Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 25
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
Microbiol Spectr ; 11(3): e0032623, 2023 06 15.
Artigo em Inglês | MEDLINE | ID: mdl-37022262

RESUMO

Epstein-Barr virus (EBV) infects host cells and establishes a latent infection that requires evasion of host innate immunity. A variety of EBV-encoded proteins that manipulate the innate immune system have been reported, but whether other EBV proteins participate in this process is unclear. EBV-encoded envelope glycoprotein gp110 is a late protein involved in virus entry into target cells and enhancement of infectivity. Here, we reported that gp110 inhibits RIG-I-like receptor pathway-mediated promoter activity of interferon-ß (IFN-ß) as well as the transcription of downstream antiviral genes to promote viral proliferation. Mechanistically, gp110 interacts with the inhibitor of NF-κB kinase (IKKi) and restrains its K63-linked polyubiquitination, leading to attenuation of IKKi-mediated activation of NF-κB and repression of the phosphorylation and nuclear translocation of p65. Additionally, gp110 interacts with an important regulator of the Wnt signaling pathway, ß-catenin, and induces its K48-linked polyubiquitination degradation via the proteasome system, resulting in the suppression of ß-catenin-mediated IFN-ß production. Taken together, these results suggest that gp110 is a negative regulator of antiviral immunity, revealing a novel mechanism of EBV immune evasion during lytic infection. IMPORTANCE Epstein-Barr virus (EBV) is a ubiquitous pathogen that infects almost all human beings, and the persistence of EBV in the host is largely due to immune escape mediated by its encoded products. Thus, elucidation of EBV's immune escape mechanisms will provide a new direction for the design of novel antiviral strategies and vaccine development. Here, we report that EBV-encoded gp110 serves as a novel viral immune evasion factor, which inhibits RIG-I-like receptor pathway-mediated interferon-ß (IFN-ß) production. Furthermore, we found that gp110 targeted two key proteins, inhibitor of NF-κB kinase (IKKi) and ß-catenin, which mediate antiviral activity and the production of IFN-ß. gp110 inhibited K63-linked polyubiquitination of IKKi and induced ß-catenin degradation via the proteasome, resulting in decreased IFN-ß production. In summary, our data provide new insights into the EBV-mediated immune evasion surveillance strategy.


Assuntos
Infecções por Vírus Epstein-Barr , NF-kappa B , Humanos , NF-kappa B/metabolismo , Herpesvirus Humano 4/genética , Complexo de Endopeptidases do Proteassoma , beta Catenina , Interferon beta , Antivirais , Glicoproteínas
2.
J Biol Chem ; 299(5): 104613, 2023 05.
Artigo em Inglês | MEDLINE | ID: mdl-36931391

RESUMO

Epstein-Barr virus (EBV) is a member of the lymphotropic virus family and is highly correlated with some human malignant tumors. It has been reported that envelope glycoprotein 110 (gp110) plays an essential role in viral fusion, DNA replication, and nucleocapsid assembly of EBV. However, it has not been established whether gp110 is involved in regulating the host's innate immunity. In this study, we found that gp110 inhibits tumor necrosis factor α-mediated NF- κB promoter activity and the downstream production of NF- κB-regulated cytokines under physiological conditions. Using dual-luciferase reporter assays, we showed that gp110 might impede the NF-κB promoter activation downstream of NF-κB transactivational subunit p65. Subsequently, we used coimmunoprecipitation assays to demonstrate that gp110 interacts with p65 during EBV lytic infection, and that the C-terminal cytoplasmic region of gp110 is the key interaction domain with p65. Furthermore, we determined that gp110 can bind to the N-terminal Rel homologous and C-terminal domains of p65. Alternatively, gp110 might not disturb the association of p65 with nontransactivational subunit p50, but we showed it restrains activational phosphorylation (at Ser536) and nuclear translocation of p65, which we also found to be executed by the C-terminal cytoplasmic region of gp110. Altogether, these data suggest that the surface protein gp110 may be a vital component for EBV to antagonize the host's innate immune response, which is also helpful for revealing the infectivity and pathogenesis of EBV.


Assuntos
Infecções por Vírus Epstein-Barr , NF-kappa B , Humanos , NF-kappa B/genética , NF-kappa B/metabolismo , Herpesvirus Humano 4/metabolismo , Infecções por Vírus Epstein-Barr/genética , Fator de Transcrição RelA/genética , Fator de Transcrição RelA/metabolismo , Transdução de Sinais , Transporte Proteico
3.
Cell Death Dis ; 13(9): 835, 2022 09 28.
Artigo em Inglês | MEDLINE | ID: mdl-36171198

RESUMO

Intersectin 1 (ITSN1) contains two isoforms: ITSN1-S and ITSN1-L, which are highly regulated by alternative splicing. Our previous results showed that the two isoforms of ITSN1 displayed opposite functions: ITSN1-S promoted glioma development, while ITSN1-L exerted an inhibitory role in glioma progression. In this study, our transcriptome analysis using a large glioma cohort indicated that the ratio of ITSN1-S/ITSN1-L was positively correlated with glioma grading and poor prognosis. We identified the RNA-binding protein polypyrimidine tract-binding protein 1 (PTBP1) as an ITSN1 pre-mRNA interaction protein through RNA pull-down assay and RNA immunoprecipitation assay. Knockdown of PTBP1 decreased the ratio of ITSN1-S/ITSN1-L. Minigene reporter assay and mutation analyses further confirmed PTBP1 targeted polypyrimidine sequences on ITSN1 exon 30 (TTGCACTTCAGTATTTT) and promoted the inclusion of ITSN1 exon 30. Subsequently, silencing PTBP1 inhibited glioma cell proliferation, migration, and invasion by down-regulating the ratio of ITSN1-S/ITSN1-L. Taken together, our study provides a novel mechanism that PTBP1 modulates the alternative splicing of ITSN1 and promotes glioma proliferation and motility by up-regulating the ratio of ITSN1-S/ITSN1-L, thereby highlighting that PTBP1 may be an attractive therapeutic target for gliomas.


Assuntos
Processamento Alternativo , Glioma , Proteínas Adaptadoras de Transporte Vesicular , Processamento Alternativo/genética , Movimento Celular/genética , Glioma/genética , Glioma/metabolismo , Ribonucleoproteínas Nucleares Heterogêneas/genética , Ribonucleoproteínas Nucleares Heterogêneas/metabolismo , Humanos , Proteína de Ligação a Regiões Ricas em Polipirimidinas/genética , Proteína de Ligação a Regiões Ricas em Polipirimidinas/metabolismo , Isoformas de Proteínas/metabolismo , Precursores de RNA/metabolismo , Proteínas de Ligação a RNA/metabolismo
4.
J Biol Chem ; 298(10): 102471, 2022 10.
Artigo em Inglês | MEDLINE | ID: mdl-36089062

RESUMO

Zika virus (ZIKV) is a re-emerging flavivirus that causes conditions such as microcephaly and testis damage. The spread of ZIKV has become a major public health concern. Recent studies indicated that antimicrobial peptides are an ideal source for screening antiviral candidates with broad-spectrum antiviral activities, including against ZIKV. We herein found that Hc-CATH, a cathelicidin antimicrobial peptide identified from the sea snake Hydrophis cyanocinctus in our previous work, conferred protection against ZIKV infection in host cells and showed preventative efficacy and therapeutic efficacy in C57BL/6J mice, Ifnar1-/- mice, and pregnant mice. Intriguingly, we revealed that Hc-CATH decreased the susceptibility of host cells to ZIKV by downregulating expression of AXL, a TAM (TYRO3, AXL and MERTK) family kinase receptor that mediates ZIKV infection, and subsequently reversed the negative regulation of AXL on host's type I interferon response. Furthermore, we showed that the cyclo-oxygenase-2/prostaglandin E2/adenylyl cyclase/protein kinase A pathway was involved in Hc-CATH-mediated AXL downregulation, and Hc-CATH in addition directly inactivated ZIKV particles by disrupting viral membrane. Finally, while we found Hc-CATH did not act on the late stage of ZIKV infection, structure-function relationship studies revealed that α-helix and phenylalanine residues are key structural requirements for its protective efficacy against initial ZIKV infection. In summary, we demonstrate that Hc-CATH provides prophylactic and therapeutic efficacy against ZIKV infection via downregulation of AXL, as well as inactivating the virion. Our findings reveal a novel mechanism of cathelicidin against viral infection and highlight the potential of Hc-CATH to prevent and treat ZIKV infection.


Assuntos
Peptídeos Antimicrobianos , Infecção por Zika virus , Zika virus , Animais , Feminino , Masculino , Camundongos , Gravidez , Peptídeos Antimicrobianos/farmacologia , Peptídeos Antimicrobianos/uso terapêutico , Antivirais/farmacologia , Antivirais/uso terapêutico , Hydrophiidae/metabolismo , Camundongos Endogâmicos C57BL , Receptores Proteína Tirosina Quinases/genética , Receptores Proteína Tirosina Quinases/metabolismo , Internalização do Vírus , Zika virus/efeitos dos fármacos , Zika virus/metabolismo , Infecção por Zika virus/tratamento farmacológico , Infecção por Zika virus/prevenção & controle , Regulação da Expressão Gênica/efeitos dos fármacos , Catelicidinas , Receptor Tirosina Quinase Axl
6.
Front Cell Infect Microbiol ; 11: 766922, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34950606

RESUMO

Coronavirus disease 2019 (COVID-19), caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), has spread worldwide and has infected more than 250 million people. A typical feature of COVID-19 is the lack of type I interferon (IFN-I)-mediated antiviral immunity in patients. However, the detailed molecular mechanisms by which SARS-CoV-2 evades the IFN-I-mediated antiviral response remain elusive. Here, we performed a comprehensive screening and identified a set of SARS-CoV-2 proteins that antagonize the IFN-I response. Subsequently, we characterized the mechanisms of two viral proteins antagonize IFN-I production and downstream signaling. SARS-CoV-2 membrane protein binds to importin karyopherin subunit alpha-6 (KPNA6) to inhibit interferon regulatory factor 3(IRF3) nuclear translocation. Further, the spike protein interacts with signal transducer and activator of transcription 1 (STAT1) to block its association with Janus kinase 1 (JAK1). This study increases our understanding of SARS-CoV-2 pathogenesis and suggests novel therapeutic targets for the treatment of COVID-19.


Assuntos
COVID-19 , Interferon Tipo I , Glicoproteína da Espícula de Coronavírus , Proteínas da Matriz Viral , Humanos , SARS-CoV-2 , Transdução de Sinais , Proteínas Virais
7.
EMBO Rep ; 22(8): e52205, 2021 08 04.
Artigo em Inglês | MEDLINE | ID: mdl-34160127

RESUMO

The type I interferon (IFN-I, IFN-α/ß)-mediated immune response is the first line of host defense against invading viruses. IFN-α/ß binds to IFN-α/ß receptors (IFNARs) and triggers the expression of IFN-stimulated genes (ISGs). Thus, stabilization of IFNARs is important for prolonging antiviral activity. Here, we report the induction of an RNA-binding motif-containing protein, RBM47, upon viral infection or interferon stimulation. Using multiple virus infection models, we demonstrate that RBM47 has broad-spectrum antiviral activity in vitro and in vivo. RBM47 has no noticeable impact on IFN production, but significantly activates the IFN-stimulated response element (ISRE) and enhances the expression of interferon-stimulated genes (ISGs). Mechanistically, RBM47 binds to the 3'UTR of IFNAR1 mRNA, increases mRNA stability, and retards the degradation of IFNAR1. In summary, this study suggests that RBM47 is an interferon-inducible RNA-binding protein that plays an essential role in enhancing host IFN downstream signaling.


Assuntos
Antivirais , Interferon Tipo I , Antivirais/farmacologia , Interferon Tipo I/metabolismo , Interferon beta/genética , RNA Mensageiro/genética , Proteínas de Ligação a RNA/genética
8.
Antiviral Res ; 187: 105021, 2021 03.
Artigo em Inglês | MEDLINE | ID: mdl-33508330

RESUMO

Cathelicidin antimicrobial peptides (human LL-37 and mouse CRAMP) are mainly virucidal to enveloped virus. However, the effects and relative mechanisms of LL-37 and CRAMP on non-enveloped virus are elusive. We herein found that CRAMP expression was significantly up-regulated post non-enveloped Enterovirus 71 (EV71) infection in different tissues of newborn ICR mice, while EV71 replication gradually declined post CRAMP up-regulation, indicating the antiviral potential of cathelicidin against EV71. In vitro antiviral assay showed that LL-37 and CRAMP markedly reduced cytopathic effects (CPE), intracellular viral RNA copy numbers, viral VP1 protein levels, and extracellular virons in U251 cells post EV71 infection, indicating that LL-37 and CRAMP significantly inhibited EV71 replication. Mechanism of action assay showed that LL-37 and CRAMP were not virucidal to EV71, but markedly regulated antiviral immune response in U251 cells. Co-incubation of LL-37 or CRAMP with U251 cells markedly increased the basal interferon-ß (IFN-ß) expression and interferon regulatory transcription factor 3 (IRF3) phosphorylation, modestly enhanced IFN-ß production and IRF3 phosphorylation upon EV71 infection, and significantly reduced interleukin-6 (IL-6) production and p38 mitogen-activated protein kinase (MAPK) activation post EV71 infection. Additionally, LL-37 and CRAMP directly inhibited viral binding to U251 cells. Collectively, LL-37 and CRAMP markedly inhibited EV71 replication via regulating antiviral response and inhibiting viral binding, providing potent candidates for peptide drug development against EV71 infection.


Assuntos
Peptídeos Catiônicos Antimicrobianos/genética , Peptídeos Catiônicos Antimicrobianos/farmacologia , Enterovirus Humano A/efeitos dos fármacos , Imunidade , Ligação Viral/efeitos dos fármacos , Animais , Antivirais/farmacologia , Linhagem Celular , Chlorocebus aethiops , Enterovirus Humano A/genética , Infecções por Enterovirus/tratamento farmacológico , Infecções por Enterovirus/virologia , Interações entre Hospedeiro e Microrganismos , Humanos , Fator Regulador 3 de Interferon/metabolismo , Interferon beta/metabolismo , Interleucina-6/metabolismo , Camundongos , Camundongos Endogâmicos ICR , Fosforilação , RNA Viral , Regulação para Cima , Células Vero , Replicação Viral/efeitos dos fármacos , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo , Catelicidinas
9.
Sci Bull (Beijing) ; 66(3): 284-296, 2021 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-36654334

RESUMO

Dengue virus (DENV) is the most common mosquito-borne flavivirus, and it affects millions of people globally every year. Currently, there are no approved drugs for the treatment of dengue infection. By screening a natural product library, we identified a novel compound, cyclovirobuxine D (Cvb D), that displays anti-DENV activity. Cvb D inhibits DENV replication in vitro in a dose-dependent manner and protects suckling mice against lethal DENV infection. Mechanistically, Cvb D regulates the expression of genes related to the cellular cholesterol pathway. As a result, Cvb D increases cellular cholesterol synthesis and accumulation, activates mTOR, and inhibits viral-dependent autophagy. Cvb D does not suppress autophagy initiation but impedes the nuclear translocation of the lysosome transcription factor TFEB. In addition, Cvb D restricts the replication of other positive-strand RNA viruses such as Zika virus and Coxsackievirus B3. We speculate that Cvb D could be a broad-spectrum antiviral drug candidate for use against positive-strand RNA viruses that require autophagy for optimal replication.

11.
Antiviral Res ; 174: 104705, 2020 02.
Artigo em Inglês | MEDLINE | ID: mdl-31926181

RESUMO

Monocyte chemotactic protein-induced protein 1 (MCPIP1) is an inflammatory regulator in immune response. Recently, MCPIP1 has also been identified as a host antiviral factor against certain virus infection including human immunodeficiency virus, dengue virus and hepatitis C virus. However, whether MCPIP1 could restrict the replication of hepatitis B virus (HBV), a DNA pararetrovirus belonging to Hepadnaviridae family, has not been investigated. In this study, we found that MCPIP1 expression was up-regulated in mouse livers upon acute HBV replication and in HBV-replicated hepatoma cells or HBV-stimulated macrophages. Enforced MCPIP1 expression by hydrodynamic DNA injection in vivo significantly inhibited HBV replication in the mouse livers. Then in vitro studies by overexpression or knockdown assays in cell-lines identified the direct antiviral effect of MCPIP1 on HBV replication. RNA immunoprecipitation and decay assay further suggested that MCPIP1 potently restricted HBV replication through directly binding viral RNA and degrading RNA via its RNase activity, but not deubiquitinase activity. Moreover, we further verified that MCPIP1 negatively regulated HBV-induced proinflammatory cytokines, such as IL-1ß, TNF-α and IL-6 in macrophages. Taken together, our data expand MCPIP1's range of viral targets to DNA virus and also demonstrate the negative regulatory role of MCPIP1 in suppressing virus-induced inflammatory response, suggesting MCPIP1 as a potential therapeutic target for treating HBV-related diseases via inducing a host defense against HBV and reducing inflammatory injury meanwhile.


Assuntos
Interações Hospedeiro-Patógeno , Inflamação , Macrófagos/imunologia , RNA Viral/genética , Ribonucleases/genética , Replicação Viral/genética , Animais , Antivirais/metabolismo , Linhagem Celular , Feminino , Células Hep G2 , Vírus da Hepatite B/fisiologia , Humanos , Fígado/imunologia , Fígado/virologia , Macrófagos/virologia , Camundongos , Camundongos Endogâmicos BALB C , Fatores de Transcrição/genética
12.
PLoS Pathog ; 14(8): e1007287, 2018 08.
Artigo em Inglês | MEDLINE | ID: mdl-30142214

RESUMO

In order to eliminate viral infections, hundreds of interferon-stimulated genes (ISGs) are induced via type I interferons (IFNs). However, the functions and mechanisms of most ISGs are largely unclear. A tripartite motif (TRIM) protein encoding gene TRIM69 is induced by dengue virus (DENV) infection as an ISG. TRIM69 restricts DENV replication, and its RING domain, which has the E3 ubiquitin ligase activity, is critical for its antiviral activity. An in vivo study further confirmed that TRIM69 contributes to the control of DENV infection in immunocompetent mice. Unlike many other TRIM family members, TRIM69 is not involved in modulation of IFN signaling. Instead, TRIM69 interacts with DENV Nonstructural Protein 3 (NS3) directly and mediates its polyubiquitination and degradation. Finally, Lys104 of NS3 is identified as the target of TRIM69-mediated ubiquitination. Our study demonstrates that TRIM69 restricts DENV replication by specifically ubiquitinating a viral nonstructural protein.


Assuntos
Vírus da Dengue/fisiologia , Interferon Tipo I/farmacologia , Proteínas com Motivo Tripartido/fisiologia , Ubiquitina-Proteína Ligases/fisiologia , Ubiquitinação , Proteínas não Estruturais Virais/metabolismo , Replicação Viral , Células A549 , Animais , Anopheles , Células Cultivadas , Regulação da Expressão Gênica/efeitos dos fármacos , Células HEK293 , Células HeLa , Células Endoteliais da Veia Umbilical Humana , Humanos , Camundongos , Processamento de Proteína Pós-Traducional/efeitos dos fármacos , Processamento de Proteína Pós-Traducional/genética , RNA Helicases/metabolismo , Serina Endopeptidases/metabolismo , Proteínas com Motivo Tripartido/efeitos dos fármacos , Proteínas com Motivo Tripartido/genética , Ubiquitina-Proteína Ligases/efeitos dos fármacos , Ubiquitina-Proteína Ligases/genética , Ubiquitinação/efeitos dos fármacos , Ubiquitinação/genética , Regulação para Cima/efeitos dos fármacos , Regulação para Cima/genética , Replicação Viral/efeitos dos fármacos , Replicação Viral/genética
13.
Artigo em Inglês | MEDLINE | ID: mdl-28824886

RESUMO

Dengue is a mosquito-borne viral disease that rapidly spread in tropic and subtropic area in recent years. DEAD (Glu-Asp-Ala-Glu)-box RNA helicases have been reported to play important roles in viral infection, either as cytosolic sensors of viral nucleic acids or as essential host factors for the replication of different viruses. In this study, we reported that DDX25, a DEAD-box RNA helicase, plays a proviral role in DENV infection. The expression levels of DDX25 mRNA and protein were upregulated in DENV infected cells. During DENV infection, the intracellular viral loads were significantly lower in DDX25 silenced cells and higher in DDX25 overexpressed cells. Meanwhile, the expression level of type I interferon (IFN) was increased in DDX25 siRNA treated cells during viral infection. Consistent with the in vitro findings, the Ddx25-transgenic mice have an increased susceptibility to lethal vesicular stomatitis virus (VSV) virus challenge. The viremia was significantly higher while the anti-viral cytokine levels were lower in Ddx25-transgenic mice. Further, DDX25 modulated RIG-I signaling pathway and blocked IFNß production, by interrupting IFN regulatory factor 3 (IRF3) and NFκB activation. Thus, DDX25 is a novel negative regulator of IFN pathway and facilitates RNA virus infection.


Assuntos
RNA Helicases DEAD-box/metabolismo , DNA Helicases/metabolismo , Interferon Tipo I/metabolismo , Infecções por Vírus de RNA/metabolismo , Animais , Linhagem Celular , Dengue , Vírus da Dengue , Células HEK293 , Humanos , Imunidade Inata , Fator Regulador 3 de Interferon/metabolismo , Pulmão/patologia , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , NF-kappa B/metabolismo , RNA Interferente Pequeno , Carga Viral , Viroses , Replicação Viral
14.
Exp Ther Med ; 13(6): 3239-3248, 2017 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-28587396

RESUMO

Intracranial aneurysm (IA) remains one of the most devastating neurological conditions. However, the pathophysiology of IA formation and rupture still remains unclear. The purpose of the present study was to identify the crucial microRNA (miRNA/miR) and genes involved in IAs and elucidate the mechanisms underlying the development of IAs. In the present study, novel miRNA regulation activities in IAs were investigated through the integration of public gene expression data of miRNA and mRNA using the Gene Expression Omnibus database, combined with bioinformatics prediction. A total of 15 differentially expressed miRNA and 1,447 differentially expressed mRNA between IAs and controls were identified. A number of miRNA-target gene pairs (770), whose expression levels were inversely correlated, were used to construct a regulatory network of miRNA-target genes in IAs. The biological functions and pathways of these target genes were revealed to be associated with IAs. Specific miRNA and genes, such as hsa-let-7f, hsa-let-7d, hsa-miR-7, RPS6KA3, TSC1 and IGF1 may possess key roles in the development of IAs. The integrated analysis in the present study may provide insights into the understanding of underlying molecular mechanisms of IAs and novel therapeutic targets.

15.
Int J Biol Sci ; 12(7): 872-83, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27313500

RESUMO

Dengue virus (DENV) causes the most prevalent arthropod-borne viral disease of humans worldwide. Glycosphingolipids (GSLs) are involved in virus infection by regulating various steps of viral-host interaction. However, the distinct role of GSLs during DENV infection remains unclear. In this study, we used mouse melanoma B16 cells and their GSL-deficient mutant counterpart GM95 cells to study the influence of GSLs on DENV infection. Surprisingly, GM95 cells were highly resistant to DENV infection compared with B16 cells. Pretreatment of B16 cells with synthetase inhibitor of GM3, the most abundant GSLs in B16 cells, or silencing GM3 synthetase T3GAL5, significantly inhibited DENV infection. DENV attachment and endocytosis were not impaired in GM95 cells, but DENV genome replication was obviously inhibited in GM95 cells compared to B16 cells. Furthermore, GM3 was colocalized with DENV viral replication complex on endoplasmic reticulum (ER) inside the B16 cells. Finally, GM3 synthetase inhibitor significantly reduced the mortality rate of suckling mice that challenged with DENV by impairing the viral replication in mouse brain. Taken together, these data indicated that GM3 was not required for DENV attachment and endocytosis, however, essential for viral genome replication. Targeting GM3 could be a novel strategy to inhibit DENV infection.


Assuntos
Vírus da Dengue/genética , Vírus da Dengue/metabolismo , Genoma Viral/genética , Glicoesfingolipídeos/metabolismo , Replicação Viral/fisiologia , Animais , Linhagem Celular Tumoral , Vírus da Dengue/fisiologia , Flavivirus/genética , Flavivirus/metabolismo , Flavivirus/fisiologia , Glicolipídeos/metabolismo , Camundongos , Proteínas não Estruturais Virais/genética , Proteínas não Estruturais Virais/metabolismo , Replicação Viral/genética
16.
Viral Immunol ; 28(8): 448-54, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-26196448

RESUMO

Hantaan virus A9 strain (HTNV A9) is an etiologic agent of hemorrhagic fever with renal syndrome in China. The virulence of the pathogenic hantaviruses is determined by their ability to alter key signaling pathways of early interferon (IFN) induction within cells. The potential role of HTNV A9 structural proteins, such as nucleocapsid (N) and envelope glycoproteins (Gn and Gc), in regulating human's innate antiviral immune response has not yet been clarified. In this study, we investigated the effect of HTNV A9 N protein on the regulation of the IFN pathway. We found that A9 N protein can influence the host innate immune response by regulating the activation of IFNß. The A9 N protein stimulates IFN response in low doses, whereas significantly inhibits IFNß production at high doses. Furthermore, A9 N protein constitutively inhibits nuclear factor kappa B activation. A high dose of A9 N protein could inhibit either Poly IC-induced IFNß or vesicular stomatitis virus-induced IFNß and interferon-stimulated gene production. Our results indicate that HTNV A9 N protein helps virus establish successful infection by downregulating the IFN response and shed new light to the understanding of the interaction between the host innate immunity and virus during Hantaan virus infection.


Assuntos
Vírus Hantaan/imunologia , Interações Hospedeiro-Patógeno , Evasão da Resposta Imune , Imunidade Inata , Interferon beta/metabolismo , Proteínas do Nucleocapsídeo/imunologia , Transdução de Sinais , Animais , China , Chlorocebus aethiops , Regulação para Baixo , Humanos , Interferon beta/antagonistas & inibidores , NF-kappa B/antagonistas & inibidores , Células Vero
17.
J Virol ; 88(20): 12163-6, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-25078699

RESUMO

The interferon (IFN)-inducible viperin protein restricts a broad range of viruses. However, whether viperin plays a role during herpes simplex virus 1 (HSV-1) infection is poorly understood. In the present study, it was shown for the first time that wild-type (WT) HSV-1 infection couldn't induce viperin production, and ectopically expressed viperin inhibited the replication of UL41-null HSV-1 but not WT viruses. The underlying molecular mechanism is that UL41 counteracts viperin's antiviral activity by reducing its mRNA accumulation.


Assuntos
Herpesvirus Humano 1/fisiologia , Proteínas/antagonistas & inibidores , Proteínas Virais/fisiologia , Células HEK293 , Humanos , Oxirredutases atuantes sobre Doadores de Grupo CH-CH , Proteínas/fisiologia , Replicação Viral/fisiologia
18.
J Virol ; 88(14): 7941-51, 2014 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-24807716

RESUMO

Nuclear factor κB (NF-κB) plays important roles in innate immune responses by regulating the expression of a large number of target genes involved in the immune and inflammatory response, apoptosis, cell proliferation, differentiation, and survival. To survive in the host cells, viruses have evolved multiple strategies to evade and subvert the host immune response. Herpes simplex virus 1 (HSV-1) bears a large DNA genome, with the capacity to encode many different viral proteins to counteract the host immune responses. In the present study, we demonstrated that HSV-1 protein kinase US3 significantly inhibited NF-κB activation and decreased the expression of inflammatory chemokine interleukin-8 (IL-8). US3 was also shown to hyperphosphorylate p65 at serine 75 and block its nuclear translocation. Two US3 mutants, K220M and D305A, still interacted with p65; however, they could not hyperphosphorylate p65, indicating that the kinase activity of US3 was indispensable for the function. The attenuation of NF-κB activation by HSV-1 US3 protein kinase may represent a critical adaptation to enable virus persistence within the host. Importance: This study demonstrated that HSV-1 protein kinase US3 significantly inhibited NF-κB activation and decreased the expression of inflammatory chemokine interleukin-8 (IL-8). US3 hyperphosphorylated p65 at serine 75 to inhibit NF-κB activation. The kinase activity of US3 was indispensable for its hyperphosphorylation of p65 and abrogation of the nuclear translocation of p65. The present study elaborated a novel mechanism of HSV-1 US3 to evade the host innate immunity.


Assuntos
Herpesvirus Humano 1/fisiologia , Interações Hospedeiro-Patógeno , Evasão da Resposta Imune , Processamento de Proteína Pós-Traducional , Proteínas Serina-Treonina Quinases/metabolismo , Fator de Transcrição RelA/antagonistas & inibidores , Fator de Transcrição RelA/metabolismo , Proteínas Virais/metabolismo , Animais , Linhagem Celular , Regulação para Baixo , Humanos , Interleucina-8/biossíntese , Proteínas Mutantes/genética , Proteínas Mutantes/metabolismo , Mutação de Sentido Incorreto , Fosforilação , Ligação Proteica , Proteínas Serina-Treonina Quinases/genética , Proteínas Virais/genética
19.
J Virol ; 87(23): 12814-27, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-24049179

RESUMO

Viral infection initiates a series of signaling cascades that lead to the transcription of interferons (IFNs), finally inducing interferon-stimulated genes (ISGs) to eliminate viruses. Viruses have evolved a variety of strategies to modulate host IFN-mediated immune responses. Herpes simplex virus 1 (HSV-1) US3, a Ser/Thr kinase conserved in alphaherpesviruses, was previously reported to counteract host innate immunity; however, the molecular mechanism is elusive. In this study, we report that US3 blocks IFN-ß production by hyperphosphorylating IFN regulatory factor 3 (IRF3). Ectopic expression of US3 protein significantly inhibited Sendai virus (SeV)-mediated activation of IFN-ß and IFN-stimulated response element (ISRE) promoters and the transcription of IFN-ß, ISG54, and ISG56. US3 was also shown to block SeV-induced dimerization and nuclear translocation of IRF3. The kinase activity was indispensable for its inhibitory function, as kinase-dead (KD) US3 mutants K220M and D305A could not inhibit IFN-ß production. Furthermore, US3 interacted with and hyperphosphorylated IRF3 at Ser175 to prevent IRF3 activation. Finally, the US3 KD mutant viruses were constructed and denoted K220M or D305A HSV-1, respectively. Cells and mice infected with both mutant viruses produced remarkably larger amounts of IFN-ß than those infected with wild-type HSV-1. For the first time, these findings provide convincing evidence that US3 hyperphosphorylates IRF3, blocks the production of IFN-ß, and subverts host innate immunity.


Assuntos
Regulação para Baixo , Herpes Simples/genética , Herpes Simples/metabolismo , Herpesvirus Humano 1/enzimologia , Fator Regulador 3 de Interferon/metabolismo , Interferon beta/genética , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Virais/metabolismo , Motivos de Aminoácidos , Animais , Feminino , Herpes Simples/virologia , Herpesvirus Humano 1/genética , Humanos , Fator Regulador 3 de Interferon/química , Fator Regulador 3 de Interferon/genética , Interferon beta/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Fosforilação , Regiões Promotoras Genéticas , Ligação Proteica , Proteínas Serina-Treonina Quinases/genética , Proteínas Virais/genética
20.
J Virol ; 87(23): 12935-48, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-24067962

RESUMO

NF-κB plays central roles in regulation of diverse biological processes, including innate and adaptive immunity and inflammation. HSV-1 is the archetypal member of the alphaherpesviruses, with a large genome encoding over 80 viral proteins, many of which are involved in virus-host interactions and show immune modulatory capabilities. In this study, we demonstrated that the HSV-1 ICP0 protein, a viral E3 ubiquitin ligase, was shown to significantly suppress tumor necrosis factor alpha (TNF-α)-mediated NF-κB activation. ICP0 was demonstrated to bind to the NF-κB subunits p65 and p50 by coimmunoprecipitation analysis. ICP0 bound to the Rel homology domain (RHD) of p65. Fluorescence microscopy demonstrated that ICP0 abolished nuclear translocation of p65 upon TNF-α stimulation. Also, ICP0 degraded p50 via its E3 ubiquitin ligase activity. The RING finger (RF) domain mutant ICP0 (ICP0-RF) lost its ability to inhibit TNF-α-mediated NF-κB activation and p65 nuclear translocation and degrade p50. Notably, the RF domain of ICP0 was sufficient to interact with p50 and abolish NF-κB reporter gene activity. Here, it is for the first time shown that HSV-1 ICP0 interacts with p65 and p50, degrades p50 through the ubiquitin-proteasome pathway, and prevents NF-κB-dependent gene expression, which may contribute to immune evasion and pathogenesis of HSV-1.


Assuntos
Herpes Simples/metabolismo , Herpesvirus Humano 1/enzimologia , Proteínas Imediatamente Precoces/metabolismo , Subunidade p50 de NF-kappa B/metabolismo , NF-kappa B/genética , Fator de Transcrição RelA/metabolismo , Ativação Transcricional , Fator de Necrose Tumoral alfa/metabolismo , Ubiquitina-Proteína Ligases/metabolismo , Núcleo Celular/genética , Núcleo Celular/metabolismo , Células HEK293 , Herpes Simples/genética , Herpes Simples/virologia , Herpesvirus Humano 1/química , Herpesvirus Humano 1/genética , Humanos , Proteínas Imediatamente Precoces/química , Proteínas Imediatamente Precoces/genética , NF-kappa B/metabolismo , Subunidade p50 de NF-kappa B/genética , Ligação Proteica , Estrutura Terciária de Proteína , Transporte Proteico , Fator de Transcrição RelA/genética , Fator de Necrose Tumoral alfa/genética , Ubiquitina-Proteína Ligases/química , Ubiquitina-Proteína Ligases/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA