Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 42
Filtrar
Más filtros

País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Nat Immunol ; 20(12): 1610-1620, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31740798

RESUMEN

The initial response to viral infection is anticipatory, with host antiviral restriction factors and pathogen sensors constantly surveying the cell to rapidly mount an antiviral response through the synthesis and downstream activity of interferons. After pathogen clearance, the host's ability to resolve this antiviral response and return to homeostasis is critical. Here, we found that isoforms of the RNA-binding protein ZAP functioned as both a direct antiviral restriction factor and an interferon-resolution factor. The short isoform of ZAP bound to and mediated the degradation of several host interferon messenger RNAs, and thus acted as a negative feedback regulator of the interferon response. In contrast, the long isoform of ZAP had antiviral functions and did not regulate interferon. The two isoforms contained identical RNA-targeting domains, but differences in their intracellular localization modulated specificity for host versus viral RNA, which resulted in disparate effects on viral replication during the innate immune response.


Asunto(s)
Infecciones por Alphavirus/inmunología , Interferones/genética , Isoformas de Proteínas/metabolismo , Proteínas de Unión al ARN/metabolismo , ARN/metabolismo , Proteínas Represoras/metabolismo , Virus Sindbis/fisiología , Infecciones por Alphavirus/genética , Retroalimentación Fisiológica , Células HEK293 , Células Hep G2 , Homeostasis , Humanos , Inmunidad Innata , Factor 3 Regulador del Interferón/genética , Factor 3 Regulador del Interferón/metabolismo , Unión Proteica , Isoformas de Proteínas/genética , ARN/genética , ARN Interferente Pequeño/genética , Proteínas de Unión al ARN/genética , Proteínas Represoras/genética , Replicación Viral
2.
Immunity ; 51(3): 451-464.e6, 2019 09 17.
Artículo en Inglés | MEDLINE | ID: mdl-31471108

RESUMEN

Type I and III interferons (IFNs) activate similar downstream signaling cascades, but unlike type I IFNs, type III IFNs (IFNλ) do not elicit strong inflammatory responses in vivo. Here, we examined the molecular mechanisms underlying this disparity. Type I and III IFNs displayed kinetic differences in expression of IFN-stimulated genes and proinflammatory responses, with type I IFNs preferentially stimulating expression of the transcription factor IRF1. Type III IFNs failed to induce IRF1 expression because of low IFNλ receptor abundance and insufficient STAT1 activation on epithelial cells and thus did not activate the IRF1 proinflammatory gene program. Rather, IFNλ stimulation preferentially induced factors implicated in tissue repair. Our findings suggest that IFN receptor compartmentalization and abundance confer a spatiotemporal division of labor where type III IFNs control viral spread at the site of the infection while restricting tissue damage; the transient induction of inflammatory responses by type I IFNs recruits immune effectors to promote protective immunity.


Asunto(s)
Factor 1 Regulador del Interferón/inmunología , Interferón Tipo I/inmunología , Interferones/inmunología , Animales , Línea Celular , Células Epiteliales/inmunología , Humanos , Inflamación/inmunología , Masculino , Ratones , Ratones Endogámicos C57BL , Factor de Transcripción STAT1/inmunología , Interferón lambda
3.
Proc Natl Acad Sci U S A ; 120(51): e2300474120, 2023 Dec 19.
Artículo en Inglés | MEDLINE | ID: mdl-38100417

RESUMEN

Seasonal influenza results in 3 to 5 million cases of severe disease and 250,000 to 500,000 deaths annually. Macrophages have been implicated in both the resolution and progression of the disease, but the drivers of these outcomes are poorly understood. We probed mouse lung transcriptomic datasets using the Digital Cell Quantifier algorithm to predict immune cell subsets that correlated with mild or severe influenza A virus (IAV) infection outcomes. We identified a unique lung macrophage population that transcriptionally resembled small serosal cavity macrophages and whose presence correlated with mild disease. Until now, the study of serosal macrophage translocation in the context of viral infections has been neglected. Here, we show that pleural macrophages (PMs) migrate from the pleural cavity to the lung after infection with IAV. We found that the depletion of PMs increased morbidity and pulmonary inflammation. There were increased proinflammatory cytokines in the pleural cavity and an influx of neutrophils within the lung. Our results show that PMs are recruited to the lung during IAV infection and contribute to recovery from influenza. This study expands our knowledge of PM plasticity and identifies a source of lung macrophages independent of monocyte recruitment and local proliferation.


Asunto(s)
Virus de la Influenza A , Gripe Humana , Infecciones por Orthomyxoviridae , Animales , Ratones , Humanos , Gripe Humana/genética , Pulmón , Macrófagos , Macrófagos Alveolares
4.
EMBO Rep ; 24(4): e56660, 2023 04 05.
Artículo en Inglés | MEDLINE | ID: mdl-36880581

RESUMEN

Interferon-induced transmembrane protein 3 (IFITM3) is an antiviral protein that alters cell membranes to block fusion of viruses. Conflicting reports identified opposing effects of IFITM3 on SARS-CoV-2 infection of cells, and its impact on viral pathogenesis in vivo remains unclear. Here, we show that IFITM3 knockout (KO) mice infected with SARS-CoV-2 experience extreme weight loss and lethality compared to mild infection in wild-type (WT) mice. KO mice have higher lung viral titers and increases in inflammatory cytokine levels, immune cell infiltration, and histopathology. Mechanistically, we observe disseminated viral antigen staining throughout the lung and pulmonary vasculature in KO mice, as well as increased heart infection, indicating that IFITM3 constrains dissemination of SARS-CoV-2. Global transcriptomic analysis of infected lungs shows upregulation of gene signatures associated with interferons, inflammation, and angiogenesis in KO versus WT animals, highlighting changes in lung gene expression programs that precede severe lung pathology and fatality. Our results establish IFITM3 KO mice as a new animal model for studying severe SARS-CoV-2 infection and overall demonstrate that IFITM3 is protective in SARS-CoV-2 infections in vivo.


Asunto(s)
COVID-19 , SARS-CoV-2 , Animales , Ratones , COVID-19/genética , Interferones/genética , Pulmón , Ratones Noqueados
5.
Proc Natl Acad Sci U S A ; 119(21): e2202012119, 2022 05 24.
Artículo en Inglés | MEDLINE | ID: mdl-35588457

RESUMEN

Severe acute respiratory syndrome coronavirus 2 (SARS­CoV-2) is a worldwide health concern, and new treatment strategies are needed. Targeting inflammatory innate immunity pathways holds therapeutic promise, but effective molecular targets remain elusive. Here, we show that human caspase-4 (CASP4) and its mouse homolog, caspase-11 (CASP11), are up-regulated in SARS­CoV-2 infections and that CASP4 expression correlates with severity of SARS­CoV-2 infection in humans. SARS­CoV-2­infected Casp11−/− mice were protected from severe weight loss and lung pathology, including blood vessel damage, compared to wild-type (WT) mice and mice lacking the caspase downstream effector gasdermin-D (Gsdmd−/−). Notably, viral titers were similar regardless of CASP11 knockout. Global transcriptomics of SARS­CoV-2­infected WT, Casp11−/−, and Gsdmd−/− lungs identified restrained expression of inflammatory molecules and altered neutrophil gene signatures in Casp11−/− mice. We confirmed that protein levels of inflammatory mediators interleukin (IL)-1ß, IL-6, and CXCL1, as well as neutrophil functions, were reduced in Casp11−/− lungs. Additionally, Casp11−/− lungs accumulated less von Willebrand factor, a marker for endothelial damage, but expressed more Kruppel-Like Factor 2, a transcription factor that maintains vascular integrity. Overall, our results demonstrate that CASP4/11 promotes detrimental SARS­CoV-2­induced inflammation and coagulopathy, largely independently of GSDMD, identifying CASP4/11 as a promising drug target for treatment and prevention of severe COVID-19.


Asunto(s)
COVID-19 , Caspasas Iniciadoras/metabolismo , SARS-CoV-2 , Tromboinflamación , Animales , COVID-19/enzimología , COVID-19/patología , Caspasas Iniciadoras/genética , Progresión de la Enfermedad , Humanos , Pulmón/patología , Ratones , Ratones Noqueados , Índice de Severidad de la Enfermedad , Tromboinflamación/enzimología , Tromboinflamación/genética
6.
J Immunol ; 208(2): 247-256, 2022 01 15.
Artículo en Inglés | MEDLINE | ID: mdl-35017214

RESUMEN

IFNs are comprised of three families of cytokines that confer protection against pathogen infection and uncontrolled cellular proliferation. The broad role IFNs play in innate and adaptive immune regulation has placed them under heavy scrutiny to position them as "friend" or "foe" across pathologies. Genetic lesions in genes involving IFN synthesis and signaling underscore the disparate outcomes of aberrant IFN signaling. Abrogation of the response leads to susceptibility to microbial infections whereas unabated IFN induction underlies a variety of inflammatory diseases and tumor immune evasion. Type I and III IFNs have overlapping roles in antiviral protection, yet the mechanisms by which they are induced and promote the expression of IFN-stimulated genes and inflammation can distinguish their biological functions. In this review, we examine the molecular factors that shape the shared and distinct roles of type I and III IFNs in immunity.


Asunto(s)
Interferón Tipo I/inmunología , Interferones/inmunología , Virosis/inmunología , Regulación de la Expresión Génica/genética , Regulación de la Expresión Génica/inmunología , Humanos , Inflamación/inmunología , Interferón Tipo I/metabolismo , Interferón Tipo I/uso terapéutico , Interferones/metabolismo , Interferones/uso terapéutico , Transducción de Señal/inmunología , Activación Transcripcional/genética , Activación Transcripcional/inmunología , Interferón lambda
7.
J Immunol ; 209(5): 845-854, 2022 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-36130131

RESUMEN

Retinoic acid-inducible gene I-like receptors (RLRs) are cytosolic RNA sensors critical for initiation of antiviral immunity. Activation of RLRs following RNA recognition leads to production of antiviral genes and IFNs for induction of broad antiviral immunity. Although the RLRs are ubiquitously expressed, much of our understanding of these molecules comes from their study in epithelial cells and fibroblasts. However, RLR activation is critical for induction of immune function and long-term protective immunity. Recent work has focused on the roles of RLRs in immune cells and their contribution to programming of effective immune responses. This new understanding of RLR function in immune cells and immune programming has led to the development of vaccines and therapeutics targeting the RLRs. This review covers recent advances in our understanding of the contribution of RLRs to immune cell function during infection and the emerging RLR-targeting strategies for induction of immunity against cancer and viral infection.


Asunto(s)
ARN Helicasas DEAD-box , Transducción de Señal , Antivirales , Proteína 58 DEAD Box , ARN Helicasas DEAD-box/metabolismo , Inmunidad Innata , ARN , Tretinoina
8.
Blood ; 138(8): 722-737, 2021 08 26.
Artículo en Inglés | MEDLINE | ID: mdl-34436524

RESUMEN

Immunopathology and intestinal stem cell (ISC) loss in the gastrointestinal (GI) tract is the prima facie manifestation of graft-versus-host disease (GVHD) and is responsible for significant mortality after allogeneic bone marrow transplantation (BMT). Approaches to prevent GVHD to date focus on immune suppression. Here, we identify interferon-λ (IFN-λ; interleukin-28 [IL-28]/IL-29) as a key protector of GI GVHD immunopathology, notably within the ISC compartment. Ifnlr1-/- mice displayed exaggerated GI GVHD and mortality independent of Paneth cells and alterations to the microbiome. Ifnlr1-/- intestinal organoid growth was significantly impaired, and targeted Ifnlr1 deficiency exhibited effects intrinsic to recipient Lgr5+ ISCs and natural killer cells. PEGylated recombinant IL-29 (PEG-rIL-29) treatment of naive mice enhanced Lgr5+ ISC numbers and organoid growth independent of both IL-22 and type I IFN and modulated proliferative and apoptosis gene sets in Lgr5+ ISCs. PEG-rIL-29 treatment improved survival, reduced GVHD severity, and enhanced epithelial proliferation and ISC-derived organoid growth after BMT. The preservation of ISC numbers in response to PEG-rIL-29 after BMT occurred both in the presence and absence of IFN-λ-signaling in recipient natural killer cells. IFN-λ is therefore an attractive and rapidly testable approach to prevent ISC loss and immunopathology during GVHD.


Asunto(s)
Trasplante de Médula Ósea , Citocinas/farmacología , Enfermedades Gastrointestinales , Enfermedad Injerto contra Huésped , Interleucinas/farmacocinética , Transducción de Señal , Animales , Citocinas/inmunología , Enfermedades Gastrointestinales/tratamiento farmacológico , Enfermedades Gastrointestinales/genética , Enfermedades Gastrointestinales/inmunología , Enfermedad Injerto contra Huésped/tratamiento farmacológico , Enfermedad Injerto contra Huésped/genética , Enfermedad Injerto contra Huésped/inmunología , Interleucinas/inmunología , Ratones , Ratones Noqueados , Receptores de Interferón/genética , Receptores de Interferón/inmunología , Índice de Severidad de la Enfermedad , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética , Transducción de Señal/inmunología , Trasplante Homólogo
9.
Immunity ; 40(6): 936-48, 2014 Jun 19.
Artículo en Inglés | MEDLINE | ID: mdl-24931123

RESUMEN

Virus infection is sensed in the cytoplasm by retinoic acid-inducible gene I (RIG-I, also known as DDX58), which requires RNA and polyubiquitin binding to induce type I interferon (IFN) and activate cellular innate immunity. We show that the human IFN-inducible oligoadenylate synthetases-like (OASL) protein has antiviral activity and mediates RIG-I activation by mimicking polyubiquitin. Loss of OASL expression reduced RIG-I signaling and enhanced virus replication in human cells. Conversely, OASL expression suppressed replication of a number of viruses in a RIG-I-dependent manner and enhanced RIG-I-mediated IFN induction. OASL interacted and colocalized with RIG-I, and through its C-terminal ubiquitin-like domain specifically enhanced RIG-I signaling. Bone-marrow-derived macrophages from mice deficient for Oasl2 showed that among the two mouse orthologs of human OASL, Oasl2 is functionally similar to human OASL. Our findings show a mechanism by which human OASL contributes to host antiviral responses by enhancing RIG-I activation.


Asunto(s)
2',5'-Oligoadenilato Sintetasa/inmunología , ARN Helicasas DEAD-box/inmunología , Infecciones por Virus ADN/inmunología , Interferón Tipo I/inmunología , Infecciones por Virus ARN/inmunología , 2',5'-Oligoadenilato Sintetasa/genética , Animales , Proteína 58 DEAD Box , Células HCT116 , Células HEK293 , Humanos , Inmunidad Innata , Factor 7 Regulador del Interferón/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Poliubiquitina , Unión Proteica/inmunología , Interferencia de ARN , ARN Interferente Pequeño , Receptores Inmunológicos , Transducción de Señal/inmunología , Replicación Viral/inmunología
10.
Trends Immunol ; 38(8): 558-566, 2017 08.
Artículo en Inglés | MEDLINE | ID: mdl-28666937

RESUMEN

miRNAs play an important role in fine-tuning host immune homeostasis and responses through the regulation of mRNA stability and translation. Studies have demonstrated that miRNA-mediated regulation of gene expression has a profound impact on immune cell development, function, and response to invading pathogens. As we continue to examine the mechanisms by which miRNAs maintain the balance between robust protective host immune responses and dysregulated responses that promote immune pathology, careful consideration of the complexity of post-transcriptional immune regulation is needed. Distinct tissue- and stimulus-specific RNA-RNA and RNA-protein interactions can modulate the functions of a given miRNA. Thus, new challenges emerge in the identification of post-transcriptional coregulatory modules and the genetic factors that impact miRNA function.


Asunto(s)
MicroARNs/inmunología , Procesamiento Postranscripcional del ARN/inmunología , Regiones no Traducidas 3'/inmunología , Animales , Variación Genética/inmunología , Humanos , MicroARNs/genética , MicroARNs/metabolismo , Estabilidad del ARN/inmunología , ARN Largo no Codificante/inmunología , ARN Mensajero/química , ARN Mensajero/inmunología , Proteínas de Unión al ARN/inmunología , Proteínas de Unión al ARN/metabolismo
11.
J Virol ; 91(11)2017 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-28331091

RESUMEN

Ebola virus (EBOV) and Reston virus (RESTV) are members of the Ebolavirus genus which greatly differ in their pathogenicity. While EBOV causes a severe disease in humans characterized by a dysregulated inflammatory response and elevated cytokine and chemokine production, there are no reported disease-associated human cases of RESTV infection, suggesting that RESTV is nonpathogenic for humans. The underlying mechanisms determining the pathogenicity of different ebolavirus species are not yet known. In this study, we dissected the host response to EBOV and RESTV infection in primary human monocyte-derived macrophages (MDMs). As expected, EBOV infection led to a profound proinflammatory response, including strong induction of type I and type III interferons (IFNs). In contrast, RESTV-infected macrophages remained surprisingly silent. Early activation of IFN regulatory factor 3 (IRF3) and NF-κB was observed in EBOV-infected, but not in RESTV-infected, MDMs. In concordance with previous results, MDMs treated with inactivated EBOV and Ebola virus-like particles (VLPs) induced NF-κB activation mediated by Toll-like receptor 4 (TLR4) in a glycoprotein (GP)-dependent manner. This was not the case in cells exposed to live RESTV, inactivated RESTV, or VLPs containing RESTV GP, indicating that RESTV GP does not trigger TLR4 signaling. Our results suggest that the lack of immune activation in RESTV-infected MDMs contributes to lower pathogenicity by preventing the cytokine storm observed in EBOV infection. We further demonstrate that inhibition of TLR4 signaling abolishes EBOV GP-mediated NF-κB activation. This finding indicates that limiting the excessive TLR4-mediated proinflammatory response in EBOV infection should be considered as a potential supportive treatment option for EBOV disease.IMPORTANCE Emerging infectious diseases are a major public health concern, as exemplified by the recent devastating Ebola virus (EBOV) outbreak. Different ebolavirus species are associated with widely varying pathogenicity in humans, ranging from asymptomatic infections for Reston virus (RESTV) to severe disease with fatal outcomes for EBOV. In this comparative study of EBOV- and RESTV-infected human macrophages, we identified key differences in host cell responses. Consistent with previous data, EBOV infection is associated with a proinflammatory signature triggered by the surface glycoprotein (GP), which can be inhibited by blocking TLR4 signaling. In contrast, infection with RESTV failed to stimulate a strong host response in infected macrophages due to the inability of RESTV GP to stimulate TLR4. We propose that disparate proinflammatory host signatures contribute to the differences in pathogenicity reported for ebolavirus species and suggest that proinflammatory pathways represent an intriguing target for the development of novel therapeutics.


Asunto(s)
Ebolavirus/inmunología , Ebolavirus/patogenicidad , Interacciones Huésped-Patógeno , Macrófagos/virología , Receptor Toll-Like 4/metabolismo , Animales , Línea Celular , Quimiocinas/inmunología , Quimiocinas/metabolismo , Chlorocebus aethiops , Citocinas/inmunología , Células Dendríticas/inmunología , Células Dendríticas/virología , Ebolavirus/fisiología , Perfilación de la Expresión Génica , Humanos , Factor 3 Regulador del Interferón/genética , Factor 3 Regulador del Interferón/inmunología , Interferones/inmunología , Macrófagos/inmunología , Macrófagos/metabolismo , Subunidad p50 de NF-kappa B/genética , Subunidad p50 de NF-kappa B/metabolismo , Receptor Toll-Like 4/genética , Receptor Toll-Like 4/inmunología , Células Vero , Virulencia
12.
J Virol ; 90(5): 2240-53, 2015 Dec 09.
Artículo en Inglés | MEDLINE | ID: mdl-26656717

RESUMEN

UNLABELLED: The 1918-1919 influenza pandemic remains the single greatest infectious disease outbreak in the past century. Mouse and nonhuman primate infection models have shown that the 1918 virus induces overly aggressive innate and proinflammatory responses. To understand the response to viral infection and the role of individual 1918 genes on the host response to the 1918 virus, we examined reassortant avian viruses nearly identical to the pandemic 1918 virus (1918-like avian virus) carrying either the 1918 hemagglutinin (HA) or PB2 gene. In mice, both genes enhanced 1918-like avian virus replication, but only the mammalian host adaptation of the 1918-like avian virus through reassortment of the 1918 PB2 led to increased lethality. Through the combination of viral genetics and host transcriptional profiling, we provide a multidimensional view of the molecular mechanisms by which the 1918 PB2 gene drives viral pathogenicity. We demonstrate that 1918 PB2 enhances immune and inflammatory responses concomitant with increased cellular infiltration in the lung. We also show for the first time, that 1918 PB2 expression results in the repression of both canonical and noncanonical Wnt signaling pathways, which are crucial for inflammation-mediated lung regeneration and repair. Finally, we utilize regulatory enrichment and network analysis to define the molecular regulators of inflammation, epithelial regeneration, and lung immunopathology that are dysregulated during influenza virus infection. Taken together, our data suggest that while both HA and PB2 are important for viral replication, only 1918 PB2 exacerbates lung damage in mice infected with a reassortant 1918-like avian virus. IMPORTANCE: As viral pathogenesis is determined in part by the host response, understanding the key host molecular driver(s) of virus-mediated disease, in relation to individual viral genes, is a promising approach to host-oriented drug efforts in preventing disease. Previous studies have demonstrated the importance of host adaptive genes, HA and PB2, in mediating disease although the mechanisms by which they do so are still poorly understood. Here, we combine viral genetics and host transcriptional profiling to show that although both 1918 HA and 1918 PB2 are important mediators of efficient viral replication, only 1918 PB2 impacts the pathogenicity of an avian influenza virus sharing high homology to the 1918 pandemic influenza virus. We demonstrate that 1918 PB2 enhances deleterious inflammatory responses and the inhibition of regeneration and repair functions coordinated by Wnt signaling in the lungs of infected mice, thereby promoting virus-associated disease.


Asunto(s)
Subtipo H1N1 del Virus de la Influenza A/enzimología , Subtipo H1N1 del Virus de la Influenza A/patogenicidad , Infecciones por Orthomyxoviridae/patología , Infecciones por Orthomyxoviridae/virología , ARN Polimerasa Dependiente del ARN/metabolismo , Proteínas Virales/metabolismo , Factores de Virulencia/metabolismo , Vía de Señalización Wnt/inmunología , Animales , Línea Celular , Modelos Animales de Enfermedad , Femenino , Perfilación de la Expresión Génica , Glicoproteínas Hemaglutininas del Virus de la Influenza/genética , Glicoproteínas Hemaglutininas del Virus de la Influenza/metabolismo , Humanos , Inflamación/patología , Subtipo H1N1 del Virus de la Influenza A/genética , Subtipo H1N1 del Virus de la Influenza A/inmunología , Pulmón/patología , Pulmón/virología , Ratones Endogámicos BALB C , ARN Polimerasa Dependiente del ARN/genética , Virus Reordenados/enzimología , Virus Reordenados/patogenicidad , Proteínas Virales/genética , Virulencia , Factores de Virulencia/genética
13.
J Immunol ; 192(12): 5933-42, 2014 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-24799566

RESUMEN

Polyomaviruses encode a large T Ag (LT), a multifunctional protein essential for the regulation of both viral and host cell gene expression and productive viral infection. Previously, we have shown that stable expression of LT protein results in upregulation of genes involved in the IFN induction and signaling pathway. In this study, we focus on the cellular signaling mechanism that leads to the induction of IFN responses by LT. Our results show that ectopic expression of SV40 LT results in the induction of IFN-stimulated genes (ISGs) in human fibroblasts and confers an antiviral state. We describe a LT-initiated DNA damage response (DDR) that activates IFN regulatory factor 1, causing IFN-ß production and consequent ISG expression in human cells. This IFN-ß and ISG induction is dependent on ataxia-telangiectasia mutated and Rad3-related (ATR) kinase, but independent of ATM. ATR kinase inhibition using a selective kinase inhibitor (ETP-46464) caused a decrease in IFN regulatory factor 1 stabilization and ISG expression. Furthermore, expression of a mutant LT that does not induce DDR also does not induce IFN-ß and ISGs. These results show that, in the absence of viral infection, LT-initiated activation of ATR-dependent DDR is sufficient for the induction of an IFN-ß-mediated innate immune response in human cells. Thus, we have uncovered a novel and critical role for ATR as a mediator of antiviral responses utilizing LT.


Asunto(s)
Antígenos Transformadores de Poliomavirus/inmunología , Daño del ADN/inmunología , Factor 1 Regulador del Interferón/inmunología , Interferón beta/inmunología , Virus 40 de los Simios/inmunología , Antígenos Transformadores de Poliomavirus/genética , Proteínas de la Ataxia Telangiectasia Mutada/antagonistas & inhibidores , Proteínas de la Ataxia Telangiectasia Mutada/genética , Proteínas de la Ataxia Telangiectasia Mutada/inmunología , Daño del ADN/genética , Células HEK293 , Humanos , Factor 1 Regulador del Interferón/genética , Interferón beta/genética , Inhibidores de Proteínas Quinasas/farmacología , Estabilidad Proteica/efectos de los fármacos , Virus 40 de los Simios/genética
14.
J Immunol ; 191(3): 1476-85, 2013 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-23804715

RESUMEN

IFN regulatory factor (IRF) 4 is a hematopoietic cell-specific transcription factor that regulates the maturation and differentiation of immune cells. Using an inducible expression system, we found that IRF4 directly induced a specific subset of IFN-stimulated genes (ISGs) in a type I IFN-independent manner in both epithelial and B cell lines. Moreover, Kaposi sarcoma-associated herpesvirus (KSHV)-encoded viral FLICE inhibitory protein (vFLIP) enhances IRF4-mediated gene induction. Coexpression of IRF4 with vFLIP significantly increased ISG60 (IFIT3) and Cig5 (RSAD2) transcription that was dependent on the ability of vFLIP to activate NF-κB. A vFLIP mutant (A57L) defective in NF-κB activation failed to enhance IRF4-mediated ISG induction. Thus, we provide a physiologically relevant mechanism by which viral protein-mediated NF-κB activation modulates specific ISG induction by IRF4. In contrast, IRF4 also acted as a negative regulator of KSHV replication and transcription activator expression after induction of KSHV lytic reactivation in KSHV-positive primary effusion lymphoma cells. Taken together, these results suggest a dual role for IRF4 in regulating ISG induction and KSHV lytic reactivation in primary effusion lymphoma cells.


Asunto(s)
Herpesvirus Humano 8/metabolismo , Factores Reguladores del Interferón/metabolismo , Péptidos y Proteínas de Señalización Intracelular/genética , Proteínas/genética , Proteína Reguladora de Apoptosis Similar a CASP8 y FADD/genética , Proteína Reguladora de Apoptosis Similar a CASP8 y FADD/metabolismo , Línea Celular Transformada , Activación Enzimática/genética , Expresión Génica , Regulación de la Expresión Génica , Células HEK293 , Herpesvirus Humano 8/genética , Humanos , Interferón Tipo I , Linfoma/genética , Linfoma/virología , Mutación , FN-kappa B/metabolismo , Oxidorreductasas actuantes sobre Donantes de Grupo CH-CH , Transcripción Genética/genética , Activación Transcripcional , Latencia del Virus , Replicación Viral/genética
15.
STAR Protoc ; 5(1): 102857, 2024 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-38285737

RESUMEN

Dot-blot analysis is a technique that allows for fast and convenient detection and identification of nucleic acids and proteins. Here, we provide a guide for nucleic acid isolation from eukaryotic cells and sample processing to detect RNA/DNA hybrids. We then provide detailed steps to quantify dot signal intensity. This protocol can be adapted for screening conditions that result in the accumulation of R-loops. For complete details on the use and execution of this protocol, please refer to Smith et al.1.


Asunto(s)
Células Eucariotas , Estructuras R-Loop , Immunoblotting , ARN
16.
JCI Insight ; 9(13)2024 May 21.
Artículo en Inglés | MEDLINE | ID: mdl-38973611

RESUMEN

Optimization of protective immune responses against SARS-CoV-2 remains an urgent worldwide priority. In this regard, type III IFN (IFN-λ) restricts SARS-CoV-2 infection in vitro, and treatment with IFN-λ limits infection, inflammation, and pathogenesis in murine models. Furthermore, IFN-λ has been developed for clinical use to limit COVID-19 severity. However, whether endogenous IFN-λ signaling has an effect on SARS-CoV-2 antiviral immunity and long-term immune protection in vivo is unknown. In this study, we identified a requirement for IFN-λ signaling in promoting viral clearance and protective immune programming in SARS-CoV-2 infection of mice. Expression of both IFN and IFN-stimulated gene (ISG) in the lungs were minimally affected by the absence of IFN-λ signaling and correlated with transient increases in viral titers. We found that IFN-λ supported the generation of protective CD8 T cell responses against SARS-CoV-2 by facilitating accumulation of CD103+ DC in lung draining lymph nodes (dLN). IFN-λ signaling specifically in DCs promoted the upregulation of costimulatory molecules and the proliferation of CD8 T cells. Intriguingly, antigen-specific CD8 T cell immunity to SARS-CoV-2 was independent of type I IFN signaling, revealing a nonredundant function of IFN-λ. Overall, these studies demonstrate a critical role for IFN-λ in protective innate and adaptive immunity upon infection with SARS-CoV-2 and suggest that IFN-λ serves as an immune adjuvant to support CD8 T cell immunity.


Asunto(s)
Linfocitos T CD8-positivos , COVID-19 , Interferón Tipo I , SARS-CoV-2 , Animales , Linfocitos T CD8-positivos/inmunología , SARS-CoV-2/inmunología , Ratones , COVID-19/inmunología , COVID-19/virología , Interferón Tipo I/inmunología , Interferón Tipo I/metabolismo , Pulmón/inmunología , Pulmón/virología , Transducción de Señal/inmunología , Modelos Animales de Enfermedad , Interferón lambda , Interferones/inmunología , Interferones/metabolismo , Ratones Endogámicos C57BL , Ratones Noqueados , Células Dendríticas/inmunología , Humanos
17.
Nat Commun ; 15(1): 2751, 2024 Mar 29.
Artículo en Inglés | MEDLINE | ID: mdl-38553499

RESUMEN

Influenza virus activates cellular inflammasome pathways, which can be both beneficial and detrimental to infection outcomes. Here, we investigate the function of the inflammasome-activated, pore-forming protein gasdermin D (GSDMD) during infection. Ablation of GSDMD in knockout (KO) mice (Gsdmd-/-) significantly attenuates influenza virus-induced weight loss, lung dysfunction, lung histopathology, and mortality compared with wild type (WT) mice, despite similar viral loads. Infected Gsdmd-/- mice exhibit decreased inflammatory gene signatures shown by lung transcriptomics. Among these, diminished neutrophil gene activation signatures are corroborated by decreased detection of neutrophil elastase and myeloperoxidase in KO mouse lungs. Indeed, directly infected neutrophils are observed in vivo and infection of neutrophils in vitro induces release of DNA and tissue-damaging enzymes that is largely dependent on GSDMD. Neutrophil depletion in infected WT mice recapitulates the reductions in mortality, lung inflammation, and lung dysfunction observed in Gsdmd-/- animals, while depletion does not have additive protective effects in Gsdmd-/- mice. These findings implicate a function for GSDMD in promoting lung neutrophil responses that amplify influenza virus-induced inflammation and pathogenesis. Targeting the GSDMD/neutrophil axis may provide a therapeutic avenue for treating severe influenza.


Asunto(s)
Neutrófilos , Orthomyxoviridae , Animales , Ratones , Neutrófilos/metabolismo , Gasderminas , Inflamasomas/genética , Inflamasomas/metabolismo , Inflamación/genética , Inflamación/metabolismo , Orthomyxoviridae/metabolismo , Proteínas de Unión a Fosfato/genética , Proteínas de Unión a Fosfato/metabolismo
18.
J Food Sci ; 88(12): 4928-4941, 2023 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-37905708

RESUMEN

This study validated an analytical technique using headspace gas chromatography with flame ionization detection to quantify acrylonitrile monomer with a quantification limit of 0.10 ± 0.04 µg kg-1 . Subsequently, the acrylonitrile migration from polypropylene granules was evaluated in food simulants water and ethanol (50% v/v) and at two temperatures (20 ± 1°C and 44 ± 2°C) for up to 6 weeks, representing the service time of a bottle. From the experimental data obtained, pseudo-second-order kinetics were adjusted to represent the acrylonitrile migration into the simulants. For water, equilibrium concentrations of 13.58 and 16.58 µg kg-1 at 20 and 44°C, respectively, were obtained, while for 50% ethanol, 15.07 and 16.40 µg kg-1 were obtained for the same temperatures. The experimental results and the values estimated from the migration kinetics indicate that the maximum acrylonitrile concentration will not exceed the tolerable specific limit established in regulations. PRACTICAL APPLICATION: The migration of compounds such as acrylonitrile can be a drawback resulting in an undesirable reduction in the shelf life of liquid foods packaged in bottles made of materials such as polypropylene. In this paper, acrylonitrile migration kinetics and a methodology are proposed to determine whether the tolerable migration limits are ever reached, which can serve as a tool for producers of this type of packaging of food to predict shelf life.


Asunto(s)
Acrilonitrilo , Embalaje de Alimentos , Polipropilenos , Acrilonitrilo/análisis , Etanol/química , Agua/química , Contaminación de Alimentos/análisis
19.
Cell Rep ; 42(8): 112805, 2023 08 29.
Artículo en Inglés | MEDLINE | ID: mdl-37467105

RESUMEN

Cellular stress in the form of disrupted transcription, loss of organelle integrity, or damage to nucleic acids can elicit inflammatory responses by activating signaling cascades canonically tasked with controlling pathogen infections. These stressors must be kept in check to prevent unscheduled activation of interferon, which contributes to autoinflammation. This study examines the role of the transcription factor myocyte enhancing factor 2A (MEF2A) in setting the threshold of transcriptional stress responses to prevent R-loop accumulation. Increases in R-loops lead to the induction of interferon and inflammatory responses in a DEAD-box helicase 41 (DDX41)-, cyclic GMP-AMP synthase (cGAS)-, and stimulator of interferon genes (STING)-dependent manner. The loss of MEF2A results in the activation of ATM and RAD3-related (ATR) kinase, which is also necessary for the activation of STING. This study identifies the role of MEF2A in sustaining transcriptional homeostasis and highlights the role of ATR in positively regulating R-loop-associated inflammatory responses.


Asunto(s)
Nucleotidiltransferasas , Transducción de Señal , Nucleotidiltransferasas/metabolismo , ARN Helicasas , Interferones , Inmunidad Innata
20.
Immunohorizons ; 7(6): 431-441, 2023 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-37289499

RESUMEN

IL-35 is an immunosuppressive cytokine with roles in cancer, autoimmunity, and infectious disease. In the conventional model of IL-35 biology, the p35 and Ebi3 domains of this cytokine interact with IL-12Rß2 and gp130, respectively, on the cell surface of regulatory T and regulatory B cells, triggering their suppression of Th cell activity. Here we use a human IL-12 bioactivity reporter cell line, protein binding assays, and primary human Th cells to demonstrate an additional mechanism by which IL-35 suppresses Th cell activity, wherein IL-35 directly inhibits the association of IL-12 with its surface receptor IL-12Rß2 and downstream IL-12-dependent activities. IL-12 binding to the surface receptor IL-12Rß1 was unaffected by IL-35. These data demonstrate that in addition to acting via regulatory T and regulatory B cells, human IL-35 can also directly suppress IL-12 bioactivity and its interaction with IL-12Rß2.


Asunto(s)
Interleucina-12 , Interleucinas , Humanos , Interleucina-12/metabolismo , Unión Proteica , Interleucinas/metabolismo , Citocinas/metabolismo , Línea Celular
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA