Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 20 de 79
Filtrar
1.
J Immunol ; 2024 Jul 05.
Artículo en Inglés | MEDLINE | ID: mdl-38967520

RESUMEN

Stimulator of IFN genes (STING) is a critical component of the innate immune system, playing an essential role in defending against DNA virus infections. However, the mechanisms governing basal STING regulation remain poorly understood. In this study, we demonstrate that the basal level of STING is critically maintained by hypoxia-inducible factor 1 (HIF-1)α through transcription. Under normal conditions, HIF-1α binds constitutively to the promoter region of STING, actively promoting its transcription. Knocking down HIF-1α results in a decrease in STING expression in multiple cell lines and zebrafish, which in turn reduces cellular responses to synthetic dsDNAs, including cell signaling and IFN production. Moreover, this decrease in STING levels leads to an increase in cellular susceptibility to DNA viruses HSV-1 and pseudorabies virus. These findings unveil a (to our knowledge) novel role of HIF-1α in maintaining basal STING levels and provide valuable insights into STING-mediated antiviral activities and associated diseases.

2.
J Virol ; 97(3): e0013423, 2023 03 30.
Artículo en Inglés | MEDLINE | ID: mdl-36916938

RESUMEN

Type I interferon (IFN-I) response plays a prominent role in innate immunity, which is frequently modulated during viral infection. Here, we report DNA methylation regulator UHRF1 as a potent negative regulator of IFN-I induction during alphaherpesvirus infection, whereas the viruses in turn regulates the transcriptional expression of UHRF1. Knockdown of UHRF1 in cells significantly increases interferon-ß (IFN-ß)-mediated gene transcription and viral inhibition against herpes simplex virus 1 (HSV1) and pseudorabies virus (PRV). Mechanistically, UHRF1 deficiency promotes IFN-I production by triggering dsRNA-sensing receptor RIG-I and activating IRF3 phosphorylation. Knockdown of UHRF1 in cells upregulates the accumulation of double-stranded RNA (dsRNA), including host endogenous retroviral sequence (ERV) transcripts, while the treatment of RNase III, known to specifically digest dsRNA, prevents IFN-ß induction by siUHRF1. Furthermore, the double-knockdown assay of UHRF1 and DNA methyltransferase DNMT1 suggests that siUHRF1-mediated DNA demethylation may play an important role in dsRNA accumulation and subsequently IFN induction. These findings establish the essential role of UHRF1 in IFN-I-induced antiviral immunity and reveal UHRF1 as a potential antivrial target. IMPORTANCE Alphaherpesviruses can establish lifelong infections and cause many diseases in humans and animals, which rely partly on their interaction with IFN-mediated innate immune response. Using alphaherpesviruses PRV and HSV-1 as models, we identified an essential role of DNA methylation regulator UHRF1 in IFN-mediated immunity against virus replication, which unravels a novel mechanism employed by epigenetic factor to control IFN-mediated antiviral immune response and highlight UHRF1, which might be a potential target for antiviral drug development.


Asunto(s)
Herpesvirus Humano 1 , Herpesvirus Suido 1 , Interferón Tipo I , Animales , Humanos , Antivirales , Proteínas Potenciadoras de Unión a CCAAT/genética , Proteínas Potenciadoras de Unión a CCAAT/metabolismo , Expresión Génica , Herpesvirus Humano 1/genética , Herpesvirus Suido 1/genética , Inmunidad Innata , Factor 3 Regulador del Interferón/genética , Factor 3 Regulador del Interferón/metabolismo , Interferón Tipo I/metabolismo , Interferón beta/metabolismo , Ubiquitina-Proteína Ligasas/genética , Ubiquitina-Proteína Ligasas/metabolismo , Alphaherpesvirinae , Receptores Inmunológicos/inmunología
3.
Angew Chem Int Ed Engl ; 63(23): e202403918, 2024 Jun 03.
Artículo en Inglés | MEDLINE | ID: mdl-38519423

RESUMEN

Precise design and tuning of Zn hopping/transfer sites with deeper understanding of the dendrite-formation mechanism is vital in artificial anode protective coating for aqueous Zn-ion batteries (AZIBs). Here, we probe into the role of anode-coating interfaces by designing a series of anhydride-based covalent organic frameworks (i.e., PI-DP-COF and PI-DT-COF) with specifically designed zigzag hopping sites and zincophilic anhydride groups that can serve as desired platforms to investigate the related Zn2+ hopping/transfer behaviours as well as the interfacial interaction. Combining theoretical calculations with experiments, the ABC stacking models of these COFs endow the structures with specific zigzag sites along the 1D channel that can accelerate Zn2+ transfer kinetics, lower surface-energy, homogenize ion-distribution or electric-filed. Attributed to these superiorities, thus-obtained optimal PI-DT-COF cells offer excellent cycling lifespan in both symmetric-cell (2000 cycles at 60 mA cm-2) and full-cell (1600 cycles at 2 A g-1), outperforming almost all the reported porous crystalline materials.

4.
J Virol ; 94(18)2020 08 31.
Artículo en Inglés | MEDLINE | ID: mdl-32641476

RESUMEN

Promyelocytic leukemia nuclear bodies (PML-NBs) possess an important intrinsic antiviral activity against alphaherpesvirus infection. PML is the structural backbone of NBs, comprising different isoforms. However, the contribution of each isoform to alphaherpesvirus restriction is not well understood. Here, we report the role of PML-NBs and swine PML (sPML) isoforms in pseudorabies virus (PRV) infection in its natural host swine cells. We found that sPML-NBs exhibit an anti-PRV activity in the context of increasing the expression level of endogenous sPML. Of four sPML isoforms cloned and examined, only isoforms sPML-II and -IIa, not sPML-I and -IVa, expressed in a sPML knockout cells inhibit PRV infection. Both the unique 7b region of sPML-II and the sumoylation-dependent normal formation of PML-NBs are required. 7b possesses a transcriptional repression activity and suppresses viral gene transcription during PRV infection with the cysteine residues 589 and 599 being critically involved. We conclude that sPML-NBs inhibit PRV infection partly by repressing viral gene transcription through the 7b region of sPML-II.IMPORTANCE PML-NBs are nuclear sites that mediate the antiviral restriction of alphaherpesvirus gene expression and replication. However, the contribution of each PML isoform to this activity of PML-NBs is not well characterized. Using PRV and its natural host swine cells as a system, we have discovered that the unique C terminus of sPML isoform II is required for PML-NBs to inhibit PRV infection by directly engaging in repression of viral gene transcription. Our study not only confirms in swine cells that PML-NBs have an antiviral function but also presents a mechanism to suggest that PML-NBs inhibit viral infection in an isoform specific manner.


Asunto(s)
Herpesvirus Suido 1/genética , Cuerpos de Inclusión Intranucleares/genética , Proteína de la Leucemia Promielocítica/genética , Transcripción Genética , Proteínas Virales/genética , Animales , Línea Celular , Células Epiteliales/metabolismo , Células Epiteliales/virología , Regulación de la Expresión Génica , Células HEK293 , Herpesvirus Suido 1/metabolismo , Herpesvirus Suido 1/patogenicidad , Interacciones Huésped-Patógeno/genética , Humanos , Cuerpos de Inclusión Intranucleares/metabolismo , Cuerpos de Inclusión Intranucleares/virología , Macrófagos/metabolismo , Macrófagos/virología , Proteína de la Leucemia Promielocítica/metabolismo , Dominios Proteicos , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Transducción de Señal , Relación Estructura-Actividad , Sumoilación , Porcinos , Proteínas Virales/metabolismo
5.
PLoS Pathog ; 15(1): e1007559, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-30682178

RESUMEN

Type I interferon response plays a prominent role against viral infection, which is frequently disrupted by viruses. Here, we report Bcl-2 associated transcription factor 1 (Bclaf1) is degraded during the alphaherpesvirus Pseudorabies virus (PRV) and Herpes simplex virus type 1 (HSV-1) infections through the viral protein US3. We further reveal that Bclaf1 functions critically in type I interferon signaling. Knockdown or knockout of Bclaf1 in cells significantly impairs interferon-α (IFNα) -mediated gene transcription and viral inhibition against US3 deficient PRV and HSV-1. Mechanistically, Bclaf1 maintains a mechanism allowing STAT1 and STAT2 to be efficiently phosphorylated in response to IFNα, and more importantly, facilitates IFN-stimulated gene factor 3 (ISGF3) binding with IFN-stimulated response elements (ISRE) for efficient gene transcription by directly interacting with ISRE and STAT2. Our studies establish the importance of Bclaf1 in IFNα-induced antiviral immunity and in the control of viral infections.


Asunto(s)
Interferones/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Represoras/metabolismo , Proteínas Supresoras de Tumor/metabolismo , Proteínas Virales/metabolismo , Alphaherpesvirinae/metabolismo , Alphaherpesvirinae/patogenicidad , Animales , Antivirales/farmacología , Línea Celular , China , Herpesvirus Humano 1/metabolismo , Herpesvirus Suido 1/metabolismo , Humanos , Inmunidad Innata/efectos de los fármacos , Interferón Tipo I/inmunología , Subunidad alfa del Factor 3 de Genes Estimulados por el Interferón/metabolismo , Interferón-alfa/metabolismo , Interferones/inmunología , Ratones , Ratones Endogámicos BALB C , Fosforilación , Proteínas Serina-Treonina Quinasas/genética , Proteínas Represoras/fisiología , Elementos de Respuesta , Factor de Transcripción STAT1/metabolismo , Factor de Transcripción STAT2/metabolismo , Transducción de Señal/inmunología , Proteínas Supresoras de Tumor/fisiología , Proteínas Virales/genética , Virosis/genética
6.
Arch Virol ; 166(7): 1943-1950, 2021 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-33982180

RESUMEN

Inflammatory responses are an important part of the innate immune response during viral infection. Various inflammasome complexes have been identified. The pyrin domain-containing 3 (NLRP3) inflammasome plays a critical role in detecting some RNA viruses, such as influenza virus. However, the effect of the NLRP3 inflammasome on infectious bursal disease virus (IBDV) replication is still unclear. Here, we report that IBDV-infection induces the transcription of NLRP3 inflammasome and IL-1ß genes in the immortalized chicken embryo fibroblast cell line DF-1. Inhibition of caspase-1 by Belnacasan (VX-765) suppressed the transcription of IL-1ß, reduced cell lysis, and significantly promoted IBDV replication in DF-1 cells. Furthermore, knockdown of NLRP3 by small interfering RNA promoted IBDV replication in the host cells. Thus, IBDV can induce NLRP3 inflammasome activation in DF-1 cells through a mechanism requiring viral replication, revealing a new antiviral mechanism employed by the host.


Asunto(s)
Virus de la Enfermedad Infecciosa de la Bolsa/inmunología , Inflamasomas/inmunología , Proteína con Dominio Pirina 3 de la Familia NLR/inmunología , Replicación Viral/inmunología , Animales , Línea Celular , Pollos/virología , Fibroblastos/inmunología , Fibroblastos/virología , Inmunidad Innata/inmunología , Interleucina-1beta/inmunología , ARN Interferente Pequeño/inmunología
7.
J Virol ; 93(21)2019 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-31413135

RESUMEN

Porcine reproductive and respiratory syndrome virus (PRRSV) is widely prevalent in pigs, resulting in significant economic losses worldwide. A compelling impact of PRRSV infection is severe pneumonia. In the present study, we found that interleukin-17 (IL-17) was upregulated by PRRSV infection. Subsequently, we demonstrated that PI3K and p38MAPK signaling pathways were essential for PRRSV-induced IL-17 production as addition of phosphatidylinositol 3-kinase (PI3K) and p38MAPK inhibitors dramatically reduced IL-17 production. Furthermore, we show here that deleting the C/EBPß and CREB binding motif in porcine IL-17 promoter abrogated its activation and that knockdown of C/EBPß and CREB remarkably impaired PRRSV-induced IL-17 production, suggesting that IL-17 expression was dependent on C/EBPß and CREB. More specifically, we demonstrate that PRRSV nonstructural protein 11 (nsp11) induced IL-17 production, which was also dependent on PI3K-p38MAPK-C/EBPß/CREB pathways. We then show that Ser74 and Phe76 amino acids were essential for nsp11 to induce IL-17 production and viral rescue. In addition, IRAK1 was required for nsp11 to activate PI3K and enhance IL-17 expression by interacting with each other. Importantly, we demonstrate that PI3K inhibitor significantly suppressed IL-17 production and lung inflammation caused by HP-PRRSV in vivo, implicating that higher IL-17 level induced by HP-PRRSV might be associated with severe lung inflammation. These findings provide new insights onto the molecular mechanisms of the PRRSV-induced IL-17 production and help us further understand the pathogenesis of PRRSV infection.IMPORTANCE Highly pathogenic porcine reproductive and respiratory syndrome virus (HP-PRRSV) associated with severe pneumonia has been one of the most important viral pathogens in pigs. IL-17 is a proinflammatory cytokine that might be associated with the strong inflammation caused by PRRSV. Therefore, we sought to determine whether PRRSV infection affects IL-17 expression, and if so, determine this might partially explain the underlying mechanisms for the strong inflammation in HP-PRRSV-infected pigs, especially in lungs. Here, we show that PRRSV significantly induced IL-17 expression, and we subsequently dissected the molecular mechanisms about how PRRSV regulated IL-17 production. Furthermore, we show that Ser74 and Phe76 in nsp11 were indispensable for IL-17 production and viral replication. Importantly, we demonstrated that PI3K inhibitor impaired IL-17 production and alleviated lung inflammation caused by HP-PRRSV infection. Our findings will help us for a better understanding of PRRSV pathogenesis.


Asunto(s)
Endorribonucleasas/metabolismo , Interleucina-17/metabolismo , Neumonía/patología , Síndrome Respiratorio y de la Reproducción Porcina/virología , Virus del Síndrome Respiratorio y Reproductivo Porcino/patogenicidad , Transducción de Señal , Proteínas no Estructurales Virales/metabolismo , Replicación Viral , Animales , Proteína beta Potenciadora de Unión a CCAAT/genética , Proteína beta Potenciadora de Unión a CCAAT/metabolismo , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/genética , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/metabolismo , Endorribonucleasas/genética , Quinasas Asociadas a Receptores de Interleucina-1/genética , Quinasas Asociadas a Receptores de Interleucina-1/metabolismo , Interleucina-17/genética , Mutación , Fosfatidilinositol 3-Quinasas/genética , Fosfatidilinositol 3-Quinasas/metabolismo , Neumonía/metabolismo , Neumonía/virología , Síndrome Respiratorio y de la Reproducción Porcina/metabolismo , Síndrome Respiratorio y de la Reproducción Porcina/patología , Porcinos , Proteínas no Estructurales Virales/genética , Proteínas Quinasas p38 Activadas por Mitógenos/genética , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
8.
Proc Natl Acad Sci U S A ; 114(36): E7564-E7573, 2017 09 05.
Artículo en Inglés | MEDLINE | ID: mdl-28827346

RESUMEN

Severe fever with thrombocytopenia syndrome virus (SFTSV) and Rift Valley fever virus (RVFV) are two arthropod-borne phleboviruses in the Bunyaviridae family, which cause severe illness in humans and animals. Glycoprotein N (Gn) is one of the envelope proteins on the virus surface and is a major antigenic component. Despite its importance for virus entry and fusion, the molecular features of the phleboviruse Gn were unknown. Here, we present the crystal structures of the Gn head domain from both SFTSV and RVFV, which display a similar compact triangular shape overall, while the three subdomains (domains I, II, and III) making up the Gn head display different arrangements. Ten cysteines in the Gn stem region are conserved among phleboviruses, four of which are responsible for Gn dimerization, as revealed in this study, and they are highly conserved for all members in Bunyaviridae Therefore, we propose an anchoring mode on the viral surface. The complex structure of the SFTSV Gn head and human neutralizing antibody MAb 4-5 reveals that helices α6 in subdomain III is the key component for neutralization. Importantly, the structure indicates that domain III is an ideal region recognized by specific neutralizing antibodies, while domain II is probably recognized by broadly neutralizing antibodies. Collectively, Gn is a desirable vaccine target, and our data provide a molecular basis for the rational design of vaccines against the diseases caused by phleboviruses and a model for bunyavirus Gn embedding on the viral surface.


Asunto(s)
Anticuerpos Neutralizantes/metabolismo , Epítopos/metabolismo , Glicoproteínas/química , Glicoproteínas/metabolismo , Phlebovirus/metabolismo , Proteínas del Envoltorio Viral/química , Proteínas del Envoltorio Viral/metabolismo , Animales , Infecciones por Bunyaviridae/virología , Línea Celular , Cristalografía por Rayos X , Epítopos/química , Fiebre del Valle del Rift/virología , Virus de la Fiebre del Valle del Rift/metabolismo , Células Sf9 , Internalización del Virus
9.
J Virol ; 92(1)2018 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-29070692

RESUMEN

Heartland virus (HRTV) is an emerging human pathogen that belongs to the newly defined family Phenuiviridae, order Bunyavirales Gn and Gc are two viral surface glycoproteins encoded by the M segment and are required for early events during infection. HRTV delivers its genome into the cytoplasm by fusion of the viral envelope and endosomal membranes under low-pH conditions. Here, we describe the crystal structure of HRTV Gc in its postfusion conformation. The structure shows that Gc displays a typical class II fusion protein conformation, and the overall structure is identical to severe fever with thrombocytopenia syndrome virus (SFTSV) Gc, which also belongs to the Phenuiviridae family. However, our structural analysis indicates that the hantavirus Gc presents distinct features in the aspects of subdomain orientation, N-linked glycosylation, the interaction pattern between protomers, and the fusion loop conformation. This suggests their family-specific subunit arrangement during the fusogenic process and supports the recent taxonomic revision of bunyaviruses. Our results provide insights into the comprehensive comparison of class II membrane fusion proteins in two bunyavirus families, yielding valuable information for treatments against these human pathogens.IMPORTANCE HRTV is an insect-borne virus found in America that can infect humans. It belongs to the newly defined family Phenuiviridae, order Bunyavirales HRTV contains three single-stranded RNA segments (L, M, and S). The M segment of the virus encodes a polyprotein precursor that is cleaved into two glycoproteins, Gn and Gc. Gc is a fusion protein facilitating virus entry into host cells. Here, we report the crystal structure of the HRTV Gc protein. The structure displays a typical class II fusion protein conformation. Comparison of HRTV Gc with a recently solved structure of another bunyavirus Gc revealed that these Gc structures display a newly defined family specificity, supporting the recent International Committee on Taxonomy of Viruses reclassification of the bunyaviruses. Our results expand the knowledge of bunyavirus fusion proteins and help us to understand bunyavirus characterizations. This study provides useful information to improve protection against and therapies for bunyavirus infections.


Asunto(s)
Glicoproteínas/química , Phlebovirus/química , Virus ARN/química , Proteínas del Envoltorio Viral/química , Proteínas Virales de Fusión/química , Bunyaviridae/química , Cristalización , Cristalografía por Rayos X , Glicosilación , Orthohantavirus/química , Orthohantavirus/clasificación , Phlebovirus/clasificación , Phlebovirus/genética , Conformación Proteica , Dominios Proteicos , Virus ARN/clasificación , Virus ARN/genética , Proteínas Virales de Fusión/metabolismo , Internalización del Virus
10.
J Gen Virol ; 99(12): 1671-1680, 2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-30382935

RESUMEN

Porcine reproductive and respiratory syndrome (PRRS) is one of the most important diseases in pigs. MicroRNAs (miRNAs) have emerged as an important regulator of virus-host cell interactions and miR-30c has been found to facilitate PRRSV replication. Here, we found that the interferon-alpha/beta receptor beta chain (IFNAR2) was down-regulated, while miR-30c was up-regulated during HV (a highly pathogenic type 2 PRRSV strain) and CH-1a (a classic type 2 PRRSV strain) infection. Subsequently, using bioinformatics analysis, we predicted that the IFNAR2 was targeted by miR-30c. A luciferase assay verified that the 3' UTR of IFNAR2 was targeted by miR-30c, as a mutation on either the target sequence or the miR-30c seed sequence reversed the luciferase activity. In addition, miR-30c and IFNAR2 mRNA were physically co-localized in RNA-induced silencing complex (RISC). Importantly, we showed that miR-30c also impaired the induction of IFN-stimulated genes (ISGs) by targeting IFNAR2. Our findings further reveal the mechanism of miR-30c promoting PRRSV replication.


Asunto(s)
Interacciones Huésped-Patógeno , Evasión Inmune , MicroARNs/metabolismo , Virus del Síndrome Respiratorio y Reproductivo Porcino/crecimiento & desarrollo , Virus del Síndrome Respiratorio y Reproductivo Porcino/patogenicidad , Receptor de Interferón alfa y beta/antagonistas & inhibidores , Animales , Células Cultivadas , Regulación de la Expresión Génica , Macrófagos Alveolares/virología , Virus del Síndrome Respiratorio y Reproductivo Porcino/inmunología , Porcinos , Enfermedades de los Porcinos
11.
J Virol ; 91(21)2017 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-28794045

RESUMEN

Alphaherpesviruses that establish persistent infections rely partly on their ability to evade host antiviral responses, notably the type I interferon (IFN) response. However, the mechanisms employed by alphaherpesviruses to avoid this response are not well understood. Pseudorabies virus (PRV) is an economically important pathogen and a useful model system for studying alphaherpesvirus biology. To identify PRV proteins that antagonize type I IFN signaling, we performed a screen by using an IFN-stimulated response element reporter in the swine cell line CRL. Unexpectedly, we identified the dUTPase UL50 as a strong inhibitor. We confirmed that UL50 has the ability to inhibit type I IFN signaling by performing ectopic expression of UL50 in cells and deletion of UL50 in PRV. Mechanistically, UL50 impeded type I IFN-induced STAT1 phosphorylation, likely by accelerating lysosomal degradation of IFN receptor 1 (IFNAR1). In addition, this UL50 activity was independent of its dUTPase activity and required amino acids 225 to 253 in the C-terminal region. The UL50 encoded by herpes simplex virus 1 (HSV-1) also possessed similar activity. Moreover, UL50-deleted PRV was more susceptible to IFN than UL50-proficient PRV. Our results suggest that in addition to its dUTPase activity, the UL50 protein of alphaherpesviruses possesses the ability to suppress type I IFN signaling by promoting lysosomal degradation of IFNAR1, thereby contributing to immune evasion. This finding reveals UL50 as a potential antiviral target.IMPORTANCE Alphaherpesviruses can establish lifelong infections and cause many diseases in humans and animals. Pseudorabies virus (PRV) is a swine alphaherpesvirus that threatens pig production. Using PRV as a model, we found that this alphaherpesvirus could utilize its encoded dUTPase UL50 to induce IFNAR1 degradation and inhibit type I IFN signaling in an enzymatic activity-independent manner. Our finding reveals a mechanism employed by an alphaherpesvirus to evade the immune response and indicates that UL50 is an important viral protein in pathogenesis and is a potential target for antiviral drug development.


Asunto(s)
Herpesvirus Suido 1/enzimología , Interferón Tipo I/farmacología , Lisosomas/metabolismo , Seudorrabia/metabolismo , Pirofosfatasas/metabolismo , Receptor de Interferón alfa y beta/metabolismo , Secuencia de Aminoácidos , Animales , Antivirales/farmacología , Células HeLa , Herpesvirus Suido 1/genética , Humanos , Evasión Inmune , Riñón/efectos de los fármacos , Riñón/metabolismo , Riñón/virología , Fosforilación , Proteolisis , Seudorrabia/tratamiento farmacológico , Seudorrabia/virología , Pirofosfatasas/genética , Receptor de Interferón alfa y beta/genética , Homología de Secuencia , Transducción de Señal , Porcinos , Proteínas Virales/genética , Proteínas Virales/metabolismo
12.
J Immunol ; 196(5): 2272-82, 2016 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-26826240

RESUMEN

Porcine reproductive and respiratory syndrome virus (PRRSV) is an economically important pathogen and has evolved several mechanisms to evade IFN-I responses. We report that a host microRNA, miR-30c, was upregulated by PRRSV via activating NF-κB and facilitated its ability to infect subject animals. Subsequently, we demonstrated that miR-30c was a potent negative regulator of IFN-I signaling by targeting JAK1, resulting in the enhancement of PRRSV infection. In addition, we found that JAK1 expression was significantly decreased by PRRSV and recovered when miR-30c inhibitor was overexpressed. Importantly, miR-30c was also upregulated by PRRSV infection in vivo, and miR-30c expression corresponded well with viral loads in lungs and porcine alveolar macrophages of PRRSV-infected pigs. Our findings identify a new strategy taken by PRRSV to escape IFN-I-mediated antiviral immune responses by engaging miR-30c and, thus, improve our understanding of its pathogenesis.


Asunto(s)
Interferón Tipo I/metabolismo , Janus Quinasa 1/genética , MicroARNs/genética , Síndrome Respiratorio y de la Reproducción Porcina/genética , Síndrome Respiratorio y de la Reproducción Porcina/metabolismo , Virus del Síndrome Respiratorio y Reproductivo Porcino/fisiología , Interferencia de ARN , Animales , Secuencia de Bases , Sitios de Unión , Línea Celular , Regulación de la Expresión Génica , Janus Quinasa 1/química , Janus Quinasa 1/metabolismo , MicroARNs/química , FN-kappa B/metabolismo , Síndrome Respiratorio y de la Reproducción Porcina/virología , ARN Mensajero/química , ARN Mensajero/genética , Transducción de Señal , Porcinos , Regulación hacia Arriba , Replicación Viral
13.
Nucleic Acids Res ; 42(5): 3464-77, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24369429

RESUMEN

All positive-stranded RNA viruses with genomes>∼7 kb encode helicases, which generally are poorly characterized. The core of the nidovirus superfamily 1 helicase (HEL1) is associated with a unique N-terminal zinc-binding domain (ZBD) that was previously implicated in helicase regulation, genome replication and subgenomic mRNA synthesis. The high-resolution structure of the arterivirus helicase (nsp10), alone and in complex with a polynucleotide substrate, now provides first insights into the structural basis for nidovirus helicase function. A previously uncharacterized domain 1B connects HEL1 domains 1A and 2A to a long linker of ZBD, which further consists of a novel RING-like module and treble-clef zinc finger, together coordinating three Zn atoms. On substrate binding, major conformational changes were evident outside the HEL1 domains, notably in domain 1B. Structural characterization, mutagenesis and biochemistry revealed that helicase activity depends on the extensive relay of interactions between the ZBD and HEL1 domains. The arterivirus helicase structurally resembles the cellular Upf1 helicase, suggesting that nidoviruses may also use their helicases for post-transcriptional quality control of their large RNA genomes.


Asunto(s)
Equartevirus/enzimología , ARN Helicasas/química , Proteínas Virales/química , Adenosina Trifosfatasas/química , Adenosina Trifosfatasas/metabolismo , ADN/metabolismo , Modelos Moleculares , Degradación de ARNm Mediada por Codón sin Sentido , Estructura Terciaria de Proteína , ARN Helicasas/genética , ARN Helicasas/metabolismo , Eliminación de Secuencia , Proteínas Virales/genética , Proteínas Virales/metabolismo , Zinc/química
14.
J Virol ; 88(18): 10934-45, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-25008936

RESUMEN

UNLABELLED: Porcine reproductive and respiratory syndrome virus (PRRSV) is a highly infectious pathogen that causes severe diseases in pigs and great economic losses to the swine industry worldwide. Type I interferons (IFNs) play a crucial role in antiviral immunity. In the present study, we demonstrated that infection with the highly pathogenic PRRSV strain JXwn06 antagonized type I IFN expression induced by poly(I·C) in both porcine alveolar macrophages (PAMs) and blood monocyte-derived macrophages (BMo). Subsequently, we showed that the inhibition of poly(I·C)-induced IFN-ß production by PRRSV was dependent on the blocking of NF-κB signaling pathways. By screening PRRSV nonstructural and structural proteins, we demonstrated that nonstructural protein 4 (nsp4), a viral 3C-like serine protease, significantly suppressed IFN-ß expression. Moreover, we verified that nsp4 inhibited NF-κB activation induced by signaling molecules, including RIG-I, VISA, TRIF, and IKKß. nsp4 was shown to target the NF-κB essential modulator (NEMO) at the E349-S350 site to mediate its cleavage. Importantly, nsp4 mutants with defective protease activity abolished its ability to cleave NEMO and inhibit IFN-ß production. These findings might have implications for our understanding of PRRSV pathogenesis and its mechanisms for evading the host immune response. IMPORTANCE: Porcine reproductive and respiratory syndrome virus (PRRSV) is a major agent of respiratory diseases in pigs. Like many other viruses, PRRSV has evolved a variety of strategies to evade host antiviral innate immunity for survival and propagation. In this study, we show that PRRSV nsp4 is a novel antagonist of the NF-κB signaling pathway, which is responsible for regulating the expression of type I interferons and other crucial cytokines. We then investigated the underlying mechanism used by nsp4 to suppress NF-κB-mediated IFN-ß production. We found that nsp4 interfered with the NF-κB signaling pathway through the cleavage of NEMO (a key regulator of NF-κB signaling) at the E349-S350 site, leading to the downregulation of IFN-ß production induced by poly(I·C). The data presented here may help us to better understand PRRSV pathogenesis.


Asunto(s)
Interferón beta/genética , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Síndrome Respiratorio y de la Reproducción Porcina/metabolismo , Virus del Síndrome Respiratorio y Reproductivo Porcino/metabolismo , Proteínas no Estructurales Virales/metabolismo , Animales , Regulación hacia Abajo , Interacciones Huésped-Patógeno , Interferón beta/metabolismo , Péptidos y Proteínas de Señalización Intracelular/antagonistas & inhibidores , Péptidos y Proteínas de Señalización Intracelular/genética , Macrófagos/metabolismo , Macrófagos/virología , FN-kappa B/genética , FN-kappa B/metabolismo , Síndrome Respiratorio y de la Reproducción Porcina/genética , Síndrome Respiratorio y de la Reproducción Porcina/virología , Virus del Síndrome Respiratorio y Reproductivo Porcino/genética , Transducción de Señal , Porcinos , Proteínas no Estructurales Virales/genética
15.
J Virol ; 88(5): 2810-20, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24352469

RESUMEN

UNLABELLED: Atypical porcine reproductive and respiratory syndrome (PRRS) caused by highly pathogenic porcine reproductive and respiratory syndrome virus (HP-PRRSV) is characterized by high fever and high mortality. However, the mechanism underlying the fever induction is still unknown. Prostaglandin E2 (PGE2), synthesized by cyclooxygenase type 1/2 (COX-1/2) enzymes, is essential for inducing fever. In this study, we found that PGE2, together with COX-1, was significantly elevated by HP-PRRSV. We subsequently demonstrated that extracellular signal-regulated kinase 1/2 (ERK1/2) and phosphorylated ERK (p-ERK) were the key nodes to trigger COX-1 expression after HP-PRRSV infection. Furthermore, we proved the direct binding of p-C/EBP-ß to the COX-1 promoter by luciferase reporter and chromatin immunoprecipitation assays. In addition, silencing of C/EBP-ß remarkably impaired the enhancement of COX-1 production induced by HP-PRRSV infection. Taken together, our results indicate that HP-PPRSV elicits the expression of COX-1 through the ERK1/2-p-C/EBP-ß signaling pathway, resulting in the increase of PGE2, which might be the cause of high fever in infected pigs. Our findings might provide new insights into the molecular mechanisms underlying the pathogenesis of HP-PRRSV infection. IMPORTANCE: The atypical PRRS caused by HP-PRRSV was characterized by high fever, high morbidity, and high mortality in pigs of all ages, yet how HP-PRRSV induces high fever in pigs remains unknown. In the present study, we found out that HP-PRRSV infection could increase PGE2 production by upregulation of COX-1, and we subsequently characterized the underlying mechanisms about how HP-PRRSV enhances COX-1 production. PGE2 plays a critical role in inducing high temperature in hosts during pathogen infections. Thus, our findings here could help us have a better understanding of HP-PRRSV pathogenesis.


Asunto(s)
Proteína beta Potenciadora de Unión a CCAAT/metabolismo , Ciclooxigenasa 1/metabolismo , Dinoprostona/biosíntesis , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Virus del Síndrome Respiratorio y Reproductivo Porcino/fisiología , Transducción de Señal , Animales , Secuencia de Bases , Clonación Molecular , Ciclooxigenasa 1/genética , Datos de Secuencia Molecular , Fosforilación , Síndrome Respiratorio y de la Reproducción Porcina/genética , Síndrome Respiratorio y de la Reproducción Porcina/metabolismo , Virus del Síndrome Respiratorio y Reproductivo Porcino/patogenicidad , Regiones Promotoras Genéticas , Elementos de Respuesta , Porcinos
16.
J Virol ; 87(2): 1159-71, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-23152505

RESUMEN

Porcine reproductive and respiratory syndrome virus (PRRSV) is one of the most important viral pathogens in the swine industry. Emerging evidence indicates that the host microRNAs (miRNAs) are involved in host-pathogen interactions. However, whether host miRNAs can target PRRSV and be used to inhibit PRRSV infection has not been reported. Recently, microRNA 181 (miR-181) has been identified as a positive regulator of immune response, and here we report that miR-181 can directly impair PRRSV infection. Our results showed that delivered miR-181 mimics can strongly inhibit PRRSV replication in vitro through specifically binding to a highly (over 96%) conserved region in the downstream of open reading frame 4 (ORF4) of the viral genomic RNA. The inhibition of PRRSV replication was specific and dose dependent. In PRRSV-infected Marc-145 cells, the viral mRNAs could compete with miR-181-targeted sequence in luciferase vector to interact with miR-181 and result in less inhibition of luciferase activity, further demonstrating the specific interactions between miR-181 and PRRSV RNAs. As expected, miR-181 and other potential PRRSV-targeting miRNAs (such as miR-206) are expressed much more abundantly in minimally permissive cells or tissues than in highly permissive cells or tissues. Importantly, highly pathogenic PRRSV (HP-PRRSV) strain-infected pigs treated with miR-181 mimics showed substantially decreased viral loads in blood and relief from PRRSV-induced fever compared to negative-control (NC)-treated controls. These results indicate the important role of host miRNAs in modulating PRRSV infection and viral pathogenesis and also support the idea that host miRNAs could be useful for RNA interference (RNAi)-mediated antiviral therapeutic strategies.


Asunto(s)
MicroARNs/metabolismo , Virus del Síndrome Respiratorio y Reproductivo Porcino/fisiología , ARN Viral/metabolismo , Replicación Viral , Animales , Línea Celular , MicroARNs/genética , MicroARNs/inmunología , Virus del Síndrome Respiratorio y Reproductivo Porcino/genética , Virus del Síndrome Respiratorio y Reproductivo Porcino/inmunología , ARN Viral/genética , Porcinos
17.
J Virol ; 87(15): 8808-12, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23740977

RESUMEN

We previously showed that microRNA 181 (miR-181) can inhibit PRRSV replication by directly targeting its genomic RNA. Here, we report that miR-181 can downregulate the PRRSV receptor CD163 in blood monocytes and porcine alveolar macrophages (PAMs) through targeting the 3' untranslated region (UTR) of CD163 mRNA. Downregulation of CD163 leads to the inhibition of PRRSV entry into PAMs and subsequently suppresses PRRSV infection. Our findings indicate that delivery of miR-181 can be used as antiviral therapy against PRRSV infection.


Asunto(s)
MicroARNs/metabolismo , Virus del Síndrome Respiratorio y Reproductivo Porcino/crecimiento & desarrollo , Receptores de Superficie Celular/antagonistas & inhibidores , Receptores Virales/antagonistas & inhibidores , Internalización del Virus , Animales , Antígenos CD/genética , Antígenos de Diferenciación Mielomonocítica/genética , Macrófagos Alveolares/virología , MicroARNs/genética , Monocitos/virología , Virus del Síndrome Respiratorio y Reproductivo Porcino/fisiología , Receptores de Superficie Celular/genética , Receptores Virales/genética , Porcinos
18.
J Virol ; 87(5): 2963-8, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-23269805

RESUMEN

H9N2 influenza viruses with an A316S substitution in hemagglutinin (HA) and a shorter neuraminidase (NA) stalk have become predominant in China. The A316S was shown to increase HA cleavage efficiency when combined with short stalk NA, and the short stalk NA improved NA enzyme activity and release of virus from erythrocytes. Single mutations or combinations of these mutations strengthened the virulence of H9N2 virus in chickens and mice.


Asunto(s)
Glicoproteínas Hemaglutininas del Virus de la Influenza/química , Glicoproteínas Hemaglutininas del Virus de la Influenza/genética , Subtipo H9N2 del Virus de la Influenza A/química , Subtipo H9N2 del Virus de la Influenza A/patogenicidad , Neuraminidasa/química , Neuraminidasa/genética , Infecciones por Orthomyxoviridae/virología , Sustitución de Aminoácidos , Animales , Embrión de Pollo , Pollos , Perros , Eritrocitos/virología , Glicoproteínas Hemaglutininas del Virus de la Influenza/metabolismo , Subtipo H9N2 del Virus de la Influenza A/genética , Células de Riñón Canino Madin Darby , Ratones , Mutación , Neuraminidasa/metabolismo , Enfermedades de las Aves de Corral/virología , Liberación del Virus
19.
Front Immunol ; 15: 1339510, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38449860

RESUMEN

African swine fever (ASF) caused by African swine fever virus (ASFV) is a highly mortal and hemorrhagic infectious disease in pigs. Previous studies have indicated that ASFV modulates interferon (IFN) production. In this study, we demonstrated that ASFV pA151R negatively regulated type I IFN production. Ectopic expression of pA151R dramatically inhibited K63-linked polyubiquitination and Ser172 phosphorylation of TANK-binding kinase 1 (TBK1). Mechanically, we demonstrated that E3 ligase TNF receptor-associated factor 6 (TRAF6) participated in the ubiquitination of TBK1 in cGAS-STING signaling pathway. We showed that pA151R interacted with TRAF6 and degraded it through apoptosis pathway, leading to the disruption of TBK1 and TRAF6 interaction. Moreover, we clarified that the amino acids H102, C109, C132, and C135 in pA151R were crucial for pA151R to inhibit type I interferon production. In addition, we verified that overexpression of pA151R facilitated DNA virus Herpes simplex virus 1 (HSV-1) replication by inhibiting IFN-ß production. Importantly, knockdown of pA151R inhibited ASFV replication and enhanced IFN-ß production in porcine alveolar macrophages (PAMs). Our findings will help understand how ASFV escapes host antiviral immune responses and develop effective ASFV vaccines.


Asunto(s)
Virus de la Fiebre Porcina Africana , Fiebre Porcina Africana , Animales , Porcinos , Ubiquitina-Proteína Ligasas , Factor 6 Asociado a Receptor de TNF , Ubiquitinación
20.
J Virol ; 86(23): 13121, 2012 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-23118452

RESUMEN

Highly pathogenic porcine reproductive and respiratory syndrome (HP-PRRS) emerged in China in 2006, and HP-PRRS virus (HP-PRRSV) has evolved continuously. Here, the complete genomic sequence of a novel HP-PRRSV field strain, JX, is reported. The present finding will contribute to further studies focusing on the evolutionary mechanism of PRRSV.


Asunto(s)
Enfermedades Transmisibles Emergentes/veterinaria , Genoma Viral/genética , Síndrome Respiratorio y de la Reproducción Porcina/epidemiología , Síndrome Respiratorio y de la Reproducción Porcina/virología , Virus del Síndrome Respiratorio y Reproductivo Porcino/genética , Animales , Secuencia de Bases , China/epidemiología , Análisis por Conglomerados , Enfermedades Transmisibles Emergentes/virología , Datos de Secuencia Molecular , Filogenia , Análisis de Secuencia de ADN/veterinaria , Porcinos
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda