Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
1.
J Virol ; 97(11): e0079523, 2023 Nov 30.
Artículo en Inglés | MEDLINE | ID: mdl-37902401

RESUMEN

IMPORTANCE: African swine fever virus (ASFV), the only known DNA arbovirus, is the causative agent of African swine fever (ASF), an acutely contagious disease in pigs. ASF has recently become a crisis in the pig industry in recent years, but there are no commercially available vaccines. Studying the immune evasion mechanisms of ASFV proteins is important for the understanding the pathogenesis of ASFV and essential information for the development of an effective live-attenuated ASFV vaccines. Here, we identified ASFV B175L, previously uncharacterized proteins that inhibit type I interferon signaling by targeting STING and 2'3'-cGAMP. The conserved B175L-zf-FCS motif specifically interacted with both cGAMP and the R238 and Y240 amino acids of STING. Consequently, this interaction interferes with the interaction of cGAMP and STING, thereby inhibiting downstream signaling of IFN-mediated antiviral responses. This novel mechanism of B175L opens a new avenue as one of the ASFV virulent genes that can contribute to the advancement of ASFV live-attenuated vaccines.


Asunto(s)
Virus de la Fiebre Porcina Africana , Fiebre Porcina Africana , Interferón Tipo I , Proteínas de la Membrana , Nucleótidos Cíclicos , Porcinos , Proteínas Virales , Animales , Fiebre Porcina Africana/inmunología , Fiebre Porcina Africana/virología , Virus de la Fiebre Porcina Africana/química , Virus de la Fiebre Porcina Africana/genética , Virus de la Fiebre Porcina Africana/inmunología , Virus de la Fiebre Porcina Africana/patogenicidad , Interferón Tipo I/antagonistas & inhibidores , Interferón Tipo I/inmunología , Proteínas de la Membrana/antagonistas & inhibidores , Proteínas de la Membrana/química , Proteínas de la Membrana/metabolismo , Nucleótidos Cíclicos/antagonistas & inhibidores , Nucleótidos Cíclicos/metabolismo , Porcinos/inmunología , Porcinos/virología , Vacunas Atenuadas/inmunología , Proteínas Virales/metabolismo , Vacunas Virales/inmunología , Interacciones Microbiota-Huesped
2.
J Med Virol ; 96(3): e29523, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-38483060

RESUMEN

Tight control of the type I interferon (IFN) signaling pathway is critical for maintaining host innate immune responses, and the ubiquitination and deubiquitination of signaling molecules are essential for signal transduction. Deubiquitinase ubiquitin-specific protein 19 (USP19) is known to be involved in deubiquitinating Beclin1, TRAF3, and TRIF for downregulation of the type I IFN signaling. Here, we show that SIAH1, a cellular E3 ubiquitin ligase that is involved in multicellular pathway, is a potent positive regulator of virus-mediated type I IFN signaling that maintains homeostasis within the antiviral immune response by targeting USP19. In the early stages of virus infection, stabilized SIAH1 directly interacts with the USP19 and simultaneously mediates K27-linked ubiquitination of 489, 490, and 610 residues of USP19 for proteasomal degradation. Additionally, we found that USP19 specifically interacts with MAVS and deubiquitinates K63-linked ubiquitinated MAVS for negative regulation of type I IFN signaling. Ultimately, we identified that SIAH1-mediated degradation of USP19 reversed USP19-mediated deubiquitination of MAVS, Beclin1, TRAF3, and TRIF, resulting in the activation of antiviral immune responses. Taken together, these findings provide new insights into the molecular mechanism of USP19 and SIAH1, and suggest a critical role of SIAH1 in antiviral immune response and homeostasis.


Asunto(s)
Interferón Tipo I , Ubiquitina , Humanos , Ubiquitina/metabolismo , Factor 3 Asociado a Receptor de TNF/genética , Beclina-1 , Ubiquitinación , Inmunidad Innata , Interferón Tipo I/metabolismo , Enzimas Desubicuitinizantes/genética , Enzimas Desubicuitinizantes/metabolismo , Proteínas Adaptadoras del Transporte Vesicular , Endopeptidasas/genética , Endopeptidasas/metabolismo
3.
Int J Mol Sci ; 25(4)2024 Feb 08.
Artículo en Inglés | MEDLINE | ID: mdl-38396775

RESUMEN

DP96R of African swine fever virus (ASFV), also known as uridine kinase (UK), encodes a virulence-associated protein. Previous studies have examined DP96R along with other genes in an effort to create live attenuated vaccines. While experiments in pigs have explored the impact of DP96R on the pathogenicity of ASFV, the precise molecular mechanism underlying this phenomenon remains unknown. Here, we describe a novel molecular mechanism by which DP96R suppresses interferon regulator factor-3 (IRF3)-mediated antiviral immune responses. DP96R interacts with a crucial karyopherin (KPNA) binding site within IRF3, disrupting the KPNA-IRF3 interaction and consequently impeding the translocation of IRF3 to the nucleus. Under this mechanistic basis, the ectopic expression of DP96R enhances the replication of DNA and RNA viruses by inhibiting the production of IFNs, whereas DP96R knock-down resulted in higher IFNs and IFN-stimulated gene (ISG) transcription during ASFV infection. Collectively, these findings underscore the pivotal role of DP96R in inhibiting IFN responses and increase our understanding of the relationship between DP96R and the virulence of ASFV.


Asunto(s)
Virus de la Fiebre Porcina Africana , Factor 3 Regulador del Interferón , Animales , Virus de la Fiebre Porcina Africana/genética , Virus de la Fiebre Porcina Africana/patogenicidad , Interferones/metabolismo , Porcinos , Proteínas Virales/metabolismo , Virulencia , Factores de Virulencia/genética , Factor 3 Regulador del Interferón/metabolismo , Humanos , Interferón Tipo I/metabolismo
4.
J Virol ; 96(15): e0102222, 2022 08 10.
Artículo en Inglés | MEDLINE | ID: mdl-35861515

RESUMEN

African swine fever virus (ASFV) is a highly pathogenic swine DNA virus with high mortality that causes African swine fever (ASF) in domestic pigs and wild boars. For efficient viral infection, ASFV has developed complex strategies to evade key components of antiviral innate immune responses. However, the immune escape mechanism of ASFV remains unclear. Upon ASFV infection, cyclic GMP-AMP (2',3'-cGAMP) synthase (cGAS), a cytosolic DNA sensor, recognizes ASFV DNA and synthesizes the second messenger 2',3'-cGAMP, which triggers interferon (IFN) production to interfere with viral replication. In this study, we demonstrated a novel immune evasion mechanism of ASFV EP364R and C129R, which blocks cellular cyclic 2',3'-cGAMP-mediated antiviral responses. ASFV EP364R and C129R with nuclease homology inhibit IFN-mediated responses by specifically interacting with 2',3'-cGAMP and exerting their phosphodiesterase (PDE) activity to cleave 2',3'-cGAMP. Particularly notable is that ASFV EP364R had a region of homology with the stimulator of interferon genes (STING) protein containing a 2',3'-cGAMP-binding motif and point mutations in the Y76S and N78A amino acids of EP364R that impaired interaction with 2',3'-cGAMP and restored subsequent antiviral responses. These results highlight a critical role for ASFV EP364R and C129R in the inhibition of IFN responses and could be used to develop ASFV live attenuated vaccines. IMPORTANCE African swine fever (ASF) is a highly contagious hemorrhagic disease in domestic pigs and wild boars caused by African swine fever virus (ASFV). ASF is a deadly epidemic disease in the global pig industry, but no drugs or vaccines are available. Understanding the pathogenesis of ASFV is essential to developing an effective live attenuated ASFV vaccine, and investigating the immune evasion mechanisms of ASFV is crucial to improve the understanding of its pathogenesis. In this study, for the first time, we identified the EP364R and C129R, uncharacterized proteins that inhibit type I interferon signaling. ASFV EP364R and C129R specifically interacted with 2',3'-cGAMP, the mammalian second messenger, and exerted phosphodiesterase activity to cleave 2',3'-cGAMP. In this study, we discovered a novel mechanism by which ASFV inhibits IFN-mediated antiviral responses, and our findings can guide the understanding of ASFV pathogenesis and the development of live attenuated ASFV vaccines.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales , Virus de la Fiebre Porcina Africana , Evasión Inmune , Proteínas de la Membrana , Nucleótidos Cíclicos , Nucleotidiltransferasas , Transducción de Señal , Proteínas Virales , Fiebre Porcina Africana/virología , Virus de la Fiebre Porcina Africana/inmunología , Virus de la Fiebre Porcina Africana/metabolismo , Animales , Interferones/antagonistas & inhibidores , Interferones/inmunología , Proteínas de la Membrana/antagonistas & inhibidores , Proteínas de la Membrana/metabolismo , Nucleótidos Cíclicos/inmunología , Nucleótidos Cíclicos/metabolismo , Nucleotidiltransferasas/antagonistas & inhibidores , Nucleotidiltransferasas/metabolismo , Hidrolasas Diéster Fosfóricas/metabolismo , Sus scrofa/virología , Porcinos , Vacunas Atenuadas , Proteínas Virales/metabolismo , Vacunas Virales
5.
Cell Rep ; 42(11): 113358, 2023 11 28.
Artículo en Inglés | MEDLINE | ID: mdl-37917584

RESUMEN

Stress granules (SGs) constitute a signaling hub that plays a critical role in type I interferon responses. Here, we report that growth arrest and DNA damage-inducible beta (Gadd45ß) act as a positive regulator of SG-mediated interferon signaling by targeting G3BP upon RNA virus infection. Gadd45ß deficiency markedly impairs SG formation and SG-mediated activation of interferon signaling in vitro. Gadd45ß knockout mice are highly susceptible to RNA virus infection, and their ability to produce interferon and cytokines is severely impaired. Specifically, Gadd45ß interacts with the RNA-binding domain of G3BP, leading to conformational expansion of G3BP1 via dissolution of its autoinhibitory electrostatic intramolecular interaction. The acidic loop 1- and RNA-binding properties of Gadd45ß markedly increase the conformational expansion and RNA-binding affinity of the G3BP1-Gadd45ß complex, thereby promoting assembly of SGs. These findings suggest a role for Gadd45ß as a component and critical regulator of G3BP1-mediated SG formation, which facilitates RLR-mediated interferon signaling.


Asunto(s)
Interferón Tipo I , Infecciones por Virus ARN , Animales , Ratones , Gránulos Citoplasmáticos/metabolismo , ADN Helicasas/metabolismo , Proteínas de Unión a Poli-ADP-Ribosa/genética , ARN , ARN Helicasas/metabolismo , Proteínas con Motivos de Reconocimiento de ARN/genética , Gránulos de Estrés
6.
Viruses ; 14(9)2022 09 16.
Artículo en Inglés | MEDLINE | ID: mdl-36146866

RESUMEN

Foot-and-mouth disease (FMD) is mainly characterized by blister formation (vesicles) in animals infected with foot-and-mouth disease virus (FMDV). However, the molecular basis of the blister formation in FMD is still unknown. BP180 is one of the main anchoring proteins connecting the dermal and epidermal layers of the skin. Previous studies have shown that the cleavage of BP180 by proteases produced by the inflammatory cells and the resulting skin loosening are major causes of the blister formation in bullous pemphigoid (BP) disease. Similar to BP, here we have demonstrated that, among the FMDV-encoded proteases, only FMDV 3Cpro contributes to the cleavage of BP180 at multiple sites, consequently inducing the degradation of BP180, leading to skin loosening. Additionally, we confirmed that FMDV 3Cpro interacts directly with BP180 and the FMDV 3Cpro C142T mutant, known to have reduced protease activity, is less effective for BP180 degradation than wild-type FMDV 3Cpro. In conclusion, for the first time, our results demonstrate the function of FMDV 3Cpro on the connective-tissue protein BP180 associated with blister formation.


Asunto(s)
Virus de la Fiebre Aftosa , Fiebre Aftosa , Animales , Vesícula , Cisteína Endopeptidasas/metabolismo , Virus de la Fiebre Aftosa/metabolismo , Péptido Hidrolasas , Proteínas Virales/genética , Proteínas Virales/metabolismo
7.
Front Immunol ; 13: 1020262, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36248821

RESUMEN

Foot-and-mouth disease virus (FMDV) is a single-stranded, positive-sense RNA virus containing at least 13 proteins. Many of these proteins show immune modulation capabilities. As a non-structural protein of the FMDV, 2B is involved in the rearrangement of the host cell membranes and the disruption of the host secretory pathway as a viroporin. Previous studies have also shown that FMDV 2B plays a role in the modulation of host type-I interferon (IFN) responses through the inhibition of expression of RIG-I and MDA5, key cytosolic sensors of the type-I IFN signaling. However, the exact molecular mechanism is poorly understood. Here, we demonstrated that FMDV 2B modulates host IFN signal pathway by the degradation of RIG-I and MDA5. FMDV 2B targeted the RIG-I for ubiquitination and proteasomal degradation by recruiting E3 ubiquitin ligase ring finger protein 125 (RNF125) and also targeted MDA5 for apoptosis-induced caspase-3- and caspase-8-dependent degradation. Ultimately, FMDV 2B significantly inhibited RNA virus-induced IFN-ß production. Importantly, we identified that the C-terminal amino acids 126-154 of FMDV 2B are essential for 2B-mediated degradation of the RIG-I and MDA5. Collectively, these results provide a clearer understanding of the specific molecular mechanisms used by FMDV 2B to inhibit the IFN responses and a rational approach to virus attenuation for future vaccine development.


Asunto(s)
Virus de la Fiebre Aftosa , Interferón Tipo I , Aminoácidos/metabolismo , Animales , Caspasa 3/metabolismo , Caspasa 8/metabolismo , Interferón Tipo I/metabolismo , Transducción de Señal , Ubiquitina-Proteína Ligasas/metabolismo , Proteínas Viroporinas
8.
Exp Mol Med ; 53(11): 1647-1668, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34782737

RESUMEN

A harmonized balance between positive and negative regulation of pattern recognition receptor (PRR)-initiated immune responses is required to achieve the most favorable outcome for the host. This balance is crucial because it must not only ensure activation of the first line of defense against viral infection but also prevent inappropriate immune activation, which results in autoimmune diseases. Recent studies have shown how signal transduction pathways initiated by PRRs are positively and negatively regulated by diverse modulators to maintain host immune homeostasis. However, viruses have developed strategies to subvert the host antiviral response and establish infection. Viruses have evolved numerous genes encoding immunomodulatory proteins that antagonize the host immune system. This review focuses on the current state of knowledge regarding key host factors that regulate innate immune signaling molecules upon viral infection and discusses evidence showing how specific viral proteins counteract antiviral responses via immunomodulatory strategies.


Asunto(s)
Genoma Viral , Interacciones Huésped-Patógeno/inmunología , Evasión Inmune , Inmunidad Innata , Transducción de Señal , Virosis/etiología , Virosis/metabolismo , Animales , Biomarcadores , Virus ADN/genética , Virus ADN/inmunología , Resistencia a la Enfermedad , Susceptibilidad a Enfermedades/inmunología , Genoma Viral/inmunología , Humanos , Quinasa I-kappa B/metabolismo , Sistema Inmunológico , Factor 3 Regulador del Interferón/metabolismo , Unión Proteica , Proteínas Serina-Treonina Quinasas , Virus ARN/genética , Virus ARN/inmunología , Receptores de Reconocimiento de Patrones/metabolismo , Factor 3 Asociado a Receptor de TNF/metabolismo , Proteínas Virales/metabolismo
9.
Viruses ; 13(9)2021 09 06.
Artículo en Inglés | MEDLINE | ID: mdl-34578357

RESUMEN

As a structural protein of the Foot-and-mouth disease virus (FMDV), VP3 plays a vital role in virus assembly and inhibiting the interferon (IFN) signal transduction to promote FMDV replication. Previous studies demonstrated that FMDV VP3 blocks the type-I IFN response by inhibiting the mRNA expression of the mitochondrial antiviral-signaling protein (MAVS); however, the underlying mechanism is poorly understood. Here, we describe the specificity of FMDV VP3 interaction with the transmembrane (TM) domain of MAVS as FMDV driven type-I IFN inhibitory mechanism for its effective replication. The TM domain of MAVS governs the mitochondria localization of MAVS, and it is a key factor in type-I IFN signaling transduction via MAVS aggregation. Thereby, the interaction of FMDV VP3 with the TM domain of MAVS leads to the inhibition of MAVS mitochondria localization, self-association, and aggregation, resulting in the suppression of type-I IFN response. Collectively, these results provide a clear understanding of a key molecular mechanism used by the FMDV VP3 for the suppression of IFN responses via targeting MAVS.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Virus de la Fiebre Aftosa , Fiebre Aftosa/inmunología , Interferón Tipo I/metabolismo , Animales , Anticuerpos Antivirales , Fiebre Aftosa/virología , Virus de la Fiebre Aftosa/genética , Células HEK293 , Células HeLa , Interacciones Huésped-Patógeno/inmunología , Humanos , Ratones , Mitocondrias/metabolismo , Células RAW 264.7 , ARN Mensajero/metabolismo , Transducción de Señal/fisiología
10.
Viruses ; 13(4)2021 03 25.
Artículo en Inglés | MEDLINE | ID: mdl-33806073

RESUMEN

Sargassum fusiforme, a plant used as a medicine and food, is regarded as a marine vegetable and health supplement to improve life expectancy. Here, we demonstrate that S. fusiforme extract (SFE) has antiviral effects against respiratory syncytial virus (RSV) in vitro and in vivo mouse model. Treatment of HEp2 cells with a non-cytotoxic concentration of SFE significantly reduced RSV replication, RSV-induced cell death, RSV gene transcription, RSV protein synthesis, and syncytium formation. Moreover, oral inoculation of SFE significantly improved RSV clearance from the lungs of BALB/c mice. Interestingly, the phenolic compounds eicosane, docosane, and tetracosane were identified as active components of SFE. Treatment with a non-cytotoxic concentration of these three components elicited similar antiviral effects against RSV infection as SFE in vitro. Together, these results suggest that SFE and its potential components are a promising natural antiviral agent candidate against RSV infection.


Asunto(s)
Antivirales/farmacología , Productos Biológicos/análisis , Productos Biológicos/farmacología , Virus Sincitial Respiratorio Humano/efectos de los fármacos , Sargassum/química , Replicación Viral/efectos de los fármacos , Animales , Antivirales/análisis , Antivirales/uso terapéutico , Línea Celular , Modelos Animales de Enfermedad , Humanos , Pulmón/efectos de los fármacos , Pulmón/virología , Ratones , Ratones Endogámicos BALB C , Fenoles/farmacología , Infecciones por Virus Sincitial Respiratorio/tratamiento farmacológico , Carga Viral/efectos de los fármacos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA