Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Cells ; 8(7)2019 07 15.
Artigo em Inglês | MEDLINE | ID: mdl-31311201

RESUMO

Zika virus (ZIKV) has emerged as an important human pathogen that can cause congenital defects in the fetus and neurological conditions in adults. The interferon (IFN) system has proven crucial in restricting ZIKV replication and pathogenesis. The canonical IFN response is triggered by the detection of viral RNA through RIG-I like receptors followed by activation of the adaptor protein MAVS on mitochondrial membranes. Recent studies have shown that a second organelle, peroxisomes, also function as a signaling platforms for the IFN response. Here, we investigated how ZIKV infection affects peroxisome biogenesis and antiviral signaling. We show that ZIKV infection depletes peroxisomes in human fetal astrocytes, a brain cell type that can support persistent infection. The peroxisome biogenesis factor PEX11B was shown to inhibit ZIKV replication, likely by increasing peroxisome numbers and enhancing downstream IFN-dependent antiviral signaling. Given that peroxisomes play critical roles in brain development and nerve function, our studies provide important insights into the roles of peroxisomes in regulating ZIKV infection and potentially neuropathogenesis.


Assuntos
Interações Hospedeiro-Patógeno , Peroxissomos/virologia , Zika virus/patogenicidade , Animais , Astrócitos/imunologia , Astrócitos/virologia , Linhagem Celular Tumoral , Células Cultivadas , Chlorocebus aethiops , Células HEK293 , Humanos , Imunidade Inata , Interferons/metabolismo , Proteínas de Membrana/metabolismo , Transdução de Sinais , Células Vero , Replicação Viral , Zika virus/fisiologia
2.
J Infect Dis ; 220(8): 1377-1387, 2019 09 13.
Artigo em Inglês | MEDLINE | ID: mdl-30799482

RESUMO

Zika virus (ZIKV) is an emerging pathogen that can cause microcephaly and other neurological defects in developing fetuses. The cellular response to ZIKV in the fetal brain is not well understood. Here, we show that ZIKV infection of human fetal astrocytes (HFAs), the most abundant cell type in the brain, results in elevated expression and secretion of fibroblast growth factor 2 (FGF2). This cytokine was shown to enhance replication and spread of ZIKV in HFAs and human fetal brain explants. The proviral effect of FGF2 is likely mediated in part by suppression of the interferon response, which would represent a novel mechanism by which viruses antagonize host antiviral defenses. We posit that FGF2-enhanced virus replication in the fetal brain contributes to the neurodevelopmental disorders associated with in utero ZIKV infection. As such, targeting FGF2-dependent signaling should be explored further as a strategy to limit replication of ZIKV.


Assuntos
Fator 2 de Crescimento de Fibroblastos/metabolismo , Microcefalia/patologia , Replicação Viral , Infecção por Zika virus/patologia , Zika virus/patogenicidade , Feto Abortado , Aedes , Animais , Astrócitos/imunologia , Astrócitos/patologia , Astrócitos/virologia , Encéfalo/citologia , Linhagem Celular , Humanos , Interferons/imunologia , Microcefalia/virologia , Cultura Primária de Células , Técnicas de Cultura de Tecidos , Zika virus/imunologia , Infecção por Zika virus/virologia
3.
Viruses ; 10(11)2018 11 17.
Artigo em Inglês | MEDLINE | ID: mdl-30453621

RESUMO

Zika virus (ZIKV) infection and persistence during pregnancy can lead to microcephaly and other fetal neurological disorders collectively known as Congenital Zika Syndrome. The immunological and virological events that contribute to the establishment of persistent ZIKV infection in humans are unclear though. Here we show that human fetal astrocytes (HFAs), the most abundant cell type in the central nervous system, become persistently infected with ZIKV resulting in continuous viral shedding for at least one month; a process that is facilitated by TIM/TAM receptors. HFAs are relatively resistant to ZIKV-induced apoptosis, a factor that may be important for chronic infection of these cells. Once infection was established, interferon treatment did not reduce virus replication. Moreover, the fact that the innate immune system was highly activated in persistently infected HFAs indicates that the virus can thrive in the presence of a sustained antiviral response. RNAseq analyses of persistently infected cells revealed that ZIKV alters host gene expression in a manner that could affect developmental processes. Conversely, data from sequencing of ZIKV genomes in persistently infected HFAs suggest that adaptive mutations were not required for establishing chronic infection. Based on these results, we postulate that HFAs are reservoirs for ZIKV in the fetal brain and that moderate apoptosis combined with inefficient antiviral response from these cells may contribute to the establishment of chronic brain infection associated with the ZIKV neurodevelopmental abnormalities.


Assuntos
Apoptose , Astrócitos/imunologia , Astrócitos/virologia , Interações Hospedeiro-Patógeno , Evasão da Resposta Imune , Interferons/antagonistas & inibidores , Zika virus/crescimento & desenvolvimento , Células Cultivadas , Perfilação da Expressão Gênica , Humanos , Análise de Sequência de RNA
4.
Sci Rep ; 8(1): 5477, 2018 04 03.
Artigo em Inglês | MEDLINE | ID: mdl-29615760

RESUMO

Zika virus is a teratogenic mosquito-transmitted flavivirus that is associated with birth defects in newborns and Guillain-Barré syndrome in adults. The virus can also be sexually transmitted, but currently, very little is known about the cell types supporting virus replication and persistence in human testes. Using primary cell cultures, we observed that Sertoli but not Leydig cells are highly susceptible to Zika virus infection, a process that is dependent on the TAM family receptor Axl. In cell culture, Sertoli cells could be productively infected with Zika virus for at least 6-weeks. Infection of Sertoli cells resulted in dramatic changes to the transcriptional profile of these cells. The most upregulated mRNA in infected cells was basic fibroblast growth factor (FGF2), a cytokine that was found to enhance Zika virus replication and support viral persistence. Together these findings provide key insights into understanding how Zika virus persists in the male reproductive tract and in turn may aid in developing antiviral therapies or strategies to minimize sexual transmission of this pathogen.


Assuntos
Células de Sertoli/virologia , Replicação Viral , Zika virus/fisiologia , Células A549 , Animais , Fator 2 de Crescimento de Fibroblastos/metabolismo , Regulação da Expressão Gênica , Humanos , Masculino , Camundongos , Proteínas Proto-Oncogênicas/metabolismo , Receptores Proteína Tirosina Quinases/metabolismo , Células de Sertoli/citologia , Células de Sertoli/metabolismo , Transdução de Sinais , Internalização do Vírus , Receptor Tirosina Quinase Axl
5.
Am J Surg ; 215(5): 788-792, 2018 05.
Artigo em Inglês | MEDLINE | ID: mdl-29352568

RESUMO

BACKGROUND: The objective of this study was to evaluate the performance of dual-energy computed tomography (DECT) for preoperative parathyroid tumor (PT) localization in primary hyperparathyroid (PHP) patients. METHODS: A retrospective review was carried out of the medical records of all PHP patients who underwent ultrasound (US), Tc-99m sestamibi noncontrast single photon emission computed tomography (CT-MIBI), DECT and parathyroidectomy at a single center. RESULTS: The sensitivities and accuracies for preoperative PT localization in the 97 patient study population were: US 40% and 93%, CT-MIBI 64.0% and 97%, and DECT 84% and 96%, respectively. In the one third of the study population that did not localize preoperatively with CT-MIBI and US, DECT correctly localized a PT in 21 cases (66%). DE-CT and US correctly localized a PT in 86% of cases, and only 5 (8%) of the cases that were accurately localized by a combination of CT-MIBI and US were not identified by DE-CT. CONCLUSIONS: DECT should be utilized as a first line preoperative PT localization study in PHP patients, and is also a sensitive salvage localization test.


Assuntos
Hiperparatireoidismo Primário/diagnóstico por imagem , Hiperparatireoidismo Primário/cirurgia , Tomografia Computadorizada por Raios X/métodos , Adulto , Idoso , Idoso de 80 Anos ou mais , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Paratireoidectomia , Doses de Radiação , Compostos Radiofarmacêuticos , Estudos Retrospectivos , Sensibilidade e Especificidade , Tomografia Computadorizada com Tomografia Computadorizada de Emissão de Fóton Único , Tecnécio Tc 99m Sestamibi , Ultrassonografia
6.
J Virol ; 91(16)2017 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-28592527

RESUMO

Zika virus (ZIKV), a member of the Flaviviridae family, has recently emerged as an important human pathogen with increasing economic and health impact worldwide. Because of its teratogenic nature and association with the serious neurological condition Guillain-Barré syndrome, a tremendous amount of effort has focused on understanding ZIKV pathogenesis. To gain further insights into ZIKV interaction with host cells, we investigated how this pathogen affects stress response pathways. While ZIKV infection induces stress signaling that leads to phosphorylation of eIF2α and cellular translational arrest, stress granule (SG) formation was inhibited. Further analysis revealed that the viral proteins NS3 and NS4A are linked to translational repression, whereas expression of the capsid protein, NS3/NS2B-3, and NS4A interfered with SG formation. Some, but not all, flavivirus capsid proteins also blocked SG assembly, indicating differential interactions between flaviviruses and SG biogenesis pathways. Depletion of the SG components G3BP1, TIAR, and Caprin-1, but not TIA-1, reduced ZIKV replication. Both G3BP1 and Caprin-1 formed complexes with capsid, whereas viral genomic RNA stably interacted with G3BP1 during ZIKV infection. Taken together, these results are consistent with a scenario in which ZIKV uses multiple viral components to hijack key SG proteins to benefit viral replication.IMPORTANCE There is a pressing need to understand ZIKV pathogenesis in order to advance the development of vaccines and therapeutics. The cellular stress response constitutes one of the first lines of defense against viral infection; therefore, understanding how ZIKV evades this antiviral system will provide key insights into ZIKV biology and potentially pathogenesis. Here, we show that ZIKV induces the stress response through activation of the UPR (unfolded protein response) and PKR (protein kinase R), leading to host translational arrest, a process likely mediated by the viral proteins NS3 and NS4A. Despite the activation of translational shutoff, formation of SG is strongly inhibited by the virus. Specifically, ZIKV hijacks the core SG proteins G3BP1, TIAR, and Caprin-1 to facilitate viral replication, resulting in impaired SG assembly. This process is potentially facilitated by the interactions of the viral RNA with G3BP1 as well as the viral capsid protein with G3BP1 and Caprin-1. Interestingly, expression of capsid proteins from several other flaviviruses also inhibited SG formation. Taken together, the present study provides novel insights into how ZIKV modulates cellular stress response pathways during replication.

7.
EMBO Rep ; 17(12): 1766-1775, 2016 12.
Artigo em Inglês | MEDLINE | ID: mdl-27797853

RESUMO

Zika virus is an emerging mosquito-borne pathogen that is associated with Guillain-Barré syndrome in adults and microcephaly and other neurological defects in newborns. Despite being declared an international emergency by the World Health Organization, comparatively little is known about its biology. Here, we investigate the strategies employed by the virus to suppress the host antiviral response. We observe that once established, Zika virus infection is impervious to interferon treatment suggesting that the virus deploys effective countermeasures to host cell defences. This is confirmed by experiments showing that Zika virus infection impairs the induction of type-I interferon as well as downstream interferon-stimulated genes. Multiple viral proteins affect these processes. Virus-mediated degradation of STAT2 acts to reduce type-I and type-III interferon-mediated signaling. Further, the NS5 of Zika virus binds to STAT2, and its expression is correlated with STAT2 degradation by the proteasome. Together, our findings provide key insights into how Zika virus blocks cellular defense systems. This in turn is important for understanding pathogenesis and may aid in designing antiviral therapies.


Assuntos
Interações Hospedeiro-Patógeno , Interferon Tipo I/metabolismo , Transdução de Sinais , Infecção por Zika virus/imunologia , Zika virus/imunologia , Zika virus/patogenicidade , Células A549 , Adulto , Células HEK293 , Humanos , Interferon Tipo I/imunologia , Ligação Proteica , Fator de Transcrição STAT2/metabolismo , Proteínas não Estruturais Virais/genética , Proteínas não Estruturais Virais/metabolismo , Infecção por Zika virus/metabolismo , Infecção por Zika virus/virologia
8.
J Virol ; 89(24): 12349-61, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26423946

RESUMO

UNLABELLED: Flaviviruses are significant human pathogens that have an enormous impact on the global health burden. Currently, there are very few vaccines against or therapeutic treatments for flaviviruses, and our understanding of how these viruses cause disease is limited. Evidence suggests that the capsid proteins of flaviviruses play critical nonstructural roles during infection, and therefore, elucidating how these viral proteins affect cellular signaling pathways could lead to novel targets for antiviral therapy. We used affinity purification to identify host cell proteins that interact with the capsid proteins of West Nile and dengue viruses. One of the cellular proteins that formed a stable complex with flavivirus capsid proteins is the peroxisome biogenesis factor Pex19. Intriguingly, flavivirus infection resulted in a significant loss of peroxisomes, an effect that may be due in part to capsid expression. We posited that capsid protein-mediated sequestration and/or degradation of Pex19 results in loss of peroxisomes, a situation that could result in reduced early antiviral signaling. In support of this hypothesis, we observed that induction of the lambda interferon mRNA in response to a viral RNA mimic was reduced by more than 80%. Together, our findings indicate that inhibition of peroxisome biogenesis may be a novel mechanism by which flaviviruses evade the innate immune system during early stages of infection. IMPORTANCE: RNA viruses infect hundreds of millions of people each year, causing significant morbidity and mortality. Chief among these pathogens are the flaviviruses, which include dengue virus and West Nile virus. Despite their medical importance, there are very few prophylactic or therapeutic treatments for these viruses. Moreover, the manner in which they subvert the innate immune response in order to establish infection in mammalian cells is not well understood. Recently, peroxisomes were reported to function in early antiviral signaling, but very little is known regarding if or how pathogenic viruses affect these organelles. We report for the first time that flavivirus infection results in significant loss of peroxisomes in mammalian cells, which may indicate that targeting of peroxisomes is a key strategy used by viruses to subvert early antiviral defenses.


Assuntos
Vírus da Dengue/metabolismo , Dengue/metabolismo , Peroxissomos/metabolismo , Transdução de Sinais , Febre do Nilo Ocidental/metabolismo , Vírus do Nilo Ocidental/metabolismo , Dengue/genética , Vírus da Dengue/genética , Células HEK293 , Humanos , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Peroxissomos/genética , Febre do Nilo Ocidental/genética , Vírus do Nilo Ocidental/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...