Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 6 de 6
Filtrar
Más filtros

Banco de datos
Tipo de estudio
Tipo del documento
Asunto de la revista
País de afiliación
Intervalo de año de publicación
1.
J Virol ; 94(12)2020 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-32238587

RESUMEN

Cyclic GMP-AMP synthase (cGAS) senses double-stranded DNA and synthesizes the second messenger cyclic GMP-AMP (cGAMP), which binds to mediator of IRF3 activation (MITA) and initiates MITA-mediated signaling, leading to induction of type I interferons (IFNs) and other antiviral effectors. Human cytomegalovirus (HCMV), a widespread and opportunistic pathogen, antagonizes the host antiviral immune response to establish latent infection. Here, we identified HCMV tegument protein UL94 as an inhibitor of the cGAS-MITA-mediated antiviral response. Ectopic expression of UL94 impaired cytosolic double-stranded DNA (dsDNA)- and DNA virus-triggered induction of type I IFNs and enhanced viral replication. Conversely, UL94 deficiency potentiated HCMV-induced transcription of type I IFNs and downstream antiviral effectors and impaired viral replication. UL94 interacted with MITA, disrupted the dimerization and translocation of MITA, and impaired the recruitment of TBK1 to the MITA signalsome. These results suggest that UL94 plays an important role in the immune evasion of HCMV.IMPORTANCE Human cytomegalovirus (HCMV), a large double-stranded DNA (dsDNA) virus, encodes more than 200 viral proteins. HCMV infection causes irreversible abnormalities of the central nervous system in newborns and severe syndromes in organ transplantation patients or AIDS patients. It has been demonstrated that HCMV has evolved multiple immune evasion strategies to establish latent infection. Previous studies pay more attention to the mechanism by which HCMV evades immune response in the early phase of infection. In this study, we identified UL94 as a negative regulator of the innate immune response, which functions in the late phase of HCMV infection.


Asunto(s)
Proteínas de la Cápside/inmunología , Citomegalovirus/inmunología , Genoma Viral , Evasión Inmune , Proteínas de la Membrana/inmunología , Proteínas Serina-Treonina Quinasas/inmunología , ARN Interferente Pequeño/genética , Proteínas de la Cápside/genética , Núcleo Celular/inmunología , Núcleo Celular/virología , GMP Cíclico/inmunología , GMP Cíclico/metabolismo , Citomegalovirus/genética , Citomegalovirus/crecimiento & desarrollo , Citosol/inmunología , Citosol/virología , ADN/inmunología , ADN/metabolismo , Fibroblastos/inmunología , Fibroblastos/virología , Regulación de la Expresión Génica , Células HEK293 , Humanos , Inmunidad Innata , Factor 3 Regulador del Interferón/genética , Factor 3 Regulador del Interferón/inmunología , Proteínas de la Membrana/genética , Cultivo Primario de Células , Unión Proteica , Multimerización de Proteína , Proteínas Serina-Treonina Quinasas/genética , Transporte de Proteínas , ARN Interferente Pequeño/inmunología , Transducción de Señal , Secuenciación del Exoma
2.
PLoS Pathog ; 13(4): e1006328, 2017 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-28414768

RESUMEN

Upon viral infection, retinoic acid-inducible gene I-like receptors (RLRs) recognize viral RNA and trigger a series of signaling events, leading to the induction of type I interferons (IFNs). These processes are delicately regulated to prevent excessive and harmful immune responses. In this study, we identified G patch domain-containing protein 3 (GPATCH3) as a negative regulator of RLR-mediated antiviral signaling pathways. Overexpression of GPATCH3 impaired RNA virus- triggered induction of downstream antiviral genes, whereas its knockdown had opposite effects and attenuated viral replication. In addition, GPATCH3-deficient cells had higher IFNB1 mRNA level compared with control cells after RNA virus infection. Mechanistically, GPATCH3 was recruited to VISA in a viral infection dependent manner and the assembly of VISA/TRAF6/TBK1 signalosome was impaired in GPATCH3-overexpressing cells. In contrast, upon viral infection, the recruitment of TRAF6 and TBK1 to VISA was enhanced in GPATCH3 deficient cells. Taking together, our findings demonstrate that GPATCH3 interacts with VISA and disrupts the assembly of virus-induced VISA signalosome therefore acts as a negative regulator of RLR-mediated innate antiviral immune responses.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/inmunología , Proteínas Portadoras/inmunología , Helicasa Inducida por Interferón IFIH1/inmunología , Receptores de Ácido Retinoico/inmunología , Virosis/inmunología , Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas Portadoras/genética , Línea Celular , Humanos , Interferón Tipo I/genética , Interferón Tipo I/inmunología , Helicasa Inducida por Interferón IFIH1/genética , Mitocondrias/genética , Mitocondrias/inmunología , Unión Proteica , Receptores de Ácido Retinoico/genética , Transducción de Señal , Virosis/genética , Virosis/virología
3.
Proc Natl Acad Sci U S A ; 111(4): 1509-14, 2014 Jan 28.
Artículo en Inglés | MEDLINE | ID: mdl-24434549

RESUMEN

TNFα and IL-1ß are two proinflammatory cytokines that play critical roles in many diseases, including rheumatoid arthritis and infectious diseases. How TNFα- and IL-1ß-mediated signaling is finely tuned is not fully elucidated. Here, we identify tripartite-motif protein 38 (TRIM38) as a critical negative regulator of TNFα- and IL-1ß-triggered signaling. Overexpression of TRIM38 inhibited activation of NF-κB and induction of downstream cytokines following TNFα and IL-1ß stimulation, whereas knockdown or knockout of TRIM38 had the opposite effects. TRIM38 constitutively interacted with critical components TGF-ß-activated kinase 1 (TAK1)-binding protein 2/3 (TAB2/3) and promoted lysosome-dependent degradation of TAB2/3 independent of its E3 ubiquitin ligase activity. Consistently, deficiency of TRIM38 resulted in abolished translocation of TAB2 to the lysosome, increased level of TAB2 in cells, and enhanced activation of TAK1 after TNFα and IL-1ß stimulation. We conclude that TRIM38 negatively regulates TNFα- and IL-1ß-induced signaling by mediating lysosome-dependent degradation of TAB2/3, two critical components in TNFα- and IL-1ß-induced signaling pathways. Our findings reveal a previously undiscovered mechanism by which cells keep the inflammatory response in check to avoid excessive harmful immune response triggered by TNFα and IL-1ß.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Proteínas Portadoras/fisiología , Interleucina-1beta/fisiología , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Lisosomas/metabolismo , FN-kappa B/metabolismo , Factor de Necrosis Tumoral alfa/fisiología , Secuencia de Bases , Proteínas Portadoras/genética , Cartilla de ADN , Humanos , Proteolisis , Interferencia de ARN , Transducción de Señal/fisiología , Proteínas de Motivos Tripartitos , Ubiquitina-Proteína Ligasas
4.
Cell Host Microbe ; 24(1): 69-80.e4, 2018 07 11.
Artículo en Inglés | MEDLINE | ID: mdl-29937271

RESUMEN

The cytosolic DNA sensor cGAS recognizes viral DNA and synthesizes the second messenger cGAMP upon viral infection. cGAMP binds to the adaptor protein MITA/STING to activate downstream signaling events, leading to induction of type I interferons (IFNs) and antiviral effector genes. Here we identify the human cytomegalovirus (HCMV) protein UL31 as an inhibitor of cGAS. UL31 interacts directly with cGAS and disassociates DNA from cGAS, thus inhibiting cGAS enzymatic functions and reducing cGAMP production. UL31 overexpression markedly reduces antiviral responses stimulated by cytosolic DNA, while knockdown or knockout of UL31 heightens HCMV-triggered induction of type I IFNs and downstream antiviral genes. Moreover, wild-type HCMV replicates more efficiently than UL31-deficient HCMV, a phenotype that is reversed in cGAS null cells. These results highlight the importance of cGAS in the host response to HCMV as well as an important viral strategy to evade this innate immune sensor.


Asunto(s)
Citomegalovirus/fisiología , Evasión Inmune/inmunología , Proteínas Nucleares/metabolismo , Nucleotidiltransferasas/antagonistas & inhibidores , Proteínas Virales/metabolismo , Citomegalovirus/genética , ADN Viral/genética , ADN Viral/metabolismo , Fibroblastos , Técnicas de Silenciamiento del Gen , Técnicas de Inactivación de Genes , Células HEK293 , Humanos , Inmunidad Innata/inmunología , Interferón Tipo I/metabolismo , Proteínas Nucleares/genética , Nucleótidos Cíclicos/metabolismo , Nucleotidiltransferasas/genética , Cultivo Primario de Células , Proteínas Virales/genética
5.
Cell Host Microbe ; 21(2): 231-243, 2017 Feb 08.
Artículo en Inglés | MEDLINE | ID: mdl-28132838

RESUMEN

Recognition of human cytomegalovirus (HCMV) DNA by the cytosolic sensor cGAS initiates STING-dependent innate antiviral responses. HCMV can antagonize host immune responses to promote latency infection. However, it is unknown whether and how HCMV targets the cGAS-STING axis for immune evasion. Here we identified the HCMV tegument protein UL82 as a negative regulator of STING-dependent antiviral responses. UL82 interacted with STING and impaired STING-mediated signaling via two mechanisms. UL82 inhibited the translocation of STING from the ER to perinuclear microsomes by disrupting the STING-iRhom2-TRAPß translocation complex. UL82 also impaired the recruitment of TBK1 and IRF3 to the STING complex. The levels of downstream antiviral genes induced by UL82-deficient HCMV were higher than those induced by wild-type HCMV. Conversely, wild-type HCMV replicated more efficiently than the UL82-deficient mutant. These findings reveal an important mechanism of immune evasion by HCMV.


Asunto(s)
Citomegalovirus/genética , Regulación Viral de la Expresión Génica , Evasión Inmune , Proteínas de la Membrana/metabolismo , Proteínas Virales/metabolismo , Citomegalovirus/fisiología , Humanos , Inmunidad Innata , Factor 3 Regulador del Interferón/genética , Factor 3 Regulador del Interferón/metabolismo , Proteínas de la Membrana/genética , Proteínas Serina-Treonina Quinasas/genética , Proteínas Serina-Treonina Quinasas/metabolismo , Transporte de Proteínas , Transducción de Señal , Proteínas Virales/genética , Replicación Viral
6.
Cell Res ; 26(3): 288-303, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26823206

RESUMEN

Recognition of viral dsRNA by Toll-like receptor 3 (TLR3) leads to induction of interferons (IFNs) and proinflammatory cytokines, and innate antiviral response. Here we identified the RNA-binding protein Mex3B as a positive regulator of TLR3-mediated signaling by expression cloning screens. Cells from Mex3b(-/-) mice exhibited reduced production of IFN-ß in response to the dsRNA analog poly(I:C) but not infection with RNA viruses. Mex3b(-/-) mice injected with poly(I:C) was more resistant to poly(I:C)-induced death. Mex3B was associated with TLR3 in the endosomes. It bound to dsRNA and increased the dsRNA-binding activity of TLR3. Mex3B also promoted the proteolytic processing of TLR3, which is critical for its activation. Mutants of Mex3B lacking its RNA-binding activity inhibited TLR3-mediated IFN-ß induction. These findings suggest that Mex3B acts as a coreceptor of TLR3 in innate antiviral response.


Asunto(s)
Inmunidad Innata , Proteínas de Unión al ARN/fisiología , Receptor Toll-Like 3/metabolismo , Proteínas Adaptadoras del Transporte Vesicular/metabolismo , Animales , Células Cultivadas , Endosomas/metabolismo , Femenino , Masculino , Ratones Noqueados , Poli I-C/farmacología , Virus ARN/fisiología , ARN Bicatenario/metabolismo , Proteínas de Unión al ARN/genética , Proteínas de Unión al ARN/metabolismo , Transducción de Señal
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA