Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 20
Filtrar
1.
Nature ; 585(7825): 414-419, 2020 09.
Artigo em Inglês | MEDLINE | ID: mdl-32641828

RESUMO

Zika virus (ZIKV) belongs to the family Flaviviridae, and is related to other viruses that cause human diseases. Unlike other flaviviruses, ZIKV infection can cause congenital neurological disorders and replicates efficiently in reproductive tissues1-3. Here we show that the envelope protein (E) of ZIKV is polyubiquitinated by the E3 ubiquitin ligase TRIM7 through Lys63 (K63)-linked polyubiquitination. Accordingly, ZIKV replicates less efficiently in the brain and reproductive tissues of Trim7-/- mice. Ubiquitinated E is present on infectious virions of ZIKV when they are released from specific cell types, and enhances virus attachment and entry into cells. Specifically, K63-linked polyubiquitin chains directly interact with the TIM1 (also known as HAVCR1) receptor of host cells, which enhances virus entry in cells as well as in brain tissue in vivo. Recombinant ZIKV mutants that lack ubiquitination are attenuated in human cells and in wild-type mice, but not in live mosquitoes. Monoclonal antibodies against K63-linked polyubiquitin specifically neutralize ZIKV and reduce viraemia in mice. Our results demonstrate that the ubiquitination of ZIKV E is an important determinant of virus entry, tropism and pathogenesis.


Assuntos
Ubiquitinação , Proteínas do Envelope Viral/química , Proteínas do Envelope Viral/metabolismo , Internalização do Vírus , Zika virus/metabolismo , Zika virus/patogenicidade , Animais , Anticorpos Monoclonais/imunologia , Anticorpos Neutralizantes/imunologia , Encéfalo/metabolismo , Linhagem Celular , Culicidae/citologia , Culicidae/virologia , Endossomos/metabolismo , Feminino , Receptor Celular 1 do Vírus da Hepatite A/metabolismo , Humanos , Masculino , Fusão de Membrana , Camundongos , Especificidade de Órgãos , Poliubiquitina/imunologia , Poliubiquitina/metabolismo , Proteínas com Motivo Tripartido/metabolismo , Ubiquitina-Proteína Ligases/metabolismo , Tropismo Viral , Viremia/imunologia , Viremia/prevenção & controle , Viremia/virologia , Replicação Viral , Zika virus/química , Zika virus/genética , Infecção por Zika virus/prevenção & controle , Infecção por Zika virus/virologia
2.
PLoS Pathog ; 17(12): e1009678, 2021 12.
Artigo em Inglês | MEDLINE | ID: mdl-34855915

RESUMO

Kyasanur Forest disease virus (KFDV) and the closely related Alkhurma hemorrhagic disease virus (AHFV) are emerging flaviviruses that cause severe viral hemorrhagic fevers in humans. Increasing geographical expansion and case numbers, particularly of KFDV in southwest India, class these viruses as a public health threat. Viral pathogenesis is not well understood and additional vaccines and antivirals are needed to effectively counter the impact of these viruses. However, current animal models of KFDV pathogenesis do not accurately reproduce viral tissue tropism or clinical outcomes observed in humans. Here, we show that pigtailed macaques (Macaca nemestrina) infected with KFDV or AHFV develop viremia that peaks 2 to 4 days following inoculation. Over the course of infection, animals developed lymphocytopenia, thrombocytopenia, and elevated liver enzymes. Infected animals exhibited hallmark signs of human disease characterized by a flushed appearance, piloerection, dehydration, loss of appetite, weakness, and hemorrhagic signs including epistaxis. Virus was commonly present in the gastrointestinal tract, consistent with human disease caused by KFDV and AHFV where gastrointestinal symptoms (hemorrhage, vomiting, diarrhea) are common. Importantly, RNAseq of whole blood revealed that KFDV downregulated gene expression of key clotting factors that was not observed during AHFV infection, consistent with increased severity of KFDV disease observed in this model. This work characterizes a nonhuman primate model for KFDV and AHFV that closely resembles human disease for further utilization in understanding host immunity and development of antiviral countermeasures.


Assuntos
Modelos Animais de Doenças , Vírus da Encefalite Transmitidos por Carrapatos/patogenicidade , Encefalite Transmitida por Carrapatos/virologia , Febres Hemorrágicas Virais/virologia , Macaca nemestrina , Animais , Chlorocebus aethiops , Citocinas/sangue , Vírus da Encefalite Transmitidos por Carrapatos/genética , Vírus da Encefalite Transmitidos por Carrapatos/imunologia , Encefalite Transmitida por Carrapatos/imunologia , Encefalite Transmitida por Carrapatos/patologia , Feminino , Células HEK293 , Febres Hemorrágicas Virais/imunologia , Febres Hemorrágicas Virais/patologia , Humanos , Linfonodos/virologia , Células Vero , Viremia
3.
J Immunol ; 200(2): 471-476, 2018 01 15.
Artigo em Inglês | MEDLINE | ID: mdl-29246952

RESUMO

Inflammatory monocyte (iMO) recruitment to the brain is a hallmark of many neurologic diseases. Prior to entering the brain, iMOs must egress into the blood from the bone marrow through a mechanism, which for known encephalitic viruses, is CCR2 dependent. In this article, we show that during La Crosse Virus-induced encephalitis, egress of iMOs was surprisingly independent of CCR2, with similar percentages of iMOs in the blood and brain of heterozygous and CCR2-/- mice following infection. Interestingly, CCR2 was required for iMO trafficking from perivascular areas to sites of virus infection within the brain. Thus, CCR2 was not essential for iMO trafficking to the blood or the brain but was essential for trafficking within the brain parenchyma. Analysis of other orthobunyaviruses showed that Jamestown Canyon virus also induced CCR2-independent iMO egress to the blood. These studies demonstrate that the CCR2 requirement for iMO egress to the blood is not universal for all viruses.


Assuntos
Antígenos Ly/metabolismo , Encefalite da Califórnia/imunologia , Encefalite da Califórnia/metabolismo , Vírus La Crosse , Monócitos/imunologia , Monócitos/metabolismo , Receptores CCR2/metabolismo , Animais , Células da Medula Óssea/imunologia , Células da Medula Óssea/metabolismo , Células da Medula Óssea/patologia , Encéfalo/imunologia , Encéfalo/metabolismo , Encéfalo/patologia , Encéfalo/virologia , Quimiotaxia de Leucócito/imunologia , Modelos Animais de Doenças , Encefalite da Califórnia/virologia , Feminino , Masculino , Camundongos , Camundongos Transgênicos , Monócitos/patologia
4.
J Virol ; 92(17)2018 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-29925658

RESUMO

Chikungunya virus (CHIKV) is a mosquito-borne alphavirus that has evolved effective mechanisms to counteract the type I interferon (IFN) response. Upon recognition of the virus, cells secrete IFNs, which signal through transmembrane receptors (IFNAR) to phosphorylate STAT proteins (pSTAT). pSTAT dimers are transported into the nucleus by importin-α5 and activate the transcription of IFN-stimulated genes (ISGs), increasing cellular resistance to infection. Subsequently, STAT proteins are shuttled back into the cytoplasm by the exportin CRM1. CHIKV nonstructural protein 2 (nsP2) reduces ISG expression by inhibiting general host cell transcription and by specifically reducing the levels of nuclear pSTAT1 via an unknown mechanism. To systematically examine where nsP2 acts within the JAK/STAT signaling cascade, we used two well-characterized mutants of nsP2, P718S and KR649AA. Both mutations abrogate nsP2's ability to shut off host transcription, but only the KR649AA mutant localizes exclusively to the cytoplasm and no longer specifically inhibits JAK/STAT signaling. These mutant nsP2 proteins did not differentially affect IFNAR expression levels or STAT1 phosphorylation in response to IFNs. Coimmunoprecipitation experiments showed that in the presence of nsP2, STAT1 still effectively bound importin-α5. Chemically blocking CRM1-mediated nuclear export in the presence of nsP2 additionally showed that nuclear translocation of STAT1 is not affected by nsP2. nsP2 putatively has five domains. Redirecting the nsP2 KR649AA mutant or just nsP2's C-terminal methyltransferase-like domain into the nucleus strongly reduced nuclear pSTAT in response to IFN stimulation. This demonstrates that the C-terminal domain of nuclear nsP2 specifically inhibits the IFN response by promoting the nuclear export of STAT1.IMPORTANCE Chikungunya virus is an emerging pathogen associated with large outbreaks on the African, Asian, European, and both American continents. In most patients, infection results in high fever, rash, and incapacitating (chronic) arthralgia. CHIKV effectively inhibits the first line of defense, the innate immune response. As a result, stimulation of the innate immune response with interferons (IFNs) is ineffective as a treatment for CHIKV disease. The IFN response requires an intact downstream signaling cascade called the JAK/STAT signaling pathway, which is effectively inhibited by CHIKV nonstructural protein 2 (nsP2) via an unknown mechanism. The research described here specifies where in the JAK/STAT signaling cascade the IFN response is inhibited and which protein domain of nsP2 is responsible for IFN inhibition. The results illuminate new aspects of antiviral defense and CHIKV counterdefense strategies and will direct the search for novel antiviral compounds.


Assuntos
Vírus Chikungunya/enzimologia , Cisteína Endopeptidases/genética , Imunidade Inata , Interferon Tipo I/antagonistas & inibidores , Fator de Transcrição STAT1/genética , Linhagem Celular , Vírus Chikungunya/genética , Vírus Chikungunya/fisiologia , Simulação por Computador , Cisteína Endopeptidases/metabolismo , Células HEK293 , Interações Hospedeiro-Patógeno/genética , Interações Hospedeiro-Patógeno/imunologia , Humanos , Imunoprecipitação , Interferon Tipo I/genética , Interferon Tipo I/imunologia , Metiltransferases/metabolismo , Mutação , Transdução de Sinais/genética , Transdução de Sinais/imunologia , Replicação Viral
5.
J Immunol ; 198(9): 3526-3535, 2017 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-28330900

RESUMO

The recent association between Zika virus (ZIKV) and neurologic complications, including Guillain-Barré syndrome in adults and CNS abnormalities in fetuses, highlights the importance in understanding the immunological mechanisms controlling this emerging infection. Studies have indicated that ZIKV evades the human type I IFN response, suggesting a role for the adaptive immune response in resolving infection. However, the inability of ZIKV to antagonize the mouse IFN response renders the virus highly susceptible to circulating IFN in murine models. Thus, as we show in this article, although wild-type C57BL/6 mice mount cell-mediated and humoral adaptive immune responses to ZIKV, these responses were not required to prevent disease. However, when the type I IFN response of mice was suppressed, then the adaptive immune responses became critical. For example, when type I IFN signaling was blocked by Abs in Rag1-/- mice, the mice showed dramatic weight loss and ZIKV infection in the brain and testes. This phenotype was not observed in Ig-treated Rag1-/- mice or wild-type mice treated with anti-type I IFNR alone. Furthermore, we found that the CD8+ T cell responses of pregnant mice to ZIKV infection were diminished compared with nonpregnant mice. It is possible that diminished cell-mediated immunity during pregnancy could increase virus spread to the fetus. These results demonstrate an important role for the adaptive immune response in the control of ZIKV infection and imply that vaccination may prevent ZIKV-related disease, particularly when the type I IFN response is suppressed as it is in humans.


Assuntos
Imunidade Adaptativa , Encéfalo/virologia , Linfócitos T CD8-Positivos/virologia , Complicações Infecciosas na Gravidez/imunologia , Testículo/virologia , Infecção por Zika virus/imunologia , Zika virus/imunologia , Animais , Anticorpos Bloqueadores/administração & dosagem , Encéfalo/imunologia , Linfócitos T CD8-Positivos/imunologia , Modelos Animais de Doenças , Feminino , Proteínas de Homeodomínio/genética , Humanos , Evasão da Resposta Imune , Interferon Tipo I/imunologia , Interferon Tipo I/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Gravidez/imunologia , Testículo/imunologia , Infecção por Zika virus/epidemiologia
6.
J Infect Dis ; 214(suppl 3): S303-S307, 2016 10 15.
Artigo em Inglês | MEDLINE | ID: mdl-27471319

RESUMO

The development of point-of-care clinical chemistry analyzers has enabled the implementation of these ancillary tests in field laboratories in resource-limited outbreak areas. The Eternal Love Winning Africa (ELWA) outbreak diagnostic laboratory, established in Monrovia, Liberia, to provide Ebola virus and Plasmodium spp. diagnostics during the Ebola epidemic, implemented clinical chemistry analyzers in December 2014. Clinical chemistry testing was performed for 68 patients in triage, including 12 patients infected with Ebola virus and 18 infected with Plasmodium spp. The main distinguishing feature in clinical chemistry of Ebola virus-infected patients was the elevation in alanine aminotransferase, aspartate aminotransferase, alkaline phosphatase, and γ-glutamyltransferase levels and the decrease in calcium. The implementation of clinical chemistry is probably most helpful when the medical supportive care implemented at the Ebola treatment unit allows for correction of biochemistry derangements and on-site clinical chemistry analyzers can be used to monitor electrolyte balance.


Assuntos
Surtos de Doenças , Epidemias , Doença pelo Vírus Ebola/diagnóstico , Doença pelo Vírus Ebola/epidemiologia , Malária/diagnóstico , Adolescente , Alanina Transaminase/análise , Fosfatase Alcalina/análise , Aspartato Aminotransferases/análise , Química Clínica , Serviços de Laboratório Clínico , Ebolavirus/imunologia , Ebolavirus/isolamento & purificação , Doença pelo Vírus Ebola/virologia , Humanos , Libéria/epidemiologia , Testes de Função Hepática , Malária/epidemiologia , Malária/parasitologia , Masculino , Plasmodium/isolamento & purificação , Plasmodium/metabolismo , Sistemas Automatizados de Assistência Junto ao Leito , gama-Glutamiltransferase/análise
7.
J Infect Dis ; 214(suppl 3): S169-S176, 2016 10 15.
Artigo em Inglês | MEDLINE | ID: mdl-27333914

RESUMO

West Africa experienced the first epidemic of Ebola virus infection, with by far the greatest number of cases in Guinea, Sierra Leone, and Liberia. The unprecedented epidemic triggered an unparalleled response, including the deployment of multiple Ebola treatment units and mobile/field diagnostic laboratories. The National Institute of Allergy and Infectious Diseases and the Centers for Disease Control and Prevention deployed a joint laboratory to Monrovia, Liberia, in August 2014 to support the newly founded Ebola treatment unit at the Eternal Love Winning Africa (ELWA) campus. The laboratory operated initially out of a tent structure but quickly moved into a fixed-wall building owing to severe weather conditions, the need for increased security, and the high sample volume. Until May 2015, when the laboratory closed, the site handled close to 6000 clinical specimens for Ebola virus diagnosis and supported the medical staff in case patient management. Laboratory operation and safety, as well as Ebola virus diagnostic assays, are described and discussed; in addition, lessons learned for future deployments are reviewed.


Assuntos
Serviços de Laboratório Clínico/organização & administração , Ebolavirus/isolamento & purificação , Epidemias/prevenção & controle , Doença pelo Vírus Ebola/epidemiologia , África Ocidental/epidemiologia , Centers for Disease Control and Prevention, U.S. , Feminino , Guiné/epidemiologia , Doença pelo Vírus Ebola/diagnóstico , Doença pelo Vírus Ebola/transmissão , Doença pelo Vírus Ebola/virologia , Humanos , Cooperação Internacional , Libéria/epidemiologia , Masculino , National Institute of Allergy and Infectious Diseases (U.S.) , Segurança , Serra Leoa/epidemiologia , Estados Unidos
8.
Nat Commun ; 14(1): 4481, 2023 07 25.
Artigo em Inglês | MEDLINE | ID: mdl-37491352

RESUMO

Inflammation in response to severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) infection drives severity of coronavirus disease 2019 (COVID-19) and is influenced by host genetics. To understand mechanisms of inflammation, animal models that reflect genetic diversity and clinical outcomes observed in humans are needed. We report a mouse panel comprising the genetically diverse Collaborative Cross (CC) founder strains crossed to human ACE2 transgenic mice (K18-hACE2) that confers susceptibility to SARS-CoV-2. Infection of CC x K18-hACE2 resulted in a spectrum of survival, viral replication kinetics, and immune profiles. Importantly, in contrast to the K18-hACE2 model, early type I interferon (IFN-I) and regulated proinflammatory responses were required for control of SARS-CoV-2 replication in PWK x K18-hACE2 mice that were highly resistant to disease. Thus, virus dynamics and inflammation observed in COVID-19 can be modeled in diverse mouse strains that provide a genetically tractable platform for understanding anti-coronavirus immunity.


Assuntos
COVID-19 , Interferon Tipo I , Humanos , Camundongos , Animais , Citocinas , SARS-CoV-2 , Camundongos Transgênicos , Inflamação/genética , Modelos Animais de Doenças , Pulmão
9.
Viruses ; 13(3)2021 03 10.
Artigo em Inglês | MEDLINE | ID: mdl-33801908

RESUMO

An evolutionary arms race has been ongoing between retroviruses and their primate hosts for millions of years. Within the last century, a zoonotic transmission introduced the Human Immunodeficiency Virus (HIV-1), a retrovirus, to the human population that has claimed the lives of millions of individuals and is still infecting over a million people every year. To counteract retroviruses such as this, primates including humans have evolved an innate immune sensor for the retroviral capsid lattice known as TRIM5α. Although the molecular basis for its ability to restrict retroviruses is debated, it is currently accepted that TRIM5α forms higher-order assemblies around the incoming retroviral capsid that are not only disruptive for the virus lifecycle, but also trigger the activation of an antiviral state. More recently, it was discovered that TRIM5α restriction is broader than previously thought because it restricts not only the human retroelement LINE-1, but also the tick-borne flaviviruses, an emergent group of RNA viruses that have vastly different strategies for replication compared to retroviruses. This review focuses on the underlying mechanisms of TRIM5α-mediated restriction of retroelements and flaviviruses and how they differ from the more widely known ability of TRIM5α to restrict retroviruses.


Assuntos
Capsídeo/imunologia , Imunidade Inata , Vírus de RNA/imunologia , Vírus de RNA/metabolismo , Proteínas com Motivo Tripartido/metabolismo , Ubiquitina-Proteína Ligases/metabolismo , Fatores de Restrição Antivirais , Capsídeo/metabolismo , Proteínas de Transporte/genética , Flavivirus/imunologia , Flavivirus/metabolismo , Humanos , Vírus de RNA/classificação , Vírus de RNA/genética , Retroviridae/imunologia , Retroviridae/metabolismo , Infecções por Retroviridae/imunologia , Infecções por Retroviridae/prevenção & controle , Proteínas com Motivo Tripartido/genética , Proteínas com Motivo Tripartido/imunologia , Ubiquitina-Proteína Ligases/genética , Ubiquitina-Proteína Ligases/imunologia
10.
Cell Rep ; 37(4): 109888, 2021 10 26.
Artigo em Inglês | MEDLINE | ID: mdl-34706234

RESUMO

Dysregulated inflammation dominated by chemokine expression is a key feature of disease following infection with the globally important human pathogens Zika virus (ZIKV) and dengue virus, but a mechanistic understanding of how pro-inflammatory responses are initiated is lacking. Mitophagy is a quality-control mechanism that regulates innate immune signaling and cytokine production through selective degradation of damaged mitochondria. Here, we demonstrate that ZIKV nonstructural protein 5 (NS5) antagonizes mitophagy by binding to the host protein Ajuba and preventing its translocation to depolarized mitochondria where it is required for PINK1 activation and downstream signaling. Consequent mitophagy suppression amplifies the production of pro-inflammatory chemokines through protein kinase R (PKR) sensing of mitochondrial RNA. In Ajuba-/- mice, ZIKV induces early expression of pro-inflammatory chemokines associated with significantly enhanced dissemination to tissues. This work identifies Ajuba as a critical regulator of mitophagy and demonstrates a role for mitophagy in limiting systemic inflammation following infection by globally important human viruses.


Assuntos
Proteínas com Domínio LIM/metabolismo , Mitofagia , Proteínas Quinases/metabolismo , Transdução de Sinais , Infecção por Zika virus/metabolismo , Zika virus/metabolismo , eIF-2 Quinase/metabolismo , Células A549 , Animais , Chlorocebus aethiops , Células HEK293 , Células HeLa , Humanos , Proteínas com Domínio LIM/genética , Camundongos , Camundongos Knockout , Proteínas Quinases/genética , Células Vero , Zika virus/genética , Infecção por Zika virus/genética , eIF-2 Quinase/genética
11.
J Virol ; 83(9): 4469-75, 2009 May.
Artigo em Inglês | MEDLINE | ID: mdl-19225008

RESUMO

The hallmark of transmissible spongiform encephalopathies (TSEs or prion diseases) is the accumulation of an abnormally folded, partially protease-resistant form (PrP-res) of the normal protease-sensitive prion protein (PrP-sen). PrP-sen is attached to the cell membrane by a glycosylphosphatidylinositol (GPI) anchor. In vitro, the anchor and the local membrane environment are important for the conversion of PrP-sen to PrP-res. In vivo, however, the anchor is not necessary because transgenic mice expressing anchorless PrP-sen accumulate PrP-res and replicate infectivity. To clarify the role of the GPI anchor in TSE infection, cells expressing GPI-anchored PrP-sen, anchorless PrP-sen, or both forms of PrP-sen were exposed to the mouse scrapie strain 22L. Cells expressing anchored PrP-sen produced PrP-res after exposure to 22L. Surprisingly, while cells expressing anchorless PrP-sen made anchorless PrP-res in the first 96 h postinfection, no PrP-res was detected at later passes. In contrast, when cells expressing both forms of PrP-sen were exposed to 22L, both anchored and anchorless PrP-res were detected over multiple passes. Consistent with the in vitro data, scrapie-infected cells expressing anchored PrP-sen transmitted disease to mice whereas cells expressing anchorless PrP-sen alone did not. These results demonstrate that the GPI anchor on PrP-sen is important for the persistent infection of cells in vitro. Our data suggest that cells expressing anchorless PrP-sen are not directly infected with scrapie. Thus, PrP-res formation in transgenic mice expressing anchorless PrP-sen may be occurring extracellularly.


Assuntos
Príons/metabolismo , Scrapie/metabolismo , Animais , Linhagem Celular , Camundongos , Camundongos Transgênicos
12.
iScience ; 15: 489-501, 2019 May 31.
Artigo em Inglês | MEDLINE | ID: mdl-31129244

RESUMO

Tick-borne flaviviruses (TBFVs) can cause life-threatening encephalitis and hemorrhagic fever. To identify virus-host interactions that may be exploited as therapeutic targets, we analyzed the TBFV polyprotein in silico for antiviral protein-binding motifs. We obtained two putative tumor necrosis factor receptor-associated factor 6 (TRAF6)-binding motifs (TBMs) within the protease domain of the viral nonstructural 3 (NS3) protein. Here, we show that TBFV NS3 interacted with TRAF6 during infection and that TRAF6 supports TBFV replication. The proviral role of TRAF6 was not seen with mosquito-borne flaviviruses, consistent with the lack of conserved TBMs. Mutation of the second TBM within NS3 disrupted TRAF6 binding, coincident with reduced abundance of mature, autocatalytically derived form of the NS3 protease and significant virus attenuation in vitro. Our studies reveal insights into how flaviviruses exploit innate immunity for the purpose of viral replication and identify a potential target for therapeutic design.

13.
Cell Rep ; 27(11): 3269-3283.e6, 2019 06 11.
Artigo em Inglês | MEDLINE | ID: mdl-31189110

RESUMO

Tripartite motif-containing protein 5α (TRIM5α) is a cellular antiviral restriction factor that prevents early events in retrovirus replication. The activity of TRIM5α is thought to be limited to retroviruses as a result of highly specific interactions with capsid lattices. In contrast to this current understanding, we show that both human and rhesus macaque TRIM5α suppress replication of specific flaviviruses. Multiple viruses in the tick-borne encephalitis complex are sensitive to TRIM5α-dependent restriction, but mosquito-borne flaviviruses, including yellow fever, dengue, and Zika viruses, are resistant. TRIM5α suppresses replication by binding to the viral protease NS2B/3 to promote its K48-linked ubiquitination and proteasomal degradation. Importantly, TRIM5α contributes to the antiviral function of IFN-I against sensitive flaviviruses in human cells. Thus, TRIM5α possesses remarkable plasticity in the recognition of diverse virus families, with the potential to influence human susceptibility to emerging flaviviruses of global concern.


Assuntos
Infecções por Flavivirus/metabolismo , Peptídeo Hidrolases/metabolismo , Complexo de Endopeptidases do Proteassoma/metabolismo , Proteínas com Motivo Tripartido/metabolismo , Ubiquitina-Proteína Ligases/metabolismo , Proteínas Virais/metabolismo , Replicação Viral , Animais , Fatores de Restrição Antivirais , Gatos , Chlorocebus aethiops , Células Dendríticas/metabolismo , Células Dendríticas/virologia , Flavivirus/patogenicidade , Flavivirus/fisiologia , Infecções por Flavivirus/virologia , Células HEK293 , Humanos , Ligação Proteica , Proteólise , Especificidade por Substrato , Ubiquitinação , Células Vero
14.
Artigo em Inglês | MEDLINE | ID: mdl-29696134

RESUMO

The common small animal disease models for Zika virus (ZIKV) are mice lacking the interferon responses, but infection of interferon receptor α/ß knock out (IFNAR-/-) mice is not uniformly lethal particularly in older animals. Here we sought to advance this model in regard to lethality for future countermeasure efficacy testing against more recent ZIKV strains from the Asian lineage, preferably the American sublineage. We first infected IFNAR-/- mice subcutaneously with the contemporary ZIKV-Paraiba strain resulting in predominantly neurological disease with ~50% lethality. Infection with ZIKV-Paraiba by different routes established a uniformly lethal model only in young mice (4-week old) upon intraperitoneal infection. However, intraperitoneal inoculation of ZIKV-French Polynesia resulted in uniform lethality in older IFNAR-/- mice (10-12-weeks old). In conclusion, we have established uniformly lethal mouse disease models for efficacy testing of antivirals and vaccines against recent ZIKV strains representing the Asian lineage.


Assuntos
Modelos Animais de Doenças , Receptor de Interferon alfa e beta/genética , Infecção por Zika virus/mortalidade , Infecção por Zika virus/patologia , Zika virus/isolamento & purificação , Aedes , Fatores Etários , Animais , Linhagem Celular , Chlorocebus aethiops , Feminino , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Células Vero , Zika virus/patogenicidade , Infecção por Zika virus/virologia
15.
Cell Host Microbe ; 23(5): 672-685.e6, 2018 05 09.
Artigo em Inglês | MEDLINE | ID: mdl-29746837

RESUMO

Progress toward understanding Zika virus (ZIKV) pathogenesis is hindered by lack of immunocompetent small animal models, in part because ZIKV fails to effectively antagonize Stat2-dependent interferon (IFN) responses in mice. To address this limitation, we first passaged an African ZIKV strain (ZIKV-Dak-41525) through Rag1-/- mice to obtain a mouse-adapted virus (ZIKV-Dak-MA) that was more virulent than ZIKV-Dak-41525 in mice treated with an anti-Ifnar1 antibody. A G18R substitution in NS4B was the genetic basis for the increased replication, and resulted in decreased IFN-ß production, diminished IFN-stimulated gene expression, and the greater brain infection observed with ZIKV-Dak-MA. To generate a fully immunocompetent mouse model of ZIKV infection, human STAT2 was introduced into the mouse Stat2 locus (hSTAT2 KI). Subcutaneous inoculation of pregnant hSTAT2 KI mice with ZIKV-Dak-MA resulted in spread to the placenta and fetal brain. An immunocompetent mouse model of ZIKV infection may prove valuable for evaluating countermeasures to limit disease.


Assuntos
Camundongos/imunologia , Infecção por Zika virus/imunologia , Zika virus/imunologia , Zika virus/patogenicidade , Animais , Encéfalo , Sobrevivência Celular , Modelos Animais de Doenças , Feminino , Doenças Fetais/metabolismo , Doenças Fetais/virologia , Proteínas de Homeodomínio/genética , Proteínas de Homeodomínio/metabolismo , Humanos , Imunidade , Transmissão Vertical de Doenças Infecciosas , Interferon beta/metabolismo , Interferons/metabolismo , Camundongos/genética , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Mutação , Placenta/metabolismo , Gravidez , Complicações Infecciosas na Gravidez/virologia , RNA Helicases/genética , Receptor de Interferon alfa e beta , Fator de Transcrição STAT2/metabolismo , Serina Endopeptidases/genética , Proteínas não Estruturais Virais/genética , Zika virus/genética , Infecção por Zika virus/virologia
16.
PLoS One ; 12(6): e0179781, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28650973

RESUMO

Tick-borne flaviviruses (TBFVs), including Powassan virus and tick-borne encephalitis virus cause encephalitis or hemorrhagic fevers in humans with case-fatality rates ranging from 1-30%. Despite severe disease in humans, TBFV infection of natural rodent hosts has little noticeable effect. Currently, the basis for resistance to disease is not known. We hypothesize that the coevolution of flaviviruses with their respective hosts has shaped the evolution of potent antiviral factors that suppress virus replication and protect the host from lethal infection. In the current study, we compared virus infection between reservoir host cells and related susceptible species. Infection of primary fibroblasts from the white-footed mouse (Peromyscus leucopus, a representative host) with a panel of vector-borne flaviviruses showed up to a 10,000-fold reduction in virus titer compared to control Mus musculus cells. Replication of vesicular stomatitis virus was equivalent in P. leucopus and M. musculus cells suggesting that restriction was flavivirus-specific. Step-wise comparison of the virus infection cycle revealed a significant block to viral RNA replication, but not virus entry, in P. leucopus cells. To understand the role of the type I interferon (IFN) response in virus restriction, we knocked down signal transducer and activator of transcription 1 (STAT1) or the type I IFN receptor (IFNAR1) by RNA interference. Loss of IFNAR1 or STAT1 significantly relieved the block in virus replication in P. leucopus cells. The major IFN antagonist encoded by TBFV, nonstructural protein 5, was functional in P. leucopus cells, thus ruling out ineffective viral antagonism of the host IFN response. Collectively, this work demonstrates that the IFN response of P. leucopus imparts a strong and virus-specific barrier to flavivirus replication. Future identification of the IFN-stimulated genes responsible for virus restriction specifically in P. leucopus will yield mechanistic insight into efficient control of virus replication and may inform the development of antiviral therapeutics.


Assuntos
Vírus da Encefalite Transmitidos por Carrapatos/imunologia , Vírus da Encefalite Transmitidos por Carrapatos/patogenicidade , Interferon Tipo I/imunologia , Peromyscus/imunologia , Peromyscus/virologia , Animais , Células Cultivadas , Modelos Animais de Doenças , Vírus da Encefalite Transmitidos por Carrapatos/genética , Encefalite Transmitida por Carrapatos/genética , Encefalite Transmitida por Carrapatos/imunologia , Encefalite Transmitida por Carrapatos/virologia , Especificidade de Hospedeiro/genética , Especificidade de Hospedeiro/imunologia , Interações Hospedeiro-Patógeno/genética , Interações Hospedeiro-Patógeno/imunologia , Humanos , Interferon Tipo I/antagonistas & inibidores , Camundongos , Peromyscus/genética , RNA Interferente Pequeno/genética , RNA Viral/genética , Receptor de Interferon alfa e beta/antagonistas & inibidores , Receptor de Interferon alfa e beta/genética , Receptor de Interferon alfa e beta/imunologia , Fator de Transcrição STAT1/antagonistas & inibidores , Fator de Transcrição STAT1/genética , Fator de Transcrição STAT1/imunologia , Transdução de Sinais/genética , Transdução de Sinais/imunologia , Proteínas não Estruturais Virais/imunologia , Replicação Viral/genética , Replicação Viral/imunologia
17.
Cell Host Microbe ; 18(1): 61-74, 2015 Jul 08.
Artigo em Inglês | MEDLINE | ID: mdl-26159719

RESUMO

Type I interferon (IFN-α/ß or IFN-I) signals through two receptor subunits, IFNAR1 and IFNAR2, to orchestrate sterile and infectious immunity. Cellular pathways that regulate IFNAR1 are often targeted by viruses to suppress the antiviral effects of IFN-I. Here we report that encephalitic flaviviruses, including tick-borne encephalitis virus and West Nile virus, antagonize IFN-I signaling by inhibiting IFNAR1 surface expression. Loss of IFNAR1 was associated with binding of the viral IFN-I antagonist, NS5, to prolidase (PEPD), a cellular dipeptidase implicated in primary immune deficiencies in humans. Prolidase was required for IFNAR1 maturation and accumulation, activation of IFNß-stimulated gene induction, and IFN-I-dependent viral control. Human fibroblasts derived from patients with genetic prolidase deficiency exhibited decreased IFNAR1 surface expression and reduced IFNß-stimulated signaling. Thus, by understanding flavivirus IFN-I antagonism, prolidase is revealed as a central regulator of IFN-I responses.


Assuntos
Dipeptidases/metabolismo , Vírus da Encefalite Transmitidos por Carrapatos/imunologia , Interações Hospedeiro-Patógeno , Interferon Tipo I/metabolismo , Receptor de Interferon alfa e beta/metabolismo , Transdução de Sinais , Vírus do Nilo Ocidental/imunologia , Fibroblastos/imunologia , Humanos , Ligação Proteica , Proteínas não Estruturais Virais/metabolismo
18.
Ticks Tick Borne Dis ; 3(1): 18-26, 2012 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-22309855

RESUMO

Ixodid ticks are vectors of human diseases such as Lyme disease, babesiosis, anaplasmosis, and tick-borne encephalitis. These diseases cause significant morbidity and mortality worldwide and are transmitted to humans during tick feeding. The tick-host-pathogen interface is a complex environment where host responses are modulated by the molecules in tick saliva to enable the acquisition of a blood meal. Disruption of host responses at the site of the tick bite may also provide an advantage for pathogens to survive and replicate. Thus, the molecules in tick saliva not only aid the tick in securing a nutrient-rich blood meal, but can also enhance the transmission and acquisition of pathogens. To investigate the effect of feeding and flavivirus infection on the salivary gland transcript expression profile in ticks, a first-generation microarray was developed using ESTs from a cDNA library derived from Ixodes scapularis salivary glands. When the salivary gland transcript profile in ticks feeding over the course of 3 days was compared to that in unfed ticks, a dramatic increase in transcripts related to metabolism was observed. Specifically, 578 transcripts were up-regulated compared to 151 down-regulated transcripts in response to feeding. When specific time points post attachment were analyzed, a temporal pattern of gene expression was observed. When Langat virus-infected ticks were compared to mock-infected ticks, transcript expression changes were observed at all 3 days of feeding. Differentially regulated transcripts include putative secreted proteins, lipocalins, Kunitz domain-containing proteins, anti-microbial peptides, and transcripts of unknown function. These studies identify salivary gland transcripts that are differentially regulated during feeding or in the context of flavivirus infection in Ixodes scapularis nymphs, a medically important disease vector. Further analysis of these transcripts may identify salivary factors that affect the transmission or replication of tick-borne flaviviruses.


Assuntos
Vetores Aracnídeos/genética , Vírus da Encefalite Transmitidos por Carrapatos/fisiologia , Regulação da Expressão Gênica/genética , Ixodes/genética , Animais , Vetores Aracnídeos/fisiologia , Vetores Aracnídeos/virologia , DNA Complementar/química , Regulação para Baixo/genética , Encefalite Transmitida por Carrapatos/transmissão , Encefalite Transmitida por Carrapatos/virologia , Perfilação da Expressão Gênica , Biblioteca Gênica , Interações Hospedeiro-Patógeno , Humanos , Ixodes/fisiologia , Ixodes/virologia , Camundongos , Ninfa/genética , Ninfa/fisiologia , Ninfa/virologia , Análise de Sequência com Séries de Oligonucleotídeos , RNA/genética , RNA/isolamento & purificação , Glândulas Salivares/fisiologia , Glândulas Salivares/virologia , Fatores de Tempo
19.
Prion ; 3(3): 134-8, 2009.
Artigo em Inglês | MEDLINE | ID: mdl-19786843

RESUMO

Normal cellular and abnormal disease-associated forms of prion protein (PrP) contain a C-terminal glycophosphatidyl-inositol (GPI) membrane anchor. The importance of the GPI membrane anchor in prion diseases is unclear but there are data to suggest that it both is and is not required for abnormal prion protein formation and prion infection. Utilizing an in vitro model of prion infection we have recently demonstrated that, while the GPI anchor is not essential for the formation of abnormal prion protein in a cell, it is necessary for the establishment of persistent prion infection. In combination with previously published data, our results suggest that GPI anchored PrP is important in the amplification and spread of prion infectivity from cell to cell.


Assuntos
Membranas Intracelulares/metabolismo , Doenças Priônicas/fisiopatologia , Príons/metabolismo , Animais , Encéfalo/patologia , Modelos Animais de Doenças , Epitopos/química , Glicosilfosfatidilinositóis/química , Humanos , Camundongos , Doenças Priônicas/metabolismo , Scrapie/fisiopatologia , Fatores de Tempo
20.
Protein Eng Des Sel ; 22(8): 515-21, 2009 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-19570812

RESUMO

The pathological isoform of the prion protein (PrP(res)) can serve as a marker for prion diseases, but more practical tests are needed for preclinical diagnosis and sensitive detection of many prion infections. Previously we showed that the quaking-induced conversion (QuIC) assay can detect sub-femtogram levels of PrP(res) in scrapie-infected hamster brain tissue and distinguish cerebral spinal fluid (CSF) samples from normal and scrapie-infected hamsters. We now report the adaptation of the QuIC reaction to prion diseases of medical and agricultural interest: human variant Creutzfeldt-Jakob disease (vCJD) and sheep scrapie. PrP(res)-positive and -negative brain homogenates from humans and sheep were discriminated within 1-2 days with a sensitivity of 10-100 fg PrP(res). More importantly, in as little as 22 h we were able to distinguish CSF samples from scrapie-infected and uninfected sheep. These results suggest the presence of prions in CSF from scrapie-infected sheep. This new method enables the relatively rapid and sensitive detection of human CJD and sheep scrapie PrP(res) and may facilitate the development of practical preclinical diagnostic and high-throughput interference tests.


Assuntos
Bioensaio/métodos , Síndrome de Creutzfeldt-Jakob/metabolismo , Príons/análise , Scrapie/metabolismo , Animais , Química Encefálica , Cricetinae , Humanos , Príons/líquido cefalorraquidiano , Príons/isolamento & purificação , Príons/metabolismo , Proteínas Recombinantes/isolamento & purificação , Proteínas Recombinantes/metabolismo , Ovinos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA