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











Base de dados
Intervalo de ano de publicação
1.
mSphere ; 2(5)2017.
Artigo em Inglês | MEDLINE | ID: mdl-28959737

RESUMO

Tacaribe virus (TCRV) is a mammalian arenavirus that was first isolated from artibeus bats in the 1950s. Subsequent experimental infection of Jamaican fruit bats (Artibeus jamaicensis) caused a disease similar to that of naturally infected bats. Although substantial attention has focused on bats as reservoir hosts of viruses that cause human disease, little is known about the interactions between bats and their pathogens. We performed a transcriptome-wide study to illuminate the response of Jamaican fruit bats experimentally infected with TCRV. Differential gene expression analysis of multiple tissues revealed global and organ-specific responses associated with innate antiviral responses, including interferon alpha/beta and Toll-like receptor signaling, activation of complement cascades, and cytokine signaling, among others. Genes encoding proteins involved in adaptive immune responses, such as gamma interferon signaling and costimulation of T cells by the CD28 family, were also altered in response to TCRV infection. Immunoglobulin gene expression was also elevated in the spleens of infected bats, including IgG, IgA, and IgE isotypes. These results indicate an active innate and adaptive immune response to TCRV infection occurred but did not prevent fatal disease. This de novo assembly provides a high-throughput data set of the Jamaican fruit bat and its host response to TCRV infection, which remains a valuable tool to understand the molecular signatures involved in antiviral responses in bats. IMPORTANCE As reservoir hosts of viruses associated with human disease, little is known about the interactions between bats and viruses. Using Jamaican fruit bats infected with Tacaribe virus (TCRV) as a model, we characterized the gene expression responses to infection in different tissues and identified pathways involved with the response to infection. This report is the most detailed gene discovery work in the species to date and the first to describe immune gene expression responses in bats during a pathogenic viral infection.

2.
Sci Rep ; 6: 21878, 2016 Feb 22.
Artigo em Inglês | MEDLINE | ID: mdl-26899616

RESUMO

The emergence of Middle East respiratory syndrome coronavirus (MERS-CoV) highlights the zoonotic potential of Betacoronaviruses. Investigations into the origin of MERS-CoV have focused on two potential reservoirs: bats and camels. Here, we investigated the role of bats as a potential reservoir for MERS-CoV. In vitro, the MERS-CoV spike glycoprotein interacted with Jamaican fruit bat (Artibeus jamaicensis) dipeptidyl peptidase 4 (DPP4) receptor and MERS-CoV replicated efficiently in Jamaican fruit bat cells, suggesting there is no restriction at the receptor or cellular level for MERS-CoV. To shed light on the intrinsic host-virus relationship, we inoculated 10 Jamaican fruit bats with MERS-CoV. Although all bats showed evidence of infection, none of the bats showed clinical signs of disease. Virus shedding was detected in the respiratory and intestinal tract for up to 9 days. MERS-CoV replicated transiently in the respiratory and, to a lesser extent, the intestinal tracts and internal organs; with limited histopathological changes observed only in the lungs. Analysis of the innate gene expression in the lungs showed a moderate, transient induction of expression. Our results indicate that MERS-CoV maintains the ability to replicate in bats without clinical signs of disease, supporting the general hypothesis of bats as ancestral reservoirs for MERS-CoV.


Assuntos
Infecções por Coronavirus/veterinária , Coronavírus da Síndrome Respiratória do Oriente Médio/fisiologia , Replicação Viral , Eliminação de Partículas Virais , Animais , Anticorpos Antivirais/sangue , Quirópteros/virologia , Chlorocebus aethiops , Infecções por Coronavirus/sangue , Infecções por Coronavirus/imunologia , Infecções por Coronavirus/virologia , Cricetinae , Dipeptidil Peptidase 4/metabolismo , Imunidade Inata , Pulmão/patologia , Pulmão/virologia , Receptores Virais/metabolismo , Células Vero , Carga Viral
3.
PLoS One ; 7(11): e48472, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-23166587

RESUMO

The Jamaican fruit bat (Artibeus jamaicensis) is one of the most common bats in the tropical Americas. It is thought to be a potential reservoir host of Tacaribe virus, an arenavirus closely related to the South American hemorrhagic fever viruses. We performed transcriptome sequencing and annotation from lung, kidney and spleen tissues using 454 and Illumina platforms to develop this species as an animal model. More than 100,000 contigs were assembled, with 25,000 genes that were functionally annotated. Of the remaining unannotated contigs, 80% were found within bat genomes or transcriptomes. Annotated genes are involved in a broad range of activities ranging from cellular metabolism to genome regulation through ncRNAs. Reciprocal BLAST best hits yielded 8,785 sequences that are orthologous to mouse, rat, cattle, horse and human. Species tree analysis of sequences from 2,378 loci was used to achieve 95% bootstrap support for the placement of bat as sister to the clade containing horse, dog, and cattle. Through substitution rate estimation between bat and human, 32 genes were identified with evidence for positive selection. We also identified 466 immune-related genes, which may be useful for studying Tacaribe virus infection of this species. The Jamaican fruit bat transcriptome dataset is a resource that should provide additional candidate markers for studying bat evolution and ecology, and tools for analysis of the host response and pathology of disease.


Assuntos
Infecções por Arenaviridae/genética , Arenavirus do Novo Mundo , Quirópteros/genética , Modelos Animais de Doenças , Filogenia , Seleção Genética , Transcriptoma/genética , Animais , Infecções por Arenaviridae/imunologia , Sequência de Bases , Quirópteros/metabolismo , Biologia Computacional , Mapeamento de Sequências Contíguas , Jamaica , Rim/metabolismo , Funções Verossimilhança , Pulmão/metabolismo , Modelos Genéticos , Anotação de Sequência Molecular , Dados de Sequência Molecular , Polimorfismo de Nucleotídeo Único/genética , Alinhamento de Sequência , Análise de Sequência de DNA , Especificidade da Espécie , Baço/metabolismo
4.
J Virol ; 86(10): 5791-9, 2012 May.
Artigo em Inglês | MEDLINE | ID: mdl-22379103

RESUMO

Tacaribe virus (TCRV) was first isolated from 11 Artibeus species bats captured in Trinidad in the 1950s during a rabies virus surveillance program. Despite significant effort, no evidence of infection of other mammals, mostly rodents, was found, suggesting that no other vertebrates harbored TCRV. For this reason, it was hypothesized that TCRV was naturally hosted by artibeus bats. This is in stark contrast to other arenaviruses with known hosts, all of which are rodents. To examine this hypothesis, we conducted experimental infections of Jamaican fruit bats (Artibeus jamaicensis) to determine whether they could be persistently infected without substantial pathology. We subcutaneously or intranasally infected bats with TCRV strain TRVL-11573, the only remaining strain of TCRV, and found that low-dose (10(4) 50% tissue culture infective dose [TCID(50)]) inoculations resulted in asymptomatic and apathogenic infection and virus clearance, while high-dose (10(6) TCID(50)) inoculations caused substantial morbidity and mortality as early as 10 days postinfection. Uninoculated cage mates failed to seroconvert, and viral RNA was not detected in their tissues, suggesting that transmission did not occur. Together, these data suggest that A. jamaicensis bats may not be a reservoir host for TCRV.


Assuntos
Infecções por Arenaviridae/veterinária , Arenaviridae/patogenicidade , Quirópteros/virologia , Reservatórios de Doenças/virologia , Animais , Arenaviridae/genética , Arenaviridae/isolamento & purificação , Arenaviridae/fisiologia , Infecções por Arenaviridae/mortalidade , Infecções por Arenaviridae/patologia , Infecções por Arenaviridae/virologia , Quirópteros/crescimento & desenvolvimento , Feminino , Masculino , Trinidad e Tobago , Virulência
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA