotype associated with enhanced membrane fusion activity. Strikingly, under specific triggering conditions (i.e., variation of receptor type and H protein origin), some F mutants also exhibited resistance to a potent morbillivirus entry inhibitor, which is known to block F triggering by enhancing the stability of prefusion F trimers. Our data reveal that the molecular nature of the F stimulus and the intrinsic stability of metastable prefusion F both regulate the efficiency of F refolding and escape from small-molecule refolding blockers. IMPORTANCE: With the aim to better characterize the thermodynamic basis of morbillivirus membrane fusion for cell entry and spread, we report here that the activation energy barrier of prefusion F trimers together with the molecular nature of the triggering "stimulus" (attachment protein and receptor types) define a "triggering range," which governs the initiation of the membrane fusion process. A central "pocket" microdomain in the globular F head contributes substantially to the regulation of the conformational stability of the prefusion complexes. The triggering range also defines the mechanism of viral escape from entry inhibitors and describes how the cellular environment can affect membrane fusion efficiency.


Assuntos
Vírus da Cinomose Canina/fisiologia , Fusão de Membrana , Proteínas Virais de Fusão/metabolismo , Substituição de Aminoácidos , Animais , Células CHO , Moléculas de Adesão Celular/genética , Moléculas de Adesão Celular/metabolismo , Fusão Celular , Chlorocebus aethiops , Cricetulus , Cães , Modelos Moleculares , Mutação , Nectinas , Ligação Proteica , Conformação Proteica , Multimerização Proteica , Estabilidade Proteica , Receptores Virais/metabolismo , Células Vero , Proteínas Virais de Fusão/química , Proteínas Virais de Fusão/genética , Proteínas Virais/química , Proteínas Virais/genética , Proteínas Virais/metabolismo , Internalização do Vírus
9.
PLoS One ; 8(3): e57488, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23554862

RESUMO

Canine distemper virus (CDV), a close relative of measles virus (MV), is widespread and well known for its broad host range. When the goal of measles eradication may be achieved, and when measles vaccination will be stopped, CDV might eventually cross the species barrier to humans and emerge as a new human pathogen. In order to get an impression how fast such alterations may occur, we characterized required adaptive mutations to the human entry receptors CD150 (SLAM) and nectin-4 as first step to infect human target cells. Recombinant wild-type CDV-A75/17(red) adapted quickly to growth in human H358 epithelial cells expressing human nectin-4. Sequencing of the viral attachment proteins (hemagglutinin, H, and fusion protein, F) genes revealed that no adaptive alteration was required to utilize human nectin-4. In contrast, the virus replicated only to low titres (10(2) pfu/ml) in Vero cells expressing human CD150 (Vero-hSLAM). After three passages using these cells virus was adapted to human CD150 and replicated to high titres (10(5) pfu/ml). Sequence analyses revealed that only one amino acid exchange in the H-protein at position 540 Asp→Gly (D540G) was required for functional adaptation to human CD150. Structural modelling suggests that the adaptive mutation D540G in H reflects the sequence alteration from canine to human CD150 at position 70 and 71 from Pro to Leu (P70L) and Gly to Glu (G71E), and compensates for the gain of a negative charge in the human CD150 molecule. Using this model system our data indicate that only a minimal alteration, in this case one adaptive mutation, is required for adaptation of CDV to the human entry receptors, and help to understand the molecular basis why this adaptive mutation occurs.


Assuntos
Antígenos CD/metabolismo , Vírus da Cinomose Canina/fisiologia , Cinomose/metabolismo , Receptores de Superfície Celular/metabolismo , Replicação Viral , Adaptação Fisiológica/genética , Substituição de Aminoácidos , Animais , Antígenos CD/genética , Moléculas de Adesão Celular/genética , Moléculas de Adesão Celular/metabolismo , Chlorocebus aethiops , Doenças Transmissíveis Emergentes/genética , Doenças Transmissíveis Emergentes/metabolismo , Doenças Transmissíveis Emergentes/transmissão , Cinomose/genética , Cinomose/transmissão , Cães , Humanos , Mutação de Sentido Incorreto , Receptores de Superfície Celular/genética , Membro 1 da Família de Moléculas de Sinalização da Ativação Linfocitária , Células Vero
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...