Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 24
Filtrar
Más filtros

Bases de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
J Biol Chem ; 300(5): 107249, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38556084

RESUMEN

Tripartite-motif protein-56 (TRIM56) positively regulates the induction of type I interferon response via the TLR3 pathway by enhancing IRF3 activation and depends on its C-terminal residues 621-750 for interacting with the adaptor TRIF. However, the precise underlying mechanism and detailed TRIM56 determinants remain unclear. Herein, we show ectopic expression of murine TRIM56 also enhances TLR3-dependent interferon-ß promoter activation, suggesting functional conservation. We found that endogenous TRIM56 and TRIF formed a complex early (0.5-2 h) after poly-I:C stimulation and that TRIM56 overexpression also promoted activation of NF-κB by poly-I:C but not that by TNF-α or IL-1ß, consistent with a specific effect on TRIF prior to the bifurcation of NF-κB and IRF3. Using transient transfection and Tet-regulated cell lines expressing various TRIM56 mutants, we demonstrated the Coiled-coil domain and a segment spanning residues ∼434-610, but not the B-box or residues 355-433, were required for TRIM56 augmentation of TLR3 signaling. Moreover, alanine substitution at each putative phosphorylation site, Ser471, Ser475, and Ser710, abrogated TRIM56 function. Concordantly, mutants bearing Ser471Ala, Ser475Ala, or Ser710Ala, or lacking the Coiled-coil domain, all lost the capacity to enhance poly-I:C-induced establishment of an antiviral state. Furthermore, the Ser710Ala mutation disrupted the TRIM56-TRIF association. Using phospho-specific antibodies, we detected biphasic phosphorylation of TRIM56 at Ser471 and Ser475 following TLR3 stimulation, with the early phase occurring at ∼0.5 to 1 h, prior to IRF3 phosphorylation. Together, these data reveal novel molecular details critical for the TRIM56 augmentation of TLR3-dependent antiviral response and highlight important roles for TRIM56 scaffolding and phosphorylation.


Asunto(s)
Proteínas Adaptadoras del Transporte Vesicular , Inmunidad Innata , Receptor Toll-Like 3 , Proteínas de Motivos Tripartitos , Animales , Humanos , Ratones , Proteínas Adaptadoras del Transporte Vesicular/metabolismo , Proteínas Adaptadoras del Transporte Vesicular/genética , Proteínas Adaptadoras del Transporte Vesicular/inmunología , Células HEK293 , Factor 3 Regulador del Interferón/metabolismo , Factor 3 Regulador del Interferón/genética , FN-kappa B/metabolismo , Fosforilación , Poli I-C/farmacología , Dominios Proteicos , Transducción de Señal , Receptor Toll-Like 3/metabolismo , Receptor Toll-Like 3/genética , Proteínas de Motivos Tripartitos/metabolismo , Proteínas de Motivos Tripartitos/genética , Ubiquitina-Proteína Ligasas/metabolismo , Ubiquitina-Proteína Ligasas/genética
2.
BMC Immunol ; 24(1): 7, 2023 04 21.
Artículo en Inglés | MEDLINE | ID: mdl-37085747

RESUMEN

BACKGROUND: Coevolution between pathogens and their hosts decreases host morbidity and mortality. Bats host and can tolerate viruses which can be lethal to other vertebrate orders, including humans. Bat adaptations to infection include localized immune response, early pathogen sensing, high interferon expression without pathogen stimulation, and regulated inflammatory response. The immune reaction is costly, and bats suppress high-cost metabolism during torpor. In the temperate zone, bats hibernate in winter, utilizing a specific behavioural adaptation to survive detrimental environmental conditions and lack of energy resources. Hibernation torpor involves major physiological changes that pose an additional challenge to bat-pathogen coexistence. Here, we compared bat cellular reaction to viral challenge under conditions simulating hibernation, evaluating the changes between torpor and euthermia. RESULTS: We infected the olfactory nerve-derived cell culture of Myotis myotis with an endemic bat pathogen, European bat lyssavirus 1 (EBLV-1). After infection, the bat cells were cultivated at two different temperatures, 37 °C and 5 °C, to examine the cell response during conditions simulating euthermia and torpor, respectively. The mRNA isolated from the cells was sequenced and analysed for differential gene expression attributable to the temperature and/or infection treatment. In conditions simulating euthermia, infected bat cells produce an excess signalling by multitude of pathways involved in apoptosis and immune regulation influencing proliferation of regulatory cell types which can, in synergy with other produced cytokines, contribute to viral tolerance. We found no up- or down-regulated genes expressed in infected cells cultivated at conditions simulating torpor compared to non-infected cells cultivated under the same conditions. When studying the reaction of uninfected cells to the temperature treatment, bat cells show an increased production of heat shock proteins (HSPs) with chaperone activity, improving the bat's ability to repair molecular structures damaged due to the stress related to the temperature change. CONCLUSIONS: The lack of bat cell reaction to infection in conditions simulating hibernation may contribute to the virus tolerance or persistence in bats. Together with the cell damage repair mechanisms induced in response to hibernation, the immune regulation may promote bats' ability to act as reservoirs of zoonotic viruses such as lyssaviruses.


Asunto(s)
Quirópteros , Hibernación , Lyssavirus , Virus , Animales , Quirópteros/fisiología , Transcriptoma
3.
J Med Virol ; 95(3): e28686, 2023 03.
Artículo en Inglés | MEDLINE | ID: mdl-36938992

RESUMEN

Recent findings in permanent cell lines suggested that SARS-CoV-2 Omicron BA.1 induces a stronger interferon response than Delta. Here, we show that BA.1 and BA.5 but not Delta induce an antiviral state in air-liquid interface cultures of primary human bronchial epithelial cells and primary human monocytes. Both Omicron subvariants caused the production of biologically active types I (α/ß) and III (λ) interferons and protected cells from super-infection with influenza A viruses. Notably, abortive Omicron infection of monocytes was sufficient to protect monocytes from influenza A virus infection. Interestingly, while influenza-like illnesses surged during the Delta wave in England, their spread rapidly declined upon the emergence of Omicron. Mechanistically, Omicron-induced interferon signaling was mediated via double-stranded RNA recognition by MDA5, as MDA5 knockout prevented it. The JAK/STAT inhibitor baricitinib inhibited the Omicron-mediated antiviral response, suggesting it is caused by MDA5-mediated interferon production, which activates interferon receptors that then trigger JAK/STAT signaling. In conclusion, our study (1) demonstrates that only Omicron but not Delta induces a substantial interferon response in physiologically relevant models, (2) shows that Omicron infection protects cells from influenza A virus super-infection, and (3) indicates that BA.1 and BA.5 induce comparable antiviral states.


Asunto(s)
COVID-19 , Subtipo H1N1 del Virus de la Influenza A , Subtipo H5N1 del Virus de la Influenza A , Virus de la Influenza A , Gripe Humana , Inhibidores de las Cinasas Janus , Humanos , SARS-CoV-2 , Interferones , Antivirales
4.
Virus Genes ; 57(1): 60-71, 2021 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-33389635

RESUMEN

Peste des petits ruminants virus (PPRV) causes an acute and highly contagious disease in domestic and wild small ruminants throughout the world, mainly by invoking immunosuppression in its natural hosts. It has been suggested that the non-structural C protein of PPRV helps in evading host responses but the molecular mechanisms by which it antagonizes the host responses have not been fully characterized. Here, we report the antagonistic effect of PPRV C protein on the expression of interferon-ß (IFN-ß) through both MAVS and RIG-I mediated pathways in vitro. Dual luciferase reporter assay and direct expression of IFN-ß mRNA analysis indicated that PPRV C significantly down regulates IFN-ß via its potential interaction with MAVS and RIG-I signaling molecules. Results further indicated that PPRV C protein significantly suppresses endogenous and exogenous IFN-ß-induced anti-viral effects in PPRV, EMCV and SVS infections in vitro. Moreover, PPRV C protein not only down regulates IFN-ß but also the downstream cytokines of interferon stimulated genes 56 (ISG56), ISG15, C-X-C motif chemokine (CXCL10) and RIG-I mediated activation of IFN promoter elements of ISRE and NF-κB. Further, this study deciphers that PPRV C protein could significantly inhibit the phosphorylation of STAT1 and interferes with the signal transmission in JAK-STAT signaling pathway. Collectively, this study indicates that PPRV C protein is important for innate immune evasion and disease progression.


Asunto(s)
Interferón beta/metabolismo , Peste de los Pequeños Rumiantes/virología , Virus de la Peste de los Pequeños Rumiantes/metabolismo , Proteínas no Estructurales Virales/metabolismo , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Animales , Chlorocebus aethiops , Proteína 58 DEAD Box/metabolismo , Células HEK293 , Humanos , Receptores Inmunológicos/metabolismo , Transducción de Señal , Células Vero
5.
J Autoimmun ; 101: 1-16, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-31047767

RESUMEN

Alteration in endogenous Interferon (IFN) system may profoundly impact immune cell function in autoimmune diseases. Here, we provide evidence that dysregulation in IFN-regulated genes and pathways are involved in B cell- and monocyte-driven pathogenic contribution to Multiple Sclerosis (MS) development and maintenance. In particular, by using an Interferome-based cell type-specific approach, we characterized an increased susceptibility to an IFN-linked caspase-3 dependent apoptotic cell death in both B cells and monocytes of MS patients that may arise from their chronic activation and persistent stimulation by activated T cells. Ongoing caspase-3 activation functionally impacts on MS monocyte properties influencing the STAT-3/IL-16 axis, thus, driving increased expression and massive release of the bio-active IL-16 triggering and perpetuating CD4+ T cell migration. Importantly, our analysis also identified a previously unknown multi-component defect in type I IFN-mediated signaling and response to virus pathways specific of MS B cells, impacting on induction of anti-viral responses and Epstein-barr virus infection control in patients. Taking advantage of cell type-specific transcriptomics and in-depth functional validation, this study revealed pathogenic contribution of endogenous IFN signaling and IFN-regulated cell processes to MS pathogenesis with implications on fate and functions of B cells and monocytes that may hold therapeutic potential.


Asunto(s)
Linfocitos B/inmunología , Linfocitos B/metabolismo , Interferón Tipo I/genética , Monocitos/inmunología , Monocitos/metabolismo , Esclerosis Múltiple/etiología , Esclerosis Múltiple/metabolismo , Transcriptoma , Adulto , Apoptosis , Biomarcadores , Estudios de Casos y Controles , Susceptibilidad a Enfermedades , Femenino , Perfilación de la Expresión Génica , Humanos , Inmunofenotipificación , Interferón Tipo I/metabolismo , Interleucina-16/genética , Masculino , Persona de Mediana Edad , Esclerosis Múltiple/patología , Especificidad de Órganos/genética , Especificidad de Órganos/inmunología , Regiones Promotoras Genéticas , Transducción de Señal
6.
Virol J ; 14(1): 213, 2017 11 03.
Artículo en Inglés | MEDLINE | ID: mdl-29100522

RESUMEN

BACKGROUND: Endothelial cells are believed to play an important role in response to virus infection. Our previous microarray analysis showed that H9N2 virus infection and inactivated viral particle inoculation increased the expression of interferon-inducible transmembrane protein 1 (IFITM1) in human umbilical vein endothelial cells (HUVECs). In present study, we deeply investigated the expression patterns of IFITM1 and IFITM1-mediated antiviral response induced by H9N2 virus infection and inactivated viral particle inoculation in HUVECs. Epithelial cells that are considered target cells of the influenza virus were selected as a reference control. METHODS: First, we quantified the expression levels of IFITM1 in HUVECs induced by H9N2 virus infection or viral particle inoculation using quantitative real-time PCR and western blot. Second, we observed whether hemagglutinin or neuraminidase affected IFITM1 expression in HUVECs. Finally, we investigated the effect of induced-IFITM1 on the antiviral state in HUVECs by siRNA and activation plasmid transfection. RESULTS: Both H9N2 virus infection and viral particle inoculation increased the expression of IFITM1 without elevating the levels of interferon-ɑ/ß in HUVECs. HA or NA protein binding alone is not sufficient to increase the levels of IFITM1 and interferon-ɑ/ß in HUVECs. IFITM1 induced by viral particle inoculation significantly decreased the virus titers in culture supernatants of HUVECs. CONCLUSIONS: Our results showed that inactivated viral particle inoculation increased the expression of IFITM1 at mRNA and protein levels. Moreover, the induction of IFITM1 expression mediated the antiviral state in HUVECs.


Asunto(s)
Antígenos de Diferenciación/metabolismo , Antivirales/metabolismo , Células Endoteliales de la Vena Umbilical Humana/virología , Subtipo H9N2 del Virus de la Influenza A/inmunología , Virión/inmunología , Antígenos de Diferenciación/genética , Línea Celular , Interacciones Huésped-Patógeno/inmunología , Humanos , Inmunidad Innata/inmunología , Subtipo H9N2 del Virus de la Influenza A/genética , Interferón-alfa/metabolismo , Interferón beta/metabolismo , ARN Interferente Pequeño/genética , Virión/genética , Inactivación de Virus
7.
Fish Shellfish Immunol ; 68: 266-271, 2017 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-28705724

RESUMEN

Double-stranded RNA (dsRNA) is produced by almost all viruses during their replicative cycle and is a potent inducer of the innate antiviral immune response including inducing expression of type I interferons (IFNs) and interferon-stimulated genes (ISGs). During lytic virus infections intracellular dsRNA can escape into the extracellular space, where surface pattern recognition receptors, such as class A scavenger receptors (SR-As) facilitate its binding and entry into neighbouring cells. Studying extracellular dsRNA entry is difficult due to the ubiquitous expression profile and compensatory dsRNA binding characteristics of SR-As; a SR-A deficient cell line has yet to be identified. The present study suggests the Chinook salmon embryonic cell line, CHSE-214, as a model for studying extracellular dsRNA sensing in vitro. CHSE-214 is unable to bind and respond to extracellular dsRNA, can only respond to dsRNA when it is transfected into the cells, and is able to bind dsRNA when overexpressing human SR-AI. The applications for this model could include elucidating: dsRNA binding and entry mechanisms, including sequence and length effects, as well as SR-A and other putative surface dsRNA receptor ligand binding studies.


Asunto(s)
ARN Bicatenario/metabolismo , ARN Viral/metabolismo , Salmón , Factores de Empalme Serina-Arginina/metabolismo , Animales , Línea Celular , Embrión no Mamífero , Acoplamiento Viral
8.
Viruses ; 16(6)2024 Jun 11.
Artículo en Inglés | MEDLINE | ID: mdl-38932230

RESUMEN

Type I interferons (IFN-Is) are pivotal in innate immunity against human immunodeficiency virus I (HIV-1) by eliciting the expression of IFN-stimulated genes (ISGs), which encompass potent host restriction factors. While ISGs restrict the viral replication within the host cell by targeting various stages of the viral life cycle, the lesser-known IFN-repressed genes (IRepGs), including RNA-binding proteins (RBPs), affect the viral replication by altering the expression of the host dependency factors that are essential for efficient HIV-1 gene expression. Both the host restriction and dependency factors determine the viral replication efficiency; however, the understanding of the IRepGs implicated in HIV-1 infection remains greatly limited at present. This review provides a comprehensive overview of the current understanding regarding the impact of the RNA-binding protein families, specifically the two families of splicing-associated proteins SRSF and hnRNP, on HIV-1 gene expression and viral replication. Since the recent findings show specifically that SRSF1 and hnRNP A0 are regulated by IFN-I in various cell lines and primary cells, including intestinal lamina propria mononuclear cells (LPMCs) and peripheral blood mononuclear cells (PBMCs), we particularly discuss their role in the context of the innate immunity affecting HIV-1 replication.


Asunto(s)
Infecciones por VIH , VIH-1 , Inmunidad Innata , Replicación Viral , VIH-1/genética , VIH-1/fisiología , Humanos , Infecciones por VIH/virología , Infecciones por VIH/genética , Infecciones por VIH/inmunología , Regulación Viral de la Expresión Génica , Factores de Empalme de ARN/metabolismo , Factores de Empalme de ARN/genética , Interferón Tipo I/metabolismo , Interferón Tipo I/genética , Interacciones Huésped-Patógeno/inmunología , Interacciones Huésped-Patógeno/genética , Interferones/metabolismo , Interferones/genética , Interferones/inmunología , Proteínas de Unión al ARN/genética , Proteínas de Unión al ARN/metabolismo
9.
J Mol Biol ; 434(6): 167389, 2022 03 30.
Artículo en Inglés | MEDLINE | ID: mdl-34883114

RESUMEN

Dendritic cells (DCs) play a pivotal role in the functional differentiation of CD4+ T cells in response to pathogens. In CD4+ T cells, HIV-1 replicates efficiently, while HIV-2, a related virus of reduced pathogenicity, is better controlled. How the DC response to HIV-1 vs HIV-2 contributes to programming an antiviral state in CD4+ T cells is not known. Here, we identify a transcriptional signature associated with progressive resistance to HIV infection in CD4+ T cells. We developed a model of naïve CD4+ T cell priming by DCs stimulated with a panel of seven viruses or synthetic ligands for the viral nucleic acid sensors cGAS and TLRs. DCs produced a cytokine response to HIV-2 infection more similar to the response to cGAS ligands than TLR ligands. In response to these signals, naive CD4+ T cells acquired a gradual antiviral resistance to subsequent HIV infection. The antiviral state was concomitant with the induction of the TH1 cytokine IFNγ and the type I interferon-stimulated gene (ISG) MX1, while the TFH cytokine IL-21 was not increased. By performing a transcriptional network analysis in T cells, we identified five distinct gene modules with characteristic ISG, TH1, TFH, IFN-I and proliferative signatures. Finally, we leverage this module to assemble a T antiviral signature of 404 genes that correlate with the antiviral state in T cells. Altogether, the study illuminates the programming of the antiviral state in T cells. The T antiviral gene signature in human CD4+ lymphocytes constitutes a resource for genetic screens and genomics analysis.


Asunto(s)
Linfocitos T CD4-Positivos , Células Dendríticas , Infecciones por VIH , VIH-2 , Transcriptoma , Linfocitos T CD4-Positivos/inmunología , Citocinas/metabolismo , Células Dendríticas/inmunología , Células Dendríticas/virología , Infecciones por VIH/genética , Infecciones por VIH/inmunología , VIH-2/genética , VIH-2/fisiología , Humanos , Ligandos , Nucleotidiltransferasas/metabolismo , Replicación Viral
10.
Pharmaceuticals (Basel) ; 15(5)2022 May 17.
Artículo en Inglés | MEDLINE | ID: mdl-35631443

RESUMEN

The synthetic compounds, Tilorone and Cridanimod, have the antiviral activity which initially had been ascribed to the capacity to induce interferon. Both drugs induce interferon in mice but not in humans. This study investigates whether these compounds have the antiviral activity in mice and rats since rats more closely resemble the human response. Viral-infection models were created in CD-1 mice and Wistar rats. Three strains of Venezuelan equine encephalitis virus were tested for the performance in these models. One virus strain is the molecularly cloned attenuated vaccine. The second strain has major virulence determinants converted to the wild-type state which are present in virulent strains. The third virus has wild-type virulence determinants, and in addition, is engineered to express green fluorescent protein. Experimentally infected animals received Tilorone or Cridanimod, and their treatment was equivalent to the pharmacopoeia-recomended human treatment regimen. Tilorone and Cridanimod show the antiviral activity in mice and rats and protect the mice from death. In rats, both drugs diminish the viremia. These drugs do not induce interferon-alpha or interferon-beta in rats. The presented observations allow postulating the existence of an interferon-independent and species-independent mechanism of action.

11.
Front Immunol ; 13: 1035532, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36439115

RESUMEN

Macrophages are key target cells of Zika virus (ZIKV) infection, implicated as a viral reservoir seeding sanctuary sites such as the central nervous system and testes. This rests on the apparent ability of macrophages to sustain ZIKV replication without experiencing cytopathic effects. ZIKV infection of macrophages triggers an innate immune response involving type I interferons (IFN-I), key antiviral cytokines that play a complex role in ZIKV pathogenesis in animal models. To investigate the functional role of the IFN-I response we generated human induced pluripotent stem cell (iPSC)-derived macrophages from a patient with complete deficiency of IFNAR2, the high affinity IFN-I receptor subunit. Accompanying the profound defect of IFN-I signalling in IFNAR2 deficient iPS-macrophages we observed significantly enhanced ZIKV replication and cell death, revealing the inherent cytopathicity of ZIKV towards macrophages. These observations were recapitulated by genetic and pharmacological ablation of IFN-I signalling in control iPS-macrophages and extended to a model of iPS-microglia. Thus, the capacity of macrophages to support noncytolytic ZIKV replication depends on an equilibrium set by IFN-I, suggesting that innate antiviral responses might counterintuitively promote ZIKV persistence via the maintenance of tissue viral reservoirs relevant to pathogenesis.


Asunto(s)
Células Madre Pluripotentes Inducidas , Infección por el Virus Zika , Virus Zika , Animales , Humanos , Receptor de Interferón alfa y beta/genética , Microglía/metabolismo , Células Madre Pluripotentes Inducidas/metabolismo , Macrófagos/metabolismo , Interferones/farmacología , Antivirales/uso terapéutico
12.
Drug Deliv Transl Res ; 11(4): 1420-1437, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-33748879

RESUMEN

The COVID-19 pandemic's high mortality rate and severe socioeconomic impact serve as a reminder of the urgent need for effective countermeasures against viral pandemic threats. In particular, effective antiviral therapeutics capable of stopping infections in its tracks is critical to reducing infection fatality rate and healthcare burden. With the field of drug delivery witnessing tremendous advancement in the last two decades owing to a panoply of nanotechnology advances, the present review summarizes and expounds on the research and development of therapeutic nanoformulations against various infectious viral pathogens, including HIV, influenza, and coronaviruses. Specifically, nanotechnology advances towards improving pathogen- and host-targeted antiviral drug delivery are reviewed, and the prospect of achieving effective viral eradication, broad-spectrum antiviral effect, and resisting viral mutations are discussed. As several COVID-19 antiviral clinical trials are met with lackluster treatment efficacy, nanocarrier strategies aimed at improving drug pharmacokinetics, biodistributions, and synergism are expected to not only contribute to the current disease treatment efforts but also expand the antiviral arsenal against other emerging viral diseases.


Asunto(s)
Antivirales/administración & dosificación , COVID-19/prevención & control , Sistemas de Liberación de Medicamentos/métodos , Interacciones Huésped-Patógeno/efectos de los fármacos , Nanopartículas/administración & dosificación , Nanotecnología/métodos , Animales , Antivirales/inmunología , COVID-19/epidemiología , COVID-19/inmunología , Sistemas de Liberación de Medicamentos/tendencias , Interacciones Huésped-Patógeno/inmunología , Humanos , Nanotecnología/tendencias , Pandemias/prevención & control , Virosis/epidemiología , Virosis/inmunología , Virosis/prevención & control , Replicación Viral/efectos de los fármacos , Replicación Viral/fisiología
13.
J Vet Med Sci ; 2020 Nov 13.
Artículo en Inglés | MEDLINE | ID: mdl-33191335

RESUMEN

Peste des petits ruminants (PPR) is an OIE-listed, acute, and highly contagious viral disease of sheep and goats caused by the PPR virus (PPRV), a morbillivirus within the Paramyxoviridae family. Here, we investigated how the PPRV protein evades the immune response using cellular models of infection. Results indicated that PPRV V protein significantly suppresses both endogenous and exogenous IFN-α- and IFN-ß-induced antiviral response with a broad-spectrum effect. The PPRV V protein significantly suppresses the production of IFN-ß and its downstream cytokines of interferon-stimulated gene 56 (ISG56), ISG15, C-X-C motif chemokine (CXCL10) as well as the RIG-IN-induced activation of IFN-responsive promoter elements (ISRE). We further found that PPRV V protein inhibits the phosphorylation of IRF3 and STAT1, reducing the production of IFNs to block transduction via JAK-STAT signaling pathway and impairs the host antiviral state.

14.
Front Immunol ; 10: 1019, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31156620

RESUMEN

Viral defense at mucosal sites depends on interferons (IFN) and IFN stimulated genes (ISGs), either of which may be constitutively expressed to maintain an "antiviral state" (AVS). However, the mechanisms that govern the AVS are poorly defined. Using a BEAS-2B respiratory epithelial cell line deficient in IRF1, we demonstrate higher susceptibility to infection with vesicular stomatitis virus (VSV) and influenza virus. IRF1-mediated restriction of VSV is IFN-independent, as blockade of types I and III IFNs and JAK-STAT signaling before infection did not affect VSV infection of either parent or IRF1 KO cells. Transcriptome analysis revealed that IRF1 regulates constitutive expression of ~300 genes, including antiviral ISGs: OAS2, BST2, and RNASEL and knockdown of any of these IRF1-dependent genes increased VSV infection. Additionally, IRF1 enhances rapid expression of IFNß and IFNλ after stimulation with poly I:C and also regulates ISG expression. Mechanistically, IRF1 enhances recruitment of BRD4 to promotor-enhancer regions of ISGs for rapid expression and maintains levels of histone H3K4me1 for optimal constitutive expression. Finally, IRF1 also regulates constitutive expression of TLR2 and TLR3 and promotes signaling through these pattern recognition receptors (PRR). These data reveal multiple roles for IRF1 toward effective anti-viral responses by maintaining IFN-independent constitutive expression of anti-viral ISGs and supporting early IFN-dependent responses to PRR stimulation.


Asunto(s)
2',5'-Oligoadenilato Sintetasa/genética , Antígenos CD/genética , Endorribonucleasas/genética , Gripe Humana/inmunología , Factor 1 Regulador del Interferón/genética , Orthomyxoviridae/inmunología , Infecciones por Rhabdoviridae/inmunología , Vesiculovirus/inmunología , Células A549 , Células Epiteliales/metabolismo , Proteínas Ligadas a GPI/genética , Regulación de la Expresión Génica/genética , Técnicas de Inactivación de Genes , Humanos , Gripe Humana/virología , Factor 1 Regulador del Interferón/metabolismo , Interferones/metabolismo , Mucosa Respiratoria/citología , Infecciones por Rhabdoviridae/virología , Transducción de Señal/genética , Transducción de Señal/inmunología , Receptores Toll-Like/metabolismo , Transfección , Replicación Viral/genética , Replicación Viral/inmunología
15.
J Leukoc Biol ; 103(3): 465-477, 2018 03.
Artículo en Inglés | MEDLINE | ID: mdl-29345347

RESUMEN

Antiviral restriction factors are structurally and functionally diverse cellular proteins that play a key role in the first line of defense against viral pathogens. Although many cell types constitutively express restriction factors at low levels, their induction in response to viral exposure and replication is often required for potent control and repulse of the invading pathogens. It is well established that type I IFNs efficiently induce antiviral restriction factors. Accumulating evidence suggests that other types of IFN, as well as specific cytokines, such as IL-27, and other activators of the cell are also capable of enhancing the expression of restriction factors and hence to establish an antiviral cellular state. Agents that efficiently induce restriction factors, increase their activity, and/or render them resistant against viral antagonists without causing general inflammation and significant side effects hold some promise for novel therapeutic or preventive strategies. In the present review, we summarize some of the current knowledge on the induction of antiretroviral restriction factors and perspectives for therapeutic application.


Asunto(s)
Antivirales/farmacología , Inmunidad Innata/inmunología , Interferones/farmacología , Virosis/tratamiento farmacológico , Animales , Humanos , Inmunidad Innata/efectos de los fármacos , Transducción de Señal , Virosis/inmunología , Virosis/virología , Replicación Viral
16.
Comput Biol Chem ; 73: 31-40, 2018 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-29413814

RESUMEN

Viral attack within host cells triggers the production of type I interferons and leads to the induction of interferon stimulated genes (ISGs). One of the ISG Mx, encodes type I interferon inducible GTPase that is responsible for the establishment of an anti-viral state within cells. Intriguingly, several isoforms of Mx have been reported in fish, but the structural analysis of fish Mx proteins remains unexplored. For the first time, we have identified and unraveled the molecular structure of Mx protein from Indian snow trout, Schizothorax richardsonii (Gray) a Coldwater fish that inhabits the water bodies in the sub-Himalayan region. The snow trout Mx coding region consists of 2518 nucleotides with an open reading frame (ORF) of 1854 nucleotides. It codes for a polypeptide of 617 amino acids with a predicted molecular weight of 70 kDa. In silico analysis of snow trout Mx protein revealed signature of dynamin family (LPRGTGIVTR) along with a tripartite GTP-binding domain (GDQSSGKS, DLPG, and TKPD). Homology modelling established that the Mx protein is an elongated structure with a G domain, bundle signaling element (BSE) and a GTPase effector domain (GED). Moreover, the GED of Mx contains two highly conserved leucine zippers at the COOH-terminal of the protein suggesting its structural similarity with human homologues. However, snow trout Mx lacks the essential features of its mammalian homologues questioning its functional characteristics. Further, a ligand binding site in the said protein has also been predicted adjacent to the GTPase switch within the G domain.


Asunto(s)
Interferón Tipo I/metabolismo , Proteínas de Resistencia a Mixovirus/metabolismo , Secuencia de Aminoácidos , Animales , Biología Computacional , Modelos Moleculares , Proteínas de Resistencia a Mixovirus/análisis , Proteínas de Resistencia a Mixovirus/genética , Filogenia , Trucha
17.
Viruses ; 9(2)2017 02 02.
Artículo en Inglés | MEDLINE | ID: mdl-28157174

RESUMEN

Myxoma virus (MYXV) is Leporipoxvirus that possesses a specific rabbit-restricted host tropism but exhibits a much broader  cellular host range in cultured cells. MYXV is able to efficiently  block all aspects of the type I interferon (IFN)-induced  antiviral  state  in rabbit cells, partially in  human  cells  and  very  poorly  in  mouse  cells.  The mechanism(s) of this species-specific inhibition of  type I IFN-induced antiviral state is not well understood. Here we demonstrate that MYXV encoded  protein  M029, a truncated relative of the vaccinia virus (VACV) E3 double-stranded RNA (dsRNA)  binding  protein  that  inhibits  protein  kinase  R (PKR),  can  also  antagonize the type I IFN-induced  antiviral state in a highly species-specific manner. In cells pre-treated with type I IFN prior to  infection,  MYXV  exploits  M029  to  overcome  the  induced  antiviral  state completely in rabbit cells,  partially  in  human  cells,  but  not at all in mouse cells. However, in cells pre-infected with MYXV,  IFN-induced  signaling  is fully  inhibited  even  in the  absence  of M029 in cells from all three species,  suggesting  that  other  MYXV  protein(s)  apart  from  M029  block  IFN  signaling  in  a  speciesindependent  manner.  We  also  show  that  the  antiviral  state  induced in rabbit, human or mouse cells  by  type  I IFN  can  inhibit M029-knockout MYXV even when PKR is genetically knocked-out, suggesting  that  M029  targets  other  host  proteins  for  this  antiviral state inhibition. Thus, the MYXV  dsRNA  binding  protein  M029  not  only  antagonizes  PKR  from  multiple  species  but  also blocks the  type I IFN antiviral state independently of PKR in a highly species-specific fashion.


Asunto(s)
Interacciones Huésped-Patógeno , Evasión Inmune , Interferón Tipo I/antagonistas & inhibidores , Myxoma virus/patogenicidad , Proteínas de Unión al ARN/metabolismo , Proteínas Virales/metabolismo , Animales , Línea Celular , Humanos , Ratones , Conejos
18.
J Interferon Cytokine Res ; 36(11): 652-665, 2016 11.
Artículo en Inglés | MEDLINE | ID: mdl-27508859

RESUMEN

Sendai virus (SeV), a murine paramyxovirus, has been used to study the induction of type I interferon (IFN) subtypes in robust quantities. Few studies have measured whether the IFN that SeV induces actually fulfills its intended purpose of interfering with virus-mediated effects in the cells in which it is produced. We determined the effects of IFN on SeV-mediated cytopathic effects (CPE) and the ability of IFN to protect against virus infection. SeV-induced biologically active IFN resulted in Jak/STAT activation and the production of a number of interferon-stimulated genes (ISGs). However, these responses did not inhibit SeV replication or CPE. This observation was not due to SeV effects on canonical IFN signaling. Furthermore, pretreating cells with type I IFN and establishing an antiviral state before infection did not mediate SeV effects. Therefore, the induction of canonical IFN signaling pathways and ISGs does not always confer protection against the IFN-inducing virus. Because type I IFNs are approved to treat various infections, our findings suggest that typical markers of IFN activity may not be indicative of a protective antiviral response and should not be used alone to determine whether an antiviral state against a particular virus is achieved.


Asunto(s)
Interferón Tipo I/inmunología , Quinasas Janus/genética , Infecciones por Respirovirus/genética , Infecciones por Respirovirus/virología , Factores de Transcripción STAT/genética , Virus Sendai/patogenicidad , Humanos , Quinasas Janus/metabolismo , Infecciones por Respirovirus/inmunología , Factores de Transcripción STAT/metabolismo , Virus Sendai/inmunología , Células Tumorales Cultivadas , Replicación Viral
19.
Virology ; 496: 59-66, 2016 09.
Artículo en Inglés | MEDLINE | ID: mdl-27254596

RESUMEN

The virus/host interaction is a complex interplay between pro- and anti-viral factors that ultimately determines the spread or halt of virus infections in tissues. This interplay develops over multiple rounds of infection. The purpose of this study was to determine how cellular-level processes combine to impact the spatial spread of infection. We measured the kinetics of virus replication (VSV), antiviral paracrine signal upregulation and secretion, spatial spread of virus and paracrine antiviral signaling, and inhibition of virus production in antiviral-exposed A549 human lung epithelial cells. We found that initially infected cells released antiviral signals 4-to-7h following production of virus. However, the subsequent rapid dissemination of signal and fast induction of a robust and persistent antiviral state ultimately led to a suppression of infection spread. This work shows how cellular responses to infection and activation of antiviral responses can integrate to ultimately control infection spread across host cell populations.


Asunto(s)
Puntos de Control del Ciclo Celular , Interacciones Huésped-Patógeno , Comunicación Paracrina , Fenómenos Fisiológicos de los Virus , Replicación Viral , Células A549 , Antivirales/metabolismo , Células Cultivadas , Citocinas/metabolismo , Interacciones Huésped-Patógeno/inmunología , Humanos , Inmunidad Innata
20.
Mol Immunol ; 76: 55-61, 2016 08.
Artículo en Inglés | MEDLINE | ID: mdl-27348633

RESUMEN

The iIFN1a (intracellular IFN-a1), that is one of the IFN-a1 variants, was shown to be functional intracellularly and act as a novel defense against an infectious hematopoietic necrosis virus (IHNV). To determine its antiviral properties, a recombinant iIFN1a was generated in Escherichia coli. Its antiviral activity against IHNV was 1.69×10(7)U/mg in CHSE-214 cells. Additionally, iIFN1a was capable of inducing comparable levels of IRF-1, IRF-2, IFN-I, IFN-γ and Mx transcription in head kidney, spleen and liver tissues at an early time point (6h), that was followed by a rapid decline 24h after induction. The recombinant protein also elicited protection against IHNV in vivo. At 6 and 24h after induction there was 100% protection against the virus, however, at 48 and 72h the protection decreased to 57 and 40%, respectively. The in vivo protection kinetics correlated with the kinetics of gene expression. The results of this study provide details of the antiviral state that was induced by iIFN1a in vivo for the first time. Additionally, this information will facilitate the development of this recombinant protein as a potential anti-viral treatment and/or adjuvant.


Asunto(s)
Enfermedades de los Peces/inmunología , Interferón-alfa/inmunología , Oncorhynchus mykiss/inmunología , Infecciones por Rhabdoviridae/veterinaria , Animales , Electroforesis en Gel de Poliacrilamida , Virus de la Necrosis Hematopoyética Infecciosa , Cinética , Oncorhynchus mykiss/virología , Reacción en Cadena de la Polimerasa , Proteínas Recombinantes/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA