Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 74
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
J Biol Chem ; 300(4): 107200, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38508315

RESUMEN

Interferon (IFN) regulatory factors (IRF) are key transcription factors in cellular antiviral responses. IRF7, a virus-inducible IRF, expressed primarily in myeloid cells, is required for transcriptional induction of interferon α and antiviral genes. IRF7 is activated by virus-induced phosphorylation in the cytoplasm, leading to its translocation to the nucleus for transcriptional activity. Here, we revealed a nontranscriptional activity of IRF7 contributing to its antiviral functions. IRF7 interacted with the pro-inflammatory transcription factor NF-κB-p65 and inhibited the induction of inflammatory target genes. Using knockdown, knockout, and overexpression strategies, we demonstrated that IRF7 inhibited NF-κB-dependent inflammatory target genes, induced by virus infection or toll-like receptor stimulation. A mutant IRF7, defective in transcriptional activity, interacted with NF-κB-p65 and suppressed NF-κB-induced gene expression. A single-action IRF7 mutant, active in anti-inflammatory function, but defective in transcriptional activity, efficiently suppressed Sendai virus and murine hepatitis virus replication. We, therefore, uncovered an anti-inflammatory function for IRF7, independent of transcriptional activity, contributing to the antiviral response of IRF7.


Asunto(s)
Factor 7 Regulador del Interferón , FN-kappa B , Animales , Humanos , Ratones , Células HEK293 , Inflamación/genética , Factor 7 Regulador del Interferón/genética , Factor 7 Regulador del Interferón/inmunología , FN-kappa B/genética , FN-kappa B/inmunología , Virus Sendai/fisiología , Factor de Transcripción ReIA/genética , Factor de Transcripción ReIA/inmunología , Replicación Viral , Mutación , Regulación de la Expresión Génica/genética , Virus de la Hepatitis Murina/fisiología , Infecciones por Coronavirus/inmunología , Infecciones por Respirovirus/inmunología
2.
Immunity ; 44(5): 1151-61, 2016 05 17.
Artículo en Inglés | MEDLINE | ID: mdl-27178468

RESUMEN

The transcription factor IRF-3 mediates cellular antiviral response by inducing the expression of interferon and other antiviral proteins. In RNA-virus infected cells, IRF-3's transcriptional activation is triggered primarily by RIG-I-like receptors (RLR), which can also activate the RLR-induced IRF-3-mediated pathway of apoptosis (RIPA). Here, we have reported that the pathway of IRF-3 activation in RIPA was independent of and distinct from the known pathway of transcriptional activation of IRF-3. It required linear polyubiquitination of two specific lysine residues of IRF-3 by LUBAC, the linear polyubiquitinating enzyme complex, which bound IRF-3 in signal-dependent fashion. To evaluate the role of RIPA in viral pathogenesis, we engineered a genetically targeted mouse, which expressed a mutant IRF-3 that was RIPA-competent but transcriptionally inert; this single-action IRF-3 could protect mice from lethal viral infection. Our observations indicated that IRF-3-mediated apoptosis of virus-infected cells could be an effective antiviral mechanism, without expression of the interferon-stimulated genes.


Asunto(s)
Apoptosis , Fibroblastos/inmunología , Factor 3 Regulador del Interferón/metabolismo , Proteínas de la Membrana/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Infecciones por Virus ARN/inmunología , Animales , Línea Celular , Clonación Molecular , Fibroblastos/virología , Humanos , Factor 3 Regulador del Interferón/genética , Ratones , Ratones Transgénicos , Mutación/genética , Receptores de Superficie Celular , Transducción de Señal , Activación Transcripcional , Ubiquitinación
3.
Proc Natl Acad Sci U S A ; 119(37): e2121385119, 2022 09 13.
Artículo en Inglés | MEDLINE | ID: mdl-36067309

RESUMEN

Interferon (IFN) regulatory factor 3 (IRF3) is a transcription factor activated by phosphorylation in the cytoplasm of a virus-infected cell; by translocating to the nucleus, it induces transcription of IFN-ß and other antiviral genes. We have previously reported IRF3 can also be activated, as a proapoptotic factor, by its linear polyubiquitination mediated by the RIG-I pathway. Both transcriptional and apoptotic functions of IRF3 contribute to its antiviral effect. Here, we report a nontranscriptional function of IRF3, namely, the repression of IRF3-mediated NF-κB activity (RIKA), which attenuated viral activation of NF-κB and the resultant inflammatory gene induction. In Irf3-/- mice, consequently, Sendai virus infection caused enhanced inflammation in the lungs. Mechanistically, RIKA was mediated by the direct binding of IRF3 to the p65 subunit of NF-κB in the cytoplasm, which prevented its nuclear import. A mutant IRF3 defective in both the transcriptional and the apoptotic activities was active in RIKA and inhibited virus replication. Our results demonstrated IRF3 deployed a three-pronged attack on virus replication and the accompanying inflammation.


Asunto(s)
Inmunidad Innata , Factor 3 Regulador del Interferón , FN-kappa B , Neumonía Viral , Transporte Activo de Núcleo Celular , Animales , Núcleo Celular/metabolismo , Expresión Génica , Factor 3 Regulador del Interferón/genética , Factor 3 Regulador del Interferón/metabolismo , Interferón beta/genética , Ratones , FN-kappa B/metabolismo , Neumonía Viral/genética , Neumonía Viral/inmunología , Virus Sendai
4.
EMBO J ; 39(22): e104106, 2020 11 16.
Artículo en Inglés | MEDLINE | ID: mdl-32926474

RESUMEN

STING (STimulator of INterferon Genes) mediates protective cellular response to microbial infection and tissue damage, but its aberrant activation can lead to autoinflammatory diseases. Upon ligand stimulation, the endoplasmic reticulum (ER) protein STING translocates to endosomes for induction of interferon production, while an alternate trafficking route delivers it directly to the autophagosomes. Here, we report that phosphorylation of a specific tyrosine residue in STING by the epidermal growth factor receptor (EGFR) is required for directing STING to endosomes, where it interacts with its downstream effector IRF3. In the absence of EGFR-mediated phosphorylation, STING rapidly transits into autophagosomes, and IRF3 activation, interferon production, and antiviral activity are compromised in cell cultures and mice, while autophagic activity is enhanced. Our observations illuminate a new connection between the tyrosine kinase activity of EGFR and innate immune functions of STING and suggest new experimental and therapeutic approaches for selective regulation of STING functions.


Asunto(s)
Receptores ErbB/metabolismo , Inmunidad Innata , Proteínas de la Membrana/metabolismo , Transporte de Proteínas/fisiología , Tirosina/metabolismo , Animales , Línea Celular , Retículo Endoplásmico/metabolismo , Endosomas/metabolismo , Receptores ErbB/genética , Técnicas de Silenciamiento del Gen , Células HeLa , Humanos , Inmunidad Innata/genética , Factor 3 Regulador del Interferón/genética , Masculino , Proteínas de la Membrana/genética , Ratones , Ratones Endogámicos C57BL , Fosforilación , Células RAW 264.7 , Transducción de Señal , Transcriptoma
5.
Proc Natl Acad Sci U S A ; 118(34)2021 08 24.
Artículo en Inglés | MEDLINE | ID: mdl-34408018

RESUMEN

Inflammatory arthritis (IA) is a common disease that affects millions of individuals worldwide. Proinflammatory events during IA pathogenesis are well studied; however, loss of protective immunity remains underexplored. Earlier, we reported that 14-3-3zeta (ζ) has a role in T-cell polarization and interleukin (IL)-17A signal transduction. Here, we demonstrate that 14-3-3ζ knockout (KO) rats develop early-onset severe arthritis in two independent models of IA, pristane-induced arthritis and collagen-induced arthritis. Arthritic 14-3-3ζ KO animals showed an increase in bone loss and immune cell infiltration in synovial joints. Induction of arthritis coincided with the loss of anti-14-3-3ζ antibodies; however, rescue experiments to supplement the 14-3-3ζ antibody by passive immunization did not suppress arthritis. Instead, 14-3-3ζ immunization during the presymptomatic phase resulted in significant suppression of arthritis in both wild-type and 14-3-3ζ KO animals. Mechanistically, 14-3-3ζ KO rats exhibited elevated inflammatory gene signatures at the messenger RNA and protein levels, particularly for IL-1ß. Furthermore, the immunization with recombinant 14-3-3ζ protein suppressed IL-1ß levels, significantly increased anti-14-3-3ζ antibody levels and collagen production, and preserved bone quality. The 14-3-3ζ protein increased collagen expression in primary rat mesenchymal cells. Together, our findings indicate that 14-3-3ζ causes immune suppression and extracellular remodeling, which lead to a previously unrecognized IA-suppressive function.


Asunto(s)
Proteínas 14-3-3/metabolismo , Proteínas 14-3-3/farmacología , Artritis/inducido químicamente , Inflamación/tratamiento farmacológico , Proteínas 14-3-3/genética , Proteínas 14-3-3/inmunología , Animales , Anticuerpos , Artritis/genética , Artritis/metabolismo , Densidad Ósea , Enfermedades Óseas/metabolismo , Enfermedades Óseas/prevención & control , Colágeno/metabolismo , Colágeno/toxicidad , Femenino , Adyuvante de Freund/farmacología , Eliminación de Gen , Regulación de la Expresión Génica/efectos de los fármacos , Regulación de la Expresión Génica/inmunología , Inmunización Pasiva , Masculino , Células Madre Mesenquimatosas/efectos de los fármacos , ARN Mensajero/genética , ARN Mensajero/metabolismo , Ratas , Terpenos/toxicidad
6.
Proc Natl Acad Sci U S A ; 117(40): 25008-25017, 2020 10 06.
Artículo en Inglés | MEDLINE | ID: mdl-32968020

RESUMEN

IL-17A is a therapeutic target in many autoimmune diseases. Most nonhematopoietic cells express IL-17A receptors and respond to extracellular IL-17A by inducing proinflammatory cytokines. The IL-17A signal transduction triggers two broad, TRAF6- and TRAF5-dependent, intracellular signaling pathways to produce representative cytokines (IL-6) and chemokines (CXCL-1), respectively. Our limited understanding of the cross-talk between these two branches has generated a crucial gap of knowledge, leading to therapeutics indiscriminately blocking IL-17A and global inhibition of its target genes. In previous work, we discovered an elevated expression of 14-3-3 proteins in inflammatory aortic disease, a rare human autoimmune disorder with increased levels of IL-17A. Here we report that 14-3-3ζ is essential for IL-17 signaling by differentially regulating the signal-induced IL-6 and CXCL-1. Using genetically manipulated human and mouse cells, and ex vivo and in vivo rat models, we uncovered a function of 14-3-3ζ. As a part of the molecular mechanism, we show that 14-3-3ζ interacts with several TRAF proteins; in particular, its interaction with TRAF5 and TRAF6 is increased in the presence of IL-17A. In contrast to TRAF6, we found TRAF5 to be an endogenous suppressor of IL-17A-induced IL-6 production, an effect countered by 14-3-3ζ. Furthermore, we observed that 14-3-3ζ interaction with TRAF proteins is required for the IL-17A-induced IL-6 levels. Together, our results show that 14-3-3ζ is an essential component of IL-17A signaling and IL-6 production, an effect that is suppressed by TRAF5. To the best of our knowledge, this report of the 14-3-3ζ-TRAF5 axis, which differentially regulates IL-17A-induced IL-6 and CXCL-1 production, is unique.


Asunto(s)
Enfermedades Autoinmunes/genética , Quimiocina CXCL1/genética , Interleucina-17/genética , Interleucina-6/genética , Proteínas 14-3-3/genética , Animales , Enfermedades Autoinmunes/patología , Quimiocinas/genética , Citocinas/genética , Regulación de la Expresión Génica/genética , Humanos , Ratones , Ratas , Transducción de Señal/genética , Factor 5 Asociado a Receptor de TNF/genética , Factor 6 Asociado a Receptor de TNF/genética
7.
J Public Health Manag Pract ; 29(6): 845-853, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37738597

RESUMEN

CONTEXT: Prior to the COVID-19 pandemic, wastewater influent monitoring for tracking disease burden in sewered communities was not performed in Ohio, and this field was only on the periphery of the state academic research community. PROGRAM: Because of the urgency of the pandemic and extensive state-level support for this new technology to detect levels of community infection to aid in public health response, the Ohio Water Resources Center established relationships and support of various stakeholders. This enabled Ohio to develop a statewide wastewater SARS-CoV-2 (severe acute respiratory syndrome coronavirus 2) monitoring network in 2 months starting in July 2020. IMPLEMENTATION: The current Ohio Coronavirus Wastewater Monitoring Network (OCWMN) monitors more than 70 unique locations twice per week, and publicly available data are updated weekly on the public dashboard. EVALUATION: This article describes the process and decisions that were made during network initiation, the network progression, and data applications, which can inform ongoing and future pandemic response and wastewater monitoring. DISCUSSION: Overall, the OCWMN established wastewater monitoring infrastructure and provided a useful tool for public health professionals responding to the pandemic.


Asunto(s)
COVID-19 , Aguas Residuales , Humanos , Ohio , Pandemias/prevención & control , Salud Pública , COVID-19/epidemiología , COVID-19/prevención & control , SARS-CoV-2
8.
J Biol Chem ; 297(5): 101274, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34619149

RESUMEN

The ubiquitously expressed transcription factor interferon (IFN) regulatory factor 3 (IRF3) is critical for the induction of antiviral genes, e.g., type-I IFN. In addition to its transcriptional function, IRF3 also activates a nontranscriptional, proapoptotic signaling pathway. While the proapoptotic function of IRF3 protects against viral infections, it is also involved in harmful immune responses that trigger hepatocyte cell death and promote liver disease. Thus, we hypothesized that a small-molecule inhibitor of the proapoptotic activity of IRF3 could alleviate fatty-acid-induced hepatocyte cell death. We conducted a high-throughput screen, which identified auranofin as a small-molecule inhibitor of the proapoptotic activity of IRF3. In addition to the nontranscriptional apoptotic pathway, auranofin also inhibited the transcriptional activity of IRF3. Using biochemical and genetic tools in human and mouse cells, we uncovered a novel mechanism of action for auranofin, in which it induces cellular autophagy to degrade IRF3 protein, thereby suppressing IRF3 functions. Autophagy-deficient cells were unable to degrade IRF3 upon auranofin treatment, suggesting that the autophagic degradation of IRF3 is a novel approach to regulate IRF3 activities. Using a physiologically relevant in vitro model, we demonstrated that auranofin inhibited fatty-acid-induced apoptotic cell death of hepatocytes. In summary, auranofin is a novel inhibitor of IRF3 functions and may represent a potential therapeutic option in diseases where IRF3 is deleterious.


Asunto(s)
Apoptosis/efectos de los fármacos , Auranofina/farmacología , Autofagia/efectos de los fármacos , Factor 3 Regulador del Interferón/metabolismo , Proteolisis/efectos de los fármacos , Transcripción Genética/efectos de los fármacos , Animales , Humanos , Factor 3 Regulador del Interferón/genética , Ratones , Células RAW 264.7
9.
Mol Cell ; 54(2): 210-1, 2014 Apr 24.
Artículo en Inglés | MEDLINE | ID: mdl-24766884

RESUMEN

Cellular interferon response to microbial infection is transient. In a recent paper in Immunity, Long et al. (2014) identify protein phosphatase 2A (PP2A) as a deactivator of phospho-interferon regulatory factor 3, the key transcription factor for interferon synthesis, thus providing one basis for the observed transiency.


Asunto(s)
Factor 3 Regulador del Interferón/metabolismo , Interferón Tipo I/metabolismo , Neuropéptidos/metabolismo , Proteína Fosfatasa 2/metabolismo , Receptor Toll-Like 3/metabolismo , Receptor Toll-Like 4/metabolismo , Animales , Humanos , Receptores de Cinasa C Activada
10.
J Biol Chem ; 295(20): 6811-6822, 2020 05 15.
Artículo en Inglés | MEDLINE | ID: mdl-32273341

RESUMEN

The interferon system is the first line of defense against virus infection. Recently, using a high-throughput genetic screen of a human interferon-stimulated gene short-hairpin RNA library, we identified a viral restriction factor, TDRD7 (Tudor domain-containing 7). TDRD7 inhibits the paramyxo-/pneumoviruses (e.g. Sendai virus and respiratory syncytial virus) by interfering with the virus-induced cellular autophagy pathway, which these viruses use for their replication. Here, we report that TDRD7 is a viral restriction factor against herpes simplex virus (HSV-1). Using knockdown, knockout, and ectopic expression systems, we demonstrate the anti-HSV-1 activity of TDRD7 in multiple human and mouse cell types. TDRD7 inhibited the virus-activated AMP-activated protein kinase (AMPK), which was essential for HSV-1 replication. Genetic ablation or chemical inhibition of AMPK activity suppressed HSV-1 replication in multiple human and mouse cells. Mechanistically, HSV-1 replication after viral entry depended on AMPK but not on its function in autophagy. The antiviral activity of TDRD7 depended on its ability to inhibit virus-activated AMPK. In summary, our results indicate that the newly identified viral restriction factor TDRD7 inhibits AMPK and thereby blocks HSV-1 replication independently of the autophagy pathway. These findings suggest that AMPK inhibition represents a potential strategy to manage HSV-1 infections.


Asunto(s)
Proteínas Quinasas Activadas por AMP/metabolismo , Autofagia , Herpesvirus Humano 1/fisiología , Ribonucleoproteínas/metabolismo , Replicación Viral , Proteínas Quinasas Activadas por AMP/genética , Animales , Chlorocebus aethiops , Células HeLa , Humanos , Ratones , Ribonucleoproteínas/genética , Células Vero
11.
PLoS Pathog ; 14(1): e1006877, 2018 01.
Artículo en Inglés | MEDLINE | ID: mdl-29381763

RESUMEN

The interferon (IFN) system represents the first line of defense against a wide range of viruses. Virus infection rapidly triggers the transcriptional induction of IFN-ß and IFN Stimulated Genes (ISGs), whose protein products act as viral restriction factors by interfering with specific stages of virus life cycle, such as entry, transcription, translation, genome replication, assembly and egress. Here, we report a new mode of action of an ISG, IFN-induced TDRD7 (tudor domain containing 7) inhibited paramyxovirus replication by inhibiting autophagy. TDRD7 was identified as an antiviral gene by a high throughput screen of an ISG shRNA library for blocking IFN's protective effect against Sendai virus (SeV) replication. The antiviral activity of TDRD7 against SeV, human parainfluenza virus 3 and respiratory syncytial virus was confirmed by its genetic ablation or ectopic expression in several types of mouse and human cells. TDRD7's antiviral action was mediated by its ability to inhibit autophagy, a cellular catabolic process which was robustly induced by SeV infection and required for its replication. Mechanistic investigation revealed that TDRD7 interfered with the activation of AMP-dependent kinase (AMPK), an enzyme required for initiating autophagy. AMPK activity was required for efficient replication of several paramyxoviruses, as demonstrated by its genetic ablation or inhibition of its activity by TDRD7 or chemical inhibitors. Therefore, our study has identified a new antiviral ISG with a new mode of action.


Asunto(s)
Antivirales/farmacología , Autofagia , Interferones/farmacología , Paramyxovirinae/fisiología , Ribonucleoproteínas/fisiología , Replicación Viral/efectos de los fármacos , Animales , Autofagia/genética , Autofagia/inmunología , Células Cultivadas , Regulación de la Expresión Génica/efectos de los fármacos , Células HEK293 , Células HeLa , Humanos , Inmunidad Innata/efectos de los fármacos , Inmunidad Innata/genética , Ratones , Ratones Endogámicos C57BL , Ribonucleoproteínas/genética , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética , Replicación Viral/genética
12.
J Immunol ; 200(8): 2809-2818, 2018 04 15.
Artículo en Inglés | MEDLINE | ID: mdl-29531172

RESUMEN

Mammalian TLRs recognize microbial infection or cell death-associated danger signals and trigger the appropriate cellular response. These responses determine the strength and the outcome of the host-microbe interaction. TLRs are transmembrane proteins located on the plasma or the endosomal membrane. Their ectodomains recognize specific microbial or endogenous ligands, and the cytoplasmic domains interact with specific proteins to activate intracellular signaling pathways. TLR9, an endosomal TLR, is activated by endocytosed DNA. Activated TLR9 recruits the cytoplasmic adapter MyD88 and other signaling proteins to induce the synthesis of inflammatory cytokines and IFN. Uncontrolled activation of TLR9 leads to the undesired overproduction of inflammatory cytokines and consequent pathogenesis. Therefore, appropriate activation and the regulation of TLR9 signaling are critical. Tyrosine (Tyr) phosphorylation of TLR9 is essential for its activation; however, the role of specific Tyr kinases is not clear. In this article, we report that epidermal growth factor receptor (EGFR), a membrane-bound protein Tyr kinase, is essential for TLR9 signaling. Genetic ablation of EGFR or pharmacological inhibition of its kinase activity attenuates TLR9-mediated induction of genes in myeloid and nonmyeloid cell types. EGFR is constitutively bound to TLR9; upon ligand stimulation, it mediates TLR9 Tyr phosphorylation, which leads to the recruitment of MyD88, activation of the signaling kinases and transcription factors, and gene induction. In mice, TLR9-mediated liver injury and death are blocked by an EGFR inhibitor or deletion of the EGFR gene from myeloid cells, which are the major producers of inflammatory cytokines.


Asunto(s)
Receptores ErbB/metabolismo , Macrófagos/metabolismo , Transducción de Señal/inmunología , Receptor Toll-Like 9/metabolismo , Animales , Receptores ErbB/inmunología , Humanos , Macrófagos/inmunología , Ratones , Fosforilación , Receptor Toll-Like 9/inmunología , Tirosina/metabolismo
13.
J Hepatol ; 70(5): 974-984, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-30710579

RESUMEN

BACKGROUND & AIMS: Interferon regulatory factor 3 (IRF3) is a transcription factor mediating antiviral responses, yet recent evidence indicates that IRF3 also has critical non-transcriptional functions, including activating RIG-I-like receptors-induced IRF-3-mediated pathway of apoptosis (RIPA) and restricting activity of NF-κB. Using a novel murine model expressing only non-transcriptional IRF3 activity (Irf3S1/S1), we tested the hypothesis that non-transcriptional functions of IRF3 modulate innate immune responses in the Gao-binge (acute-on-chronic) model of alcohol-related liver disease. METHODS: IRF3 and IRF3-mediated signals were analysed in liver samples from 5 patients transplanted for alcoholic hepatitis and 5 healthy controls. C57BL/6, Irf3-/- and Irf3S1/S1 mice were exposed to Gao-binge ethanol-induced liver injury. IRF3-mediated RIPA was investigated in cultured macrophages. RESULTS: Phospho-IRF3 and IRF3-mediated signals were elevated in livers of patients with alcoholic hepatitis. In C57BL/6 mice, Gao-binge ethanol exposure activated IRF3 signaling and resulted in hepatocellular injury. Indicators of liver injury were differentially impacted by Irf3 genotype. Irf3-/-, but not Irf3S1/S1, mice were protected from steatosis, elevated alanine/aspartate aminotransferase levels and inflammatory cytokine expression. In contrast, neutrophil accumulation and endoplasmic reticulum stress were independent of genotype. Protection from Gao-binge injury in Irf3-/- mice was associated with an increased ratio of Ly6Clow (restorative) to Ly6Chigh (inflammatory) cells compared to C57BL/6 and Irf3S1/S1 mice. Reduced ratios of Ly6Clow/Ly6Chigh in C57BL/6 and Irf3S1/S1 mice were associated with increased apoptosis in the Ly6Clow population in response to Gao-binge. Activation of primary macrophage cultures with Poly (I:C) induced translocation of IRF3 to the mitochondria, where it associated with Bax and activated caspases 3 and 9, processes indicative of activation of the RIPA pathway. CONCLUSIONS: Taken together, these data identify that the non-transcriptional function of IRF3 plays an important role in modulating the innate immune environment in response to Gao-binge ethanol exposure, via regulation of immune cell apoptosis. LAY SUMMARY: Activation of the innate immune system contributes to inflammation in the progression of alcohol-related liver disease, as well as to the resolution of injury. Here we show that the protein IRF3 modulates the innate immune environment of the liver in a mouse model of alcoholic hepatitis. It does this by increasing the apoptotic cell death of immune cells that promote the resolution of injury.


Asunto(s)
Hepatitis Alcohólica/inmunología , Factor 3 Regulador del Interferón/fisiología , Hígado/inmunología , Animales , Apoptosis , Modelos Animales de Enfermedad , Estrés del Retículo Endoplásmico , Hepatitis Alcohólica/etiología , Humanos , Inmunidad Innata , Hígado/patología , Activación de Macrófagos , Ratones , Ratones Endogámicos C57BL , Monocitos/fisiología , Neutrófilos/fisiología , Transcripción Genética
14.
PLoS Pathog ; 11(3): e1004779, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25811886

RESUMEN

Innate immunity is the first line of defense against microbial insult. The transcription factor, IRF3, is needed by mammalian cells to mount innate immune responses against many microbes, especially viruses. IRF3 remains inactive in the cytoplasm of uninfected cells; upon virus infection, it gets phosphorylated and then translocates to the nucleus, where it binds to the promoters of antiviral genes and induces their expression. Such genes include type I interferons (IFNs) as well as Interferon Stimulated Genes (ISGs). IRF3-/- cells support enhanced replication of many viruses and therefore, the corresponding mice are highly susceptible to viral pathogenesis. Here, we provide evidence for an unexpected pro-microbial role of IRF3: the replication of the protozoan parasite, Toxoplasma gondii, was significantly impaired in IRF3-/- cells. In exploring whether the transcriptional activity of IRF3 was important for its pro-parasitic function, we found that ISGs induced by parasite-activated IRF3 were indeed essential, whereas type I interferons were not important. To delineate the signaling pathway that activates IRF3 in response to parasite infection, we used genetically modified human and mouse cells. The pro-parasitic signaling pathway, which we termed PISA (Parasite-IRF3 Signaling Activation), activated IRF3 without any involvement of the Toll-like receptor or RIG-I-like receptor pathways, thereby ruling out a role of parasite-derived RNA species in activating PISA. Instead, PISA needed the presence of cGAS, STING, TBK1 and IRF3, indicating the necessity of DNA-triggered signaling. To evaluate the physiological significance of our in vitro findings, IRF3-/- mice were challenged with parasite infection and their morbidity and mortality were measured. Unlike WT mice, the IRF3-/- mice did not support replication of the parasite and were resistant to pathogenesis caused by it. Our results revealed a new paradigm in which the antiviral host factor, IRF3, plays a cell-intrinsic pro-parasitic role.


Asunto(s)
Factor 3 Regulador del Interferón/inmunología , Transducción de Señal/inmunología , Toxoplasma/inmunología , Toxoplasmosis/inmunología , Animales , Proteína 58 DEAD Box , ARN Helicasas DEAD-box/genética , ARN Helicasas DEAD-box/inmunología , Técnicas de Silenciamiento del Gen , Células HEK293 , Humanos , Factor 3 Regulador del Interferón/genética , Proteínas de la Membrana/genética , Proteínas de la Membrana/inmunología , Ratones , Ratones Noqueados , Proteínas Serina-Treonina Quinasas/genética , Proteínas Serina-Treonina Quinasas/inmunología , Receptores Inmunológicos , Transducción de Señal/genética , Toxoplasmosis/genética
15.
EMBO Rep ; 16(11): 1535-47, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26341626

RESUMEN

Mammalian Toll-like receptors (TLR) recognize microbial products and elicit transient immune responses that protect the infected host from disease. TLR4--which signals from both plasma and endosomal membranes--is activated by bacterial lipopolysaccharides (LPS) and induces many cytokine genes, the prolonged expression of which causes septic shock in mice. We report here that the expression of some TLR4-induced genes in myeloid cells requires the protein kinase activity of the epidermal growth factor receptor (EGFR). EGFR inhibition affects TLR4-induced responses differently depending on the target gene. The induction of interferon-ß (IFN-ß) and IFN-inducible genes is strongly inhibited, whereas TNF-α induction is enhanced. Inhibition is specific to the IFN-regulatory factor (IRF)-driven genes because EGFR is required for IRF activation downstream of TLR--as is IRF co-activator ß-catenin--through the PI3 kinase/AKT pathway. Administration of an EGFR inhibitor to mice protects them from LPS-induced septic shock and death by selectively blocking the IFN branch of TLR4 signaling. These results demonstrate a selective regulation of TLR4 signaling by EGFR and highlight the potential use of EGFR inhibitors to treat septic shock syndrome.


Asunto(s)
Receptores ErbB/metabolismo , Choque Séptico/metabolismo , Transducción de Señal , Receptor Toll-Like 4/metabolismo , Animales , Receptores ErbB/antagonistas & inhibidores , Receptores ErbB/genética , Gefitinib , Factores Reguladores del Interferón/metabolismo , Interferón beta/genética , Interferón beta/metabolismo , Lipopolisacáridos/administración & dosificación , Ratones , Análisis por Micromatrices , Células Mieloides/metabolismo , FN-kappa B/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Inhibidores de Proteínas Quinasas/farmacología , Quinazolinas/farmacología , Células RAW 264.7 , Choque Séptico/prevención & control , Choque Séptico/terapia , Transducción de Señal/efectos de los fármacos , Factor de Necrosis Tumoral alfa/genética , Factor de Necrosis Tumoral alfa/metabolismo
16.
Physiol Genomics ; 52(5): 217-221, 2020 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-32275178
17.
EMBO J ; 29(10): 1762-73, 2010 May 19.
Artículo en Inglés | MEDLINE | ID: mdl-20360684

RESUMEN

Upon infection with many RNA viruses, the cytoplasmic retinoic acid inducible gene-I (RIG-I) pathway activates the latent transcription factor IRF-3, causing its nuclear translocation and the induction of many antiviral genes, including those encoding interferons. Here, we report a novel and distinct activity of IRF-3, in virus-infected cells, that induces apoptosis. Using genetically defective mouse and human cell lines, we demonstrated that, although both pathways required the presence of RIG-I, IPS1, TRAF3 and TBK1, only the apoptotic pathway required the presence of TRAF2 and TRAF6 in addition. More importantly, transcriptionally inactive IRF-3 mutants, such as the one missing its DNA-binding domain, could efficiently mediate apoptosis. Apoptosis was triggered by the direct interaction of IRF-3, through a newly identified BH3 domain, with the pro-apoptotic protein Bax, their co-translocation to the mitochondria and the resulting activation of the mitochondrial apoptotic pathway. Thus, IRF-3 is a dual-action cytoplasmic protein that, upon activation, translocates to the nucleus or to the mitochondrion and triggers two complementary antiviral responses of the infected cell.


Asunto(s)
Apoptosis , Regulación Viral de la Expresión Génica , Factor 3 Regulador del Interferón/metabolismo , Proteína X Asociada a bcl-2/metabolismo , Transporte Activo de Núcleo Celular , Animales , Línea Celular , Citoplasma/metabolismo , Humanos , Ratones , Transporte de Proteínas , ARN Bicatenario/metabolismo , Tretinoina/metabolismo
18.
J Virol ; 87(1): 16-24, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-23077293

RESUMEN

Infection of cultured cells by paramyxoviruses causes cell death, mediated by a newly discovered apoptotic pathway activated by virus infection. The key proapoptotic protein in this pathway is interferon regulatory factor 3 (IRF-3), which upon activation by virus infection binds BAX, translocates it to mitochondria, and triggers apoptosis. When IRF-3-knockdown cells were infected with Sendai virus (SeV), persistent infection (PI) was established. The PI cells produced infectious SeV continuously and constitutively expressed many innate immune genes. Interferon signaling was blocked in these cells. The elevated levels of IRF-3-driven genes in the PI cells indicated that the amount of residual IRF-3 activated by endogenous SeV was high enough to drive the transcriptional effects of IRF-3 but too low to trigger its apoptotic activity. We confirmed this IRF-3 threshold idea by generating a tetracycline (Tet)-inducible cell line for IRF-3 expression, which enabled us to express various levels of IRF-3. PI could be established in the Tet-off cell line, and as expected, when doxycycline was withdrawn, the cells underwent apoptosis. Finally, we tested for PI establishment in 12 mouse embryo fibroblasts by natural selection. Eleven lines became persistently infected; although seven out of them had low IRF-3 levels, four did not. When one of the latter four was further analyzed, we observed that it expressed a very low level of caspase 3, the final executor protease of the apoptotic pathway. These results demonstrated that SeV PI can arise from infection of normal wild-type cells, but only if they can find a way to impair the IRF-3-dependent apoptotic pathway.


Asunto(s)
Apoptosis , Interacciones Huésped-Patógeno , Factor 3 Regulador del Interferón/metabolismo , Virus Sendai/patogenicidad , Animales , Línea Celular , Fibroblastos/virología , Humanos , Ratones , Replicación Viral
19.
J Immunol ; 188(6): 2825-33, 2012 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-22323545

RESUMEN

dsRNA is a common pathogen-associated molecular pattern that is recognized by cellular TLR3 and used by virus-infected cells to activate specific transcription factors and trigger induction of antiviral genes. In this article, we report a new branch of TLR3 signaling that does not lead to gene induction but affects many cellular properties, such as cell migration, adhesion, and proliferation. We demonstrated that the migration of multiple cell lineages was affected by dsRNA treatment or influenza virus infection in a TLR3-dependent fashion. Surprisingly, for this effect of TLR3 signaling, the adaptor proteins, TRIF and MyD88, were not required. The effects of the new pathway were mediated by the proto-oncoprotein c-Src, which bound to TLR3 after dsRNA stimulation of cells. The response was biphasic: upon dsRNA treatment, we observed an immediate increase in cell motility followed by its strong inhibition. Our results indicate that the first phase was mediated by dsRNA-induced phosphorylation and activation of Src, whereas the second phase resulted from the sequestration of activated Src in lipid rafts, thus decreasing its active cytoplasmic pool. As expected, two other functions of Src, its effect on cell adhesion and cell proliferation, were also inhibited by dsRNA treatment. These results demonstrate that activated TLR3 can engage Src to trigger multiple cellular effects and reveal a possible link between innate immune response and cell growth regulation. This study also provides a rare example of TLR-mediated cellular effects that do not require gene induction and the first example, to our knowledge, of an adaptor-independent effect of any TLR.


Asunto(s)
Transducción de Señal/inmunología , Receptor Toll-Like 3/metabolismo , Virosis/inmunología , Proteínas Adaptadoras del Transporte Vesicular/inmunología , Proteínas Adaptadoras del Transporte Vesicular/metabolismo , Animales , Adhesión Celular/inmunología , Movimiento Celular/inmunología , Proliferación Celular , Células HEK293 , Humanos , Immunoblotting , Inmunoprecipitación , Microdominios de Membrana/inmunología , Microdominios de Membrana/metabolismo , Ratones , ARN Bicatenario/inmunología , ARN Viral/inmunología , Receptor Toll-Like 3/inmunología
20.
Viruses ; 16(3)2024 03 14.
Artículo en Inglés | MEDLINE | ID: mdl-38543816

RESUMEN

Interferons (IFNs) are cytokines that inhibit viral replication in host cells by triggering innate immune responses through the transcriptional induction of various IFN-stimulated genes (ISGs) [...].


Asunto(s)
Interferones , Virosis , Humanos , Interferones/genética , Citocinas , Inmunidad Innata , Replicación Viral
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA