Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 4 de 4
Filtrar
Mais filtros

Base de dados
Tipo de documento
Assunto da revista
País de afiliação
Intervalo de ano de publicação
1.
Parasitol Res ; 118(7): 2277-2285, 2019 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-31119381

RESUMO

Malaria-associated bacteremia accounts for up to one-third of deaths from severe malaria, and non-typhoidal Salmonella (NTS) has been reported as a major complication of severe malarial infection. Patients who develop NTS bacteremia during Plasmodium infection show higher mortality rates than individuals with malaria alone. Systemic bacteremia can be caused by a wound or translocation from epithelial or endothelial sites. NTS is an intestinal pathogen, however the contribution of bacterial translocation from the intestinal tract during Plasmodium infection is not well studied. Here, we investigated the integrity of the intestinal barrier function of P. chabaudi-infected mice using large molecules and Salmonella infection. Intestinal histology and the adaptive immune response to malaria were also studied using light microscopy and flow cytometry. P. chabaudi infection compromised intestinal barrier function, which led to increased intestinal cellular infiltration. In addition, we observed increased serum lipopolysaccharide binding protein and leakage of soluble molecules from the intestine into the blood in infected mice. Plasmodium infection also increased intestinal translocation and dissemination of NTS to the liver. The adaptive immune response to P. chabaudi infection was also significantly impacted by NTS translocation. Reduced B and T cell activation were observed in co-infected animals, suggesting interference in the malaria-specific immune responses by bacteremia. These studies demonstrate that P. chabaudi infection induces failure of the barrier function of the intestinal wall and enhanced intestinal bacterial translocation, affecting anti-malarial immunity.


Assuntos
Imunidade Adaptativa , Malária/imunologia , Plasmodium chabaudi/imunologia , Infecções por Salmonella/imunologia , Salmonella/imunologia , Animais , Bacteriemia , Coinfecção , Modelos Animais de Doenças , Feminino , Microbioma Gastrointestinal , Intestinos/microbiologia , Intestinos/patologia , Ativação Linfocitária , Malária/complicações , Malária/parasitologia , Malária/patologia , Camundongos , Camundongos Endogâmicos C57BL , Parasitemia , Infecções por Salmonella/complicações , Infecções por Salmonella/microbiologia , Infecções por Salmonella/patologia
2.
J Gen Virol ; 94(Pt 7): 1441-1450, 2013 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-23515022

RESUMO

Rift Valley fever is a mosquito-borne zoonotic disease endemic to sub-Saharan Africa. Rift Valley fever virus (RVFV; genus Phlebovirus, family Bunyaviridae) causes high rates of abortion and fetal malformation in pregnant ruminants, and haemorrhagic fever, neurological disorders or blindness in humans. The MP-12 strain is a highly efficacious and safe live-attenuated vaccine candidate for both humans and ruminants. However, MP-12 lacks a marker to differentiate infected from vaccinated animals. In this study, we originally aimed to characterize the efficacy of a recombinant RVFV MP-12 strain encoding Toscana virus (TOSV) NSs gene in place of MP-12 NSs (rMP12-TOSNSs). TOSV NSs promotes the degradation of dsRNA-dependent protein kinase (PKR) and inhibits interferon-ß gene up-regulation without suppressing host general transcription. Unexpectedly, rMP12-TOSNSs increased death in vaccinated outbred mice and inbred BALB/c or C57BL/6 mice. Immunohistochemistry showed diffusely positive viral antigens in the thalamus, hypothalamus and brainstem, including the medulla. No viral antigens were detected in spleen or liver, which is similar to the antigen distribution of moribund mice infected with MP-12. These results suggest that rMP12-TOSNSs retains neuroinvasiveness in mice. Our findings demonstrate that rMP12-TOSNSs causes neuroinvasion without any hepatic disease and will be useful for studying the neuroinvasion mechanism of RVFV and TOSV.


Assuntos
Encéfalo/virologia , Doenças do Sistema Nervoso/virologia , Febre do Vale de Rift/prevenção & controle , Vírus da Febre do Flebótomo Napolitano/genética , Vírus da Febre do Flebótomo Napolitano/patogenicidade , Vacinas Atenuadas/efeitos adversos , Proteínas não Estruturais Virais/metabolismo , Vacinas Virais/efeitos adversos , Animais , Linhagem Celular , Chlorocebus aethiops , Feminino , Humanos , Imunoglobulina G/sangue , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Febre do Vale de Rift/imunologia , Vírus da Febre do Vale do Rift/imunologia , Vírus da Febre do Flebótomo Napolitano/imunologia , Vacinação , Vacinas Atenuadas/administração & dosagem , Vacinas Atenuadas/imunologia , Células Vero , Proteínas não Estruturais Virais/imunologia , Vacinas Virais/administração & dosagem , Vacinas Virais/imunologia
3.
Viruses ; 8(6)2016 05 24.
Artigo em Inglês | MEDLINE | ID: mdl-27231931

RESUMO

The Rift Valley fever virus (RVFV) M-segment encodes the 78 kD, NSm, Gn, and Gc proteins. The 1st AUG generates the 78 kD-Gc precursor, the 2nd AUG generates the NSm-Gn-Gc precursor, and the 3rd AUG makes the NSm'-Gn-Gc precursor. To understand biological changes due to abolishment of the precursors, we quantitatively measured Gn secretion using a reporter assay, in which a Gaussia luciferase (gLuc) protein is fused to the RVFV M-segment pre-Gn region. Using the reporter assay, the relative expression of Gn/gLuc fusion proteins was analyzed among various AUG mutants. The reporter assay showed efficient secretion of Gn/gLuc protein from the precursor made from the 2nd AUG, while the removal of the untranslated region upstream of the 2nd AUG (AUG2-M) increased the secretion of the Gn/gLuc protein. Subsequently, recombinant MP-12 strains encoding mutations in the pre-Gn region were rescued, and virological phenotypes were characterized. Recombinant MP-12 encoding the AUG2-M mutation replicated slightly less efficiently than the control, indicating that viral replication is further influenced by the biological processes occurring after Gn expression, rather than the Gn abundance. This study showed that, not only the abolishment of AUG, but also the truncation of viral UTR, affects the expression of Gn protein by the RVFV M-segment.


Assuntos
Análise Mutacional de DNA , Perfilação da Expressão Gênica , Glicoproteínas/metabolismo , Precursores de Proteínas/metabolismo , Vírus da Febre do Vale do Rift/genética , Proteínas Virais/metabolismo , Animais , Linhagem Celular , Códon de Iniciação , Genes Reporter , Glicoproteínas/genética , Humanos , Luciferases/análise , Luciferases/genética , Precursores de Proteínas/genética , Proteínas Recombinantes de Fusão , Vírus da Febre do Vale do Rift/fisiologia , Proteínas Virais/genética , Replicação Viral
4.
Viruses ; 8(5)2016 05 23.
Artigo em Inglês | MEDLINE | ID: mdl-27223297

RESUMO

Rift Valley fever is a mosquito-transmitted, zoonotic disease that infects humans and ruminants. Dendritic cell specific intercellular adhesion molecule 3 (ICAM-3) grabbing non-integrin (DC-SIGN) acts as a receptor for members of the phlebovirus genus. The Rift Valley fever virus (RVFV) glycoproteins (Gn/Gc) encode five putative N-glycan sequons (asparagine (N)-any amino acid (X)-serine (S)/threonine (T)) at positions: N438 (Gn), and N794, N829, N1035, and N1077 (Gc). The N-glycosylation profile and significance in viral infection via DC-SIGN have not been elucidated. Gc N-glycosylation was first evaluated by using Gc asparagine (N) to glutamine (Q) mutants. Subsequently, we generated a series of recombinant RVFV MP-12 strain mutants, which encode N-to-Q mutations, and the infectivity of each mutant in Jurkat cells stably expressing DC-SIGN was evaluated. Results showed that Gc N794, N1035, and N1077 were N-glycosylated but N829 was not. Gc N1077 was heterogeneously N-glycosylated. RVFV Gc made two distinct N-glycoforms: "Gc-large" and "Gc-small", and N1077 was responsible for "Gc-large" band. RVFV showed increased infection of cells expressing DC-SIGN compared to cells lacking DC-SIGN. Infection via DC-SIGN was increased in the presence of either Gn N438 or Gc N1077. Our study showed that N-glycans on the Gc and Gn surface glycoproteins redundantly support RVFV infection via DC-SIGN.


Assuntos
Moléculas de Adesão Celular/metabolismo , Glicoproteínas/metabolismo , Lectinas Tipo C/metabolismo , Polissacarídeos/metabolismo , Receptores de Superfície Celular/metabolismo , Receptores Virais/metabolismo , Vírus da Febre do Vale do Rift/fisiologia , Proteínas Estruturais Virais/metabolismo , Ligação Viral , Substituição de Aminoácidos , Glicoproteínas/genética , Humanos , Células Jurkat , Proteínas Mutantes/genética , Proteínas Mutantes/metabolismo , Vírus da Febre do Vale do Rift/genética , Proteínas Estruturais Virais/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA