Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Front Immunol ; 14: 1173605, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37435074

RESUMEN

Human metapneumovirus (HMPV) is a pneumovirus that may cause severe respiratory disease in humans. HMPV infection has been found to increase susceptibility to bacterial superinfections leading to increased morbidity and mortality. The molecular mechanisms underlying HMPV-mediated increase in bacterial susceptibility are poorly understood and largely understudied. Type I interferons (IFNs), while critical for antiviral defenses, may often have detrimental effects by skewing the host immune response and cytokine output of immune cells. It is currently unknown if HMPV skews the inflammatory response in human macrophages triggered by bacterial stimuli. Here we report that HMPV pre-infection impacts production of specific cytokines. HMPV strongly suppresses IL-1ß transcription in response to LPS or heat-killed Pseudomonas aeruginosa and Streptococcus pneumonia, while enhancing mRNA levels of IL-6, TNF-α and IFN-ß. We demonstrate that in human macrophages the HMPV-mediated suppression of IL-1ß transcription requires TANK-binding kinase 1 (TBK1) and signaling via the IFN-ß-IFNAR axis. Interestingly, our results show that HMPV pre-infection did not impair the LPS-stimulated activation of NF-κB and HIF-1α, transcription factors that stimulate IL-1ß mRNA synthesis in human cells. Furthermore, we determined that sequential HMPV-LPS treatment resulted in accumulation of the repressive epigenetic mark H3K27me3 at the IL1B promoter. Thus, for the first time we present data revealing the molecular mechanisms by which HMPV shapes the cytokine output of human macrophages exposed to bacterial pathogens/LPS, which appears to be dependent on epigenetic reprogramming at the IL1B promoter leading to reduced synthesis of IL-1ß. These results may improve current understanding of the role of type I IFNs in respiratory disease mediated not only by HMPV, but also by other respiratory viruses that are associated with superinfections.


Asunto(s)
Infecciones Bacterianas , Interferón beta , Interleucina-1beta , Infecciones por Paramyxoviridae , Sobreinfección , Humanos , Citocinas , Metapneumovirus , Transcripción Genética , Infecciones Bacterianas/inmunología , Infecciones por Paramyxoviridae/inmunología
2.
Front Immunol ; 12: 563336, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34248923

RESUMEN

The innate immune and host-protective responses to viruses, such as the airway pathogen human metapneumovirus (HMPV), depend on interferons (IFNs) that is induced through TANK-binding kinase 1 (TBK1) and IFN regulatory factors (IRFs). The transcription factor IRF1 is important for host resistance against several viruses and has a key role in induction of IFN-λ at mucosal surfaces. In most cell types IRF1 is expressed at very low levels, but its mRNA is rapidly induced when the demand for IRF1 activity arises. Despite general recognition of the importance of IRF1 to antiviral responses, the molecular mechanisms by which IRF1 is regulated during viral infections are not well understood. Here we identify the serine/threonine kinase TBK1 and IFN-ß as critical regulators of IRF1 mRNA and protein levels in human monocyte-derived macrophages. We find that inhibition of TBK1 activity either by the semi-selective TBK1/IKKε inhibitor BX795 or by siRNA-mediated knockdown abrogates HMPV-induced expression of IRF1. Moreover, we show that canonical NF-κB signaling is involved in IRF1 induction and that the TBK1/IKKε inhibitor BX795, but not siTBK1 treatment, impairs HMPV-induced phosphorylation of the NF-κB subunit p65. At later time-points of the infection, IRF1 expression depended heavily on IFN-ß-mediated signaling via the IFNAR-STAT1 pathway. Hence, our results suggest that TBK1 activation and TBK1/IKKε-mediated phosphorylation of the NF-κB subunit p65 control transcription of IRF1. Our study identifies a novel mechanism for IRF1 induction in response to viral infection of human macrophages that could be relevant not only to defense against HMPV, but also to other viral, bacterial and fungal pathogens.


Asunto(s)
Inmunidad Innata , Factor 1 Regulador del Interferón/genética , Factor 1 Regulador del Interferón/inmunología , Interferón Tipo I/inmunología , Metapneumovirus/inmunología , Proteínas Serina-Treonina Quinasas/inmunología , Línea Celular , Células Cultivadas , Humanos , Interferón Tipo I/genética , Metapneumovirus/genética , Monocitos/inmunología , Monocitos/virología , Proteínas Serina-Treonina Quinasas/genética , Transducción de Señal
3.
J Virol ; 95(3)2021 01 13.
Artículo en Inglés | MEDLINE | ID: mdl-33208442

RESUMEN

Hepatitis C virus (HCV) infection triggers Golgi fragmentation through the Golgi-resident protein immunity-related GTPase M (IRGM). Here, we report the roles of NLRP3 (NOD-, LRR- and pyrin domain-containing protein 3) and ASC (apoptosis-associated speck-like protein containing a caspase activation and recruitment domain [CARD]), two inflammasome components, in the initial events leading to this fragmentation. We show that ASC resides at the Golgi with IRGM at homeostasis. Upon infection, ASC dissociates from both IRGM and the Golgi and associates with HCV-induced NLRP3. NLRP3 silencing inhibits Golgi fragmentation. ASC silencing disrupts the Golgi structure in both control and infected cells and reduces the localization of IRGM at the Golgi. IRGM depletion in the ASC-silenced cells cannot totally restore the Golgi structure. These data highlight a role for ASC, upstream of the formation of the inflammasome, in regulating IRGM through its control on the Golgi. A similar mechanism occurs in response to nigericin treatment, but not in cells infected with another member of the Flaviviridae family, Zika virus (ZIKV). We propose a model for a newly ascribed function of the inflammasome components in Golgi structural remodeling during certain stimuli.IMPORTANCE Numerous pathogens can affect cellular homeostasis and organelle dynamics. Hepatitis C virus (HCV) triggers Golgi fragmentation through the immunity-related GTPase M (IRGM), a resident Golgi protein, to enhance its lipid supply for replication. Here, we reveal the role of the inflammasome components NLRP3 and ASC in this process, thus uncovering a new interplay between effectors of inflammation and viral infection or stress. We show that the inflammasome component ASC resides at the Golgi under homeostasis and associates with IRGM. Upon HCV infection, ASC is recruited to NLRP3 and dissociates from IRGM, causing Golgi fragmentation. Our results uncover that aside from their known function in the inflammation response, these host defense regulators also ensure the maintenance of intact intracellular structure in homeostasis, while their activation relieves factors leading to Golgi remodeling.


Asunto(s)
Proteínas Adaptadoras de Señalización CARD/metabolismo , Proteínas de Unión al GTP/metabolismo , Aparato de Golgi/fisiología , Hepacivirus/aislamiento & purificación , Hepatitis C/virología , Inflamasomas/metabolismo , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Apoptosis , Proteínas Adaptadoras de Señalización CARD/genética , Proteínas de Unión al GTP/genética , Aparato de Golgi/virología , Hepatitis C/metabolismo , Hepatitis C/patología , Humanos , Proteína con Dominio Pirina 3 de la Familia NLR/genética
4.
Virus Res ; 287: 198094, 2020 10 02.
Artículo en Inglés | MEDLINE | ID: mdl-32730781

RESUMEN

The past century has witnessed major advances in the control of many infectious diseases, yet outbreaks and epidemics caused by (re-) emerging RNA viruses continue to pose a global threat to human health. As illustrated by the global COVID19 pandemic, high healthcare costs, economic disruption and loss of productivity reinforce the unmet medical need to develop new antiviral strategies to combat not only the current pandemic but also future viral outbreaks. Pivotal for effective anti-viral defense is the innate immune system, a first line host response that senses and responds to virus infection. While molecular details of the innate immune response are well characterized, this research field is now being revolutionized with the recognition that cell metabolism has a major impact on the antiviral and inflammatory responses to virus infections. A detailed understanding of the role of metabolic regulation with respect to antiviral and inflammatory responses, together with knowledge of the strategies used by viruses to exploit immunometabolic pathways, will ultimately change our understanding and treatment of pathogenic viral diseases. INITIATE is a Marie Sklodowska-Curie Actions Innovative Training Network (MSCA-ITN), with the goal to train 15 early stage PhD researchers (ESRs) to become experts in antiviral immunometabolism (https://initiate-itn.eu/). To this end, INITIATE brings together a highly complementary international team of academic and corporate leaders from 7 European countries, with outstanding track records in the historically distinct research fields of virology, immunology and metabolism. The ESRs of INITIATE are trained in these interdisciplinary research fields through individual investigator-driven research projects, specialized scientific training events, workshops on academia-industry interactions, outreach & communication. INITIATE will deliver a new generation of creative and entrepreneurial researchers who will be able to face the inevitable future challenges in combating viral diseases.


Asunto(s)
Betacoronavirus/inmunología , Investigación Biomédica/métodos , Infecciones por Coronavirus/tratamiento farmacológico , Educación Médica/métodos , Inmunidad Innata/inmunología , Neumonía Viral/tratamiento farmacológico , Antivirales/uso terapéutico , Betacoronavirus/efectos de los fármacos , COVID-19 , Infecciones por Coronavirus/economía , Atención a la Salud/economía , Atención a la Salud/métodos , Interacciones Huésped-Patógeno/fisiología , Humanos , Pandemias/economía , Neumonía Viral/economía , SARS-CoV-2
5.
Mediators Inflamm ; 2019: 4964239, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31686982

RESUMEN

Human metapneumovirus (HMPV) may cause severe respiratory disease. The early innate immune response to viruses like HMPV is characterized by induction of antiviral interferons (IFNs) and proinflammatory immune mediators that are essential in shaping adaptive immune responses. Although innate immune responses to HMPV have been comprehensively studied in mice and murine immune cells, there is less information on these responses in human cells, comparing different cell types infected with the same HMPV strain. The aim of this study was to characterize the HMPV-induced mRNA expression of critical innate immune mediators in human primary cells relevant for airway disease. In particular, we determined type I versus type III IFN expression in human epithelial cells and monocyte-derived macrophages (MDMs) and dendritic cells (MDDCs). In epithelial cells, HMPV induced only low levels of IFN-ß mRNA, while a robust mRNA expression of IFN-λs was found in epithelial cells, MDMs, and MDDCs. In addition, we determined induction of the interferon regulatory factors (IRFs) IRF1, IRF3, and IRF7 and critical inflammatory cytokines (IL-6, IP-10, and IL-1ß). Interestingly, IRF1 mRNA was predominantly induced in MDMs and MDDCs. Overall, our results suggest that for HMPV infection of MDDCs, MDMs, NECs, and A549 cells (the cell types examined), cell type is a strong determinator of the ability of HMPV to induce different innate immune mediators. HMPV induces the transcription of IFN-ß and IRF1 to higher extents in MDMs and MDDCs than in A549s and NECs, whereas the induction of type III IFN-λ and IRF7 is considerable in MDMs, MDDCs, and A549 epithelial cells.


Asunto(s)
Inmunidad Innata/fisiología , Metapneumovirus/patogenicidad , Infecciones por Paramyxoviridae/inmunología , Células A549 , Células Cultivadas , Quimiocina CXCL10/metabolismo , Técnica del Anticuerpo Fluorescente , Humanos , Inmunidad Innata/genética , Interleucina-6/metabolismo , Macrófagos/metabolismo , Metapneumovirus/inmunología , Microscopía Confocal , Infecciones por Paramyxoviridae/metabolismo , ARN Mensajero/metabolismo , Transducción de Señal
6.
Viruses ; 11(10)2019 10 18.
Artículo en Inglés | MEDLINE | ID: mdl-31635418

RESUMEN

Viruses are the major causes of acute and chronic infectious diseases in the world. According to the World Health Organization, there is an urgent need for better control of viral diseases. Repurposing existing antiviral agents from one viral disease to another could play a pivotal role in this process. Here, we identified novel activities of obatoclax and emetine against herpes simplex virus type 2 (HSV-2), echovirus 1 (EV1), human metapneumovirus (HMPV) and Rift Valley fever virus (RVFV) in cell cultures. Moreover, we demonstrated novel activities of emetine against influenza A virus (FLUAV), niclosamide against HSV-2, brequinar against human immunodeficiency virus 1 (HIV-1), and homoharringtonine against EV1. Our findings may expand the spectrum of indications of these safe-in-man agents and reinforce the arsenal of available antiviral therapeutics pending the results of further in vitro and in vivo tests.


Asunto(s)
Antivirales/farmacología , Virus/efectos de los fármacos , Animales , Antivirales/uso terapéutico , Compuestos de Bifenilo/farmacología , Técnicas de Cultivo de Célula , Chlorocebus aethiops , Perros , Emetina/farmacología , Enterovirus Humano B/efectos de los fármacos , VIH-1/efectos de los fármacos , Herpesvirus Humano 2/efectos de los fármacos , Homoharringtonina/farmacología , Humanos , Indoles , Células de Riñón Canino Madin Darby , Niclosamida/farmacología , Pirroles/farmacología , Células Vero , Virosis/tratamiento farmacológico , Virus/clasificación
7.
Front Immunol ; 10: 2186, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31636628

RESUMEN

Viruses are one of the major causes of acute and chronic infectious diseases and thus a major contributor to the global burden of disease. Several studies have shown how viruses have evolved to hijack basic cellular pathways and evade innate immune response by modulating key host factors and signaling pathways. A collective view of these multiple studies could advance our understanding of virus-host interactions and provide new therapeutic perspectives for the treatment of viral diseases. Here, we performed an integrative meta-analysis to elucidate the 17 different host-virus interactomes. Network and bioinformatics analyses showed how viruses with small genomes efficiently achieve the maximal effect by targeting multifunctional and highly connected host proteins with a high occurrence of disordered regions. We also identified the core cellular process subnetworks that are targeted by all the viruses. Integration with functional RNA interference (RNAi) datasets showed that a large proportion of the targets are required for viral replication. Furthermore, we performed an interactome-informed drug re-purposing screen and identified novel activities for broad-spectrum antiviral agents against hepatitis C virus and human metapneumovirus. Altogether, these orthogonal datasets could serve as a platform for hypothesis generation and follow-up studies to broaden our understanding of the viral evasion landscape.


Asunto(s)
Interacciones Microbiota-Huesped , Mapas de Interacción de Proteínas , Virosis/inmunología , Proteína Coat de Complejo I/fisiología , Biología Computacional , Humanos , Evasión Inmune , Transducción de Señal/fisiología , Virosis/tratamiento farmacológico , Replicación Viral
8.
Genome Announc ; 6(21)2018 May 24.
Artículo en Inglés | MEDLINE | ID: mdl-29798923

RESUMEN

Here, we report the complete genome sequences of human metapneumovirus (HMPV) prior to and after passaging in LLC-MK2 cells. Paired comparisons of the 13,335-nucleotide genomes revealed that the virus acquired the T10736C transition in its genome, which did not affect the amino acid sequences of HMPV proteins.

9.
Sci Rep ; 8(1): 883, 2018 01 17.
Artículo en Inglés | MEDLINE | ID: mdl-29343779

RESUMEN

Thymic stromal lymphopoietin (TSLP) is associated with several allergic diseases including asthma. Two isoforms of TSLP exist in humans, a long form (lfTSLP) and a short form (sfTSLP), displaying distinct immunological functions. Recently, TSLP was found to be upregulated in human airway cells upon human metapneumovirus (hMPV) infection, yet it remains unclear if the two isoforms are regulated differently during hMPV infection. Importantly, the molecular mechanisms underlying hMPV-mediated TSLP induction remain undescribed. In this study, we characterized the expression and regulation of TSLP in hMPV-infected human airway cells. We demonstrated that hMPV strongly induced the expression of pro-inflammatory lfTSLP in human airway epithelial cells and lung fibroblasts. Further, knockdown of pattern recognition receptors retinoic acid-inducible gene I (RIG-I) or Toll-like receptor 3 (TLR3), as well as downstream signal transducers, abrogated hMPV-mediated lfTSLP induction. Importantly, silencing of TANK-binding kinase 1 (TBK1) also impaired hMPV-mediated lfTSLP induction, which could be attributed to compromised NF-κB activation. Overall, these results suggest that TBK1 may be instrumental for hMPV-mediated activation of NF-κB downstream RIG-I and TLR3, leading to a specific induction of lfTSLP in hMPV-infected human airway cells.


Asunto(s)
Citocinas/genética , Regulación de la Expresión Génica/genética , Inflamación/genética , Metapneumovirus/patogenicidad , Infecciones por Paramyxoviridae/genética , Transducción de Señal/genética , Células A549 , Asma/genética , Asma/virología , Línea Celular Tumoral , Proteína 58 DEAD Box/genética , Células Epiteliales/virología , Humanos , Hipersensibilidad/genética , Hipersensibilidad/virología , Inflamación/virología , FN-kappa B/genética , Infecciones por Paramyxoviridae/virología , Proteínas Serina-Treonina Quinasas/genética , Receptor Toll-Like 3/genética , Linfopoyetina del Estroma Tímico
10.
Proc Natl Acad Sci U S A ; 114(17): E3462-E3471, 2017 04 25.
Artículo en Inglés | MEDLINE | ID: mdl-28389568

RESUMEN

Positive-stranded RNA viruses, such as hepatitis C virus (HCV), assemble their viral replication complexes by remodeling host intracellular membranes to a membranous web. The precise composition of these replication complexes and the detailed mechanisms by which they are formed are incompletely understood. Here we show that the human immunity-related GTPase M (IRGM), known to contribute to autophagy, plays a previously unrecognized role in this process. We show that IRGM is localized at the Golgi apparatus and regulates the fragmentation of Golgi membranes in response to HCV infection, leading to colocalization of Golgi vesicles with replicating HCV. Our results show that IRGM controls phosphorylation of GBF1, a guanine nucleotide exchange factor for Arf-GTPases, which normally operates in Golgi membrane dynamics and vesicle coating in resting cells. We also find that HCV triggers IRGM-mediated phosphorylation of the early autophagy initiator ULK1, thereby providing mechanistic insight into the role of IRGM in HCV-mediated autophagy. Collectively, our results identify IRGM as a key Golgi-situated regulator that links intracellular membrane remodeling by autophagy and Golgi fragmentation with viral replication.


Asunto(s)
Autofagia , Proteínas de Unión al GTP/metabolismo , Aparato de Golgi/metabolismo , Hepacivirus/fisiología , Membranas Intracelulares/metabolismo , Replicación Viral/fisiología , Homólogo de la Proteína 1 Relacionada con la Autofagia/genética , Homólogo de la Proteína 1 Relacionada con la Autofagia/metabolismo , Línea Celular Tumoral , Proteínas de Unión al GTP/genética , Aparato de Golgi/genética , Aparato de Golgi/virología , Factores de Intercambio de Guanina Nucleótido/genética , Factores de Intercambio de Guanina Nucleótido/metabolismo , Humanos , Membranas Intracelulares/virología , Péptidos y Proteínas de Señalización Intracelular/genética , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Fosforilación/genética
11.
PLoS One ; 11(5): e0155484, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27171557

RESUMEN

Human metapneumovirus (hMPV) causes severe airway infection in children that may be caused by an unfavorable immune response. The nature of the innate immune response to hMPV in naturally occurring infections in children is largely undescribed, and it is unknown if inflammasome activation is implicated in disease pathogenesis. We examined nasopharynx aspirates and blood samples from hMPV-infected children without detectable co-infections. The expression of inflammatory and antiviral genes were measured in nasal airway secretions by relative mRNA quantification while blood plasma proteins were determined by a multiplex immunoassay. Several genes were significantly up-regulated at mRNA and protein level in the hMPV infected children. Most apparent was the expression of the chemokine IP-10, the pro-inflammatory cytokine IL-18 in addition to the interferon inducible gene ISG54. Interestingly, children experiencing more severe disease, as indicated by a severity index, had significantly more often up-regulation of the inflammasome-associated genes IL-1ß and NLRP3. Overall, our data point to cytokines, particularly inflammasome-associated, that might be important in hMPV mediated lung disease and the antiviral response in children with severe infection. Our study is the first to demonstrate that inflammasome components are associated with increased illness severity in hMPV-infected children.


Asunto(s)
Antivirales/uso terapéutico , Citocinas/genética , Metapneumovirus/patogenicidad , Infecciones por Paramyxoviridae/genética , Infecciones por Paramyxoviridae/virología , Antivirales/farmacología , Estudios de Casos y Controles , Niño , Preescolar , Citocinas/metabolismo , Femenino , Regulación de la Expresión Génica/efectos de los fármacos , Genotipo , Humanos , Lactante , Inflamasomas/genética , Inflamasomas/metabolismo , Interferones/genética , Interferones/metabolismo , Masculino , Metapneumovirus/efectos de los fármacos , Nasofaringe/patología , Nasofaringe/virología , Infecciones por Paramyxoviridae/sangre , Infecciones por Paramyxoviridae/tratamiento farmacológico
12.
Cell Signal ; 25(9): 1804-12, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23707530

RESUMEN

Antiviral responses can be triggered by the cytoplasmic RNA helicase RIG-I that binds to viral RNA. RIG-I-mediated signaling stimulates the transcription factors IRF3 and NF-κB and their activation mechanisms have been intensively studied. Here we examined Sendai virus (SV)-mediated activation of the transcription factor CREB and the role of its feedback repressor ICER in production of endogenous antiviral proteins. We show that SV infection and the mitochondrial adapter protein MAVS promote CREB phosphorylation that is dependent upon p38 MAPK and MK2. ICER is induced by CREB and acts as a feedback repressor of CRE-dependent transcription. We found that SV infection stimulated induction of ICER mRNA and protein expression. Surprisingly, ectopic expression and siRNA-mediated knockdown of ICER revealed that ICER is a positive regulator of the production of antiviral IFN-ß and IP10 during SV infection. In contrast, ICER did not affect SV-elicited phosphorylation of IRF3, NF-κB or ATF2/c-Jun, transcription factors governing IFN-ß and IP10 synthesis. However, expression of ICER increased total IRF3 protein levels during SV infection. These results point to a novel role of ICER in antiviral immune signaling acting to increase levels of antiviral effectors.


Asunto(s)
Modulador del Elemento de Respuesta al AMP Cíclico/inmunología , ARN Helicasas DEAD-box/inmunología , Interacciones Huésped-Patógeno , Interferón beta/inmunología , Infecciones por Respirovirus/inmunología , Virus Sendai/fisiología , Factor de Transcripción Activador 2/inmunología , Proteínas Adaptadoras Transductoras de Señales/inmunología , AMP Cíclico/inmunología , Modulador del Elemento de Respuesta al AMP Cíclico/genética , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/inmunología , Proteína 58 DEAD Box , Células HEK293 , Humanos , Factor 3 Regulador del Interferón/inmunología , Interferón beta/genética , ARN Mensajero/genética , Receptores Inmunológicos , Infecciones por Respirovirus/genética , Activación Transcripcional , eIF-2 Quinasa/inmunología , Proteínas Quinasas p38 Activadas por Mitógenos/inmunología
13.
Cytokine ; 57(3): 337-46, 2012 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-22200507

RESUMEN

Prolonged release of cytokines after activation of the innate immune system may lead to systemic infection and inflammatory diseases. Many cytokines with short half-lives contain adenine- and uridine-rich elements (AREs) in their 3'-untranslated region (UTR), which mediate mRNA destabilization. The Toll-like receptors (TLRs) TLR3 and TLR4 induce immune responses via the adaptor proteins TRIF or TRIF and MyD88, respectively, leading to IFN-ß production. The 3'-UTR of IFN-ß mRNA contains an ARE sequence. We demonstrate that the TLR3 ligand dsRNA and the TLR4 ligand LPS induce stabilization of IFN-ß mRNA transcripts in monocyte-derived dendritic cells. In cells from TRIF(-/-) and MyD88(-/-) mice we found that dsRNA-induced stabilization of IFN-ß mRNA is TRIF-dependent. MAPK-activated protein 2 (MK2) has previously been found to regulate mRNA stabilization. We show that dsRNA elicits increased MK2 activation, mediated by TRIF and p38 MAPK. Chemical inhibition of p38 and MK2, and siRNA knockdown of MK2 relieved dsRNA-triggered prolongation of IFN-ß mRNA half-life. Taken together, these results suggest that TLR3 induces signaling mechanisms involving TRIF, p38 MAPK and MK2 to enhance stabilization of IFN-ß mRNA contributing to enhanced IFN-ß levels during pathogen infections.


Asunto(s)
Interferón beta/genética , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Estabilidad del ARN , Receptor Toll-Like 3/metabolismo , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo , Regiones no Traducidas 3'/genética , Proteínas Adaptadoras del Transporte Vesicular/metabolismo , Animales , Secuencia de Bases , Células Dendríticas/citología , Células Dendríticas/efectos de los fármacos , Células Dendríticas/enzimología , Activación Enzimática/efectos de los fármacos , Regulación de la Expresión Génica/efectos de los fármacos , Células HEK293 , Semivida , Humanos , Lipopolisacáridos/farmacología , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Ratones , Datos de Secuencia Molecular , Monocitos/citología , Poli I-C/farmacología , Estabilidad del ARN/efectos de los fármacos , ARN Bicatenario/farmacología , ARN Mensajero/genética , ARN Mensajero/metabolismo , Secuencias Reguladoras de Ácido Ribonucleico/genética , Factor de Necrosis Tumoral alfa/genética
14.
J Lipids ; 2011: 532145, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21912747

RESUMEN

Oxidized low-density lipoproteins (LDLs) play an important role during the development of atherosclerosis characterized by intimal inflammation and macrophage accumulation. A key component of LDL is lysophosphatidylcholine (lysoPC). LysoPC is a strong proinflammatory mediator, and its mechanism is uncertain, but it has been suggested to be mediated via the platelet activating factor (PAF) receptor. Here, we report that PAF triggers a pertussis toxin- (PTX-) sensitive intracellular signaling pathway leading to sequential activation of sPLA(2), PLD, cPLA(2), and AA release in human-derived monocytes. In contrast, lysoPC initiates two signaling pathways, one sequentially activating PLD and cPLA(2), and a second parallel PTX-sensitive pathway activating cPLA(2) with concomitant activation of sPLA(2), all leading to AA release. In conclusion, lysoPC and PAF stimulate AA release by divergent pathways suggesting involvement of independent receptors. Elucidation of monocyte lysoPC-specific signaling mechanisms will aid in the development of novel strategies for atherosclerosis prevention, diagnosis, and therapy.

15.
J Biol Chem ; 285(32): 24904-14, 2010 Aug 06.
Artículo en Inglés | MEDLINE | ID: mdl-20511230

RESUMEN

The transcription factor interferon regulatory factor-3 (IRF3) regulates expression of type I interferon-beta and plays an important role in antiviral immunity. Despite the biological importance of IRF3, its in vivo phosphorylation pattern has not been reported. In this study, we have identified residues in IRF3 that are phosphorylated in vivo after infection with Sendai virus. We found that Sendai virus induced phosphorylation of the C-terminal residues Thr(390) and Ser(396), in addition to either Ser(385) or Ser(386). Moreover, Ser(173) and Ser(175) were constitutively phosphorylated. Ser(396) has previously been suggested to be the major target of the IRF3-activating kinase TBK1 (TANK-binding kinase-1), whereas Thr(390) has not previously been implicated in IRF3 regulation. Mutagenesis studies indicated that phosphorylation of Thr(390) promotes Ser(396) phosphorylation and binding to the coactivator cAMP-response element-binding protein. Taken together, our results show that IRF3 is subject to multiple interdependent phosphorylations, and we identify Thr(390) as a novel in vivo phosphorylation site that modulates the phosphorylation status of TBK1-targeted Ser(396).


Asunto(s)
Regulación Viral de la Expresión Génica , Factor 3 Regulador del Interferón/química , Secuencia de Aminoácidos , Sitios de Unión , Humanos , Espectrometría de Masas/métodos , Datos de Secuencia Molecular , Mutagénesis , Fosforilación , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Recombinantes/química , Virus Sendai/metabolismo , Homología de Secuencia de Aminoácido , Serina/química , Treonina/química
16.
J Biol Chem ; 285(22): 17011-9, 2010 May 28.
Artículo en Inglés | MEDLINE | ID: mdl-20351107

RESUMEN

Interferon regulatory factors (IRFs) are crucial for transcription during innate immune responses. We have previously shown that the tyrosine kinase c-Src enhances IRF-3-dependent transcription in response to viral double-stranded RNA. In this study, we show that c-Src has distinct roles in Toll-like receptor (TLR)-mediated activation of IRF-5 and IRF-3. Surprisingly, c-Src inhibition markedly enhanced IRF-5 activation after treatment with unmethylated CpG, while suppressing IRF-3 activation. Also, CpG-elicited interleukin-6 mRNA production was increased, whereas IP10 mRNA synthesis was reduced in cells deficient in c-Src. Interestingly, c-Src regulated TLR-stimulated induction of activating transcription factor 3 (ATF3), a transcriptional repressor. Depletion of ATF3 by small interfering RNA markedly enhanced interleukin-6 production after CpG treatment, whereas IP10 production was reduced. These results demonstrate functional specificity for c-Src in TLR-stimulated responses and suggest that c-Src modulation and ATF3 activity may contribute to differential regulation of IRF-3- versus IRF-5-mediated gene expression.


Asunto(s)
Proteínas Tirosina Quinasas/metabolismo , Receptores Toll-Like/metabolismo , Factor de Transcripción Activador 3/metabolismo , Animales , Proteína Tirosina Quinasa CSK , Línea Celular , Islas de CpG , Humanos , Factor 3 Regulador del Interferón/metabolismo , Factores Reguladores del Interferón/metabolismo , Interleucina-6/metabolismo , Ratones , Modelos Biológicos , Interferencia de ARN , Transducción de Señal , Familia-src Quinasas
17.
J Biol Chem ; 284(28): 19122-31, 2009 Jul 10.
Artículo en Inglés | MEDLINE | ID: mdl-19419966

RESUMEN

Antiviral immune responses are initiated through Toll-like receptors (TLRs) and RIG-I (retinoic acid-inducible gene-I)-like RNA helicases that recognize nucleic acids from distinct viruses. In this study, we show that the tyrosine kinase c-Src participates in antiviral responses induced by the cytoplasmic RNA helicase RIG-I. Sendai virus (SV), which is recognized by RIG-I, induced c-Src phosphorylation. Functional impairment of c-Src through chemical inhibition or transient expression of a c-Src kinase-inactive mutant attenuated production of endogenous antiviral proteins after SV infection or after expression of RIG-I or its adapter protein MAVS. Importantly, SV-stimulated synthesis of antiviral proteins was significantly impaired in cells treated with c-Src small interfering RNA and in cells from c-Src-deficient mice. In addition, we found that c-Src interacted with components of the RIG-I pathway: RIG-I, MAVS, and TRAF3 (tumor necrosis factor receptor-associated factor-3). The interaction between c-Src and TRAF3 was found to occur within the RING domain of TRAF3. Taken together, our results suggest that c-Src enhances RIG-I-mediated signaling, acting at the level of TRAF3.


Asunto(s)
Antivirales/metabolismo , ARN Helicasas DEAD-box/metabolismo , Regulación de la Expresión Génica , Factor 3 Asociado a Receptor de TNF/metabolismo , Familia-src Quinasas/fisiología , Animales , Proteína 58 DEAD Box , Genes Reporteros , Humanos , Factor 3 Regulador del Interferón/metabolismo , Ratones , Modelos Biológicos , ARN Interferente Pequeño/metabolismo , Receptores Inmunológicos , Virus Sendai/metabolismo , Transducción de Señal , Familia-src Quinasas/metabolismo
18.
EMBO J ; 25(14): 3335-46, 2006 Jul 26.
Artículo en Inglés | MEDLINE | ID: mdl-16858407

RESUMEN

Double-stranded RNA (dsRNA) is produced during the replication cycle of most viruses and triggers antiviral immune responses through Toll-like receptor 3 (TLR3). However, the molecular mechanisms and subcellular compartments associated with dsRNA-TLR3-mediated signaling are largely unknown. Here we show that c-Src tyrosine kinase is activated by dsRNA in human monocyte-derived dendritic cells, and is recruited to TLR3 in a dsRNA-dependent manner. DsRNA-induced activation of interferon-regulatory factor 3 and signal transducer and activator of transcription 1 was abolished in Src kinase-deficient cells, and restored by adding back c-Src, suggesting a central role of c-Src in antiviral immunity. We also provide evidence that TLR3 is localized in the endoplasmic reticulum of unstimulated cells, moves to dsRNA-containing endosomes in response to dsRNA, and colocalizes with c-Src on endosomes containing dsRNA in the lumen. These results provide novel insight into the molecular mechanisms of TLR3-mediated signaling, which may contribute to the understanding of innate immune responses during viral infections.


Asunto(s)
Endosomas/enzimología , Endosomas/virología , Proteínas Tirosina Quinasas/fisiología , ARN Bicatenario/antagonistas & inhibidores , Rhinovirus/fisiología , Transducción de Señal/fisiología , Receptor Toll-Like 3/fisiología , Animales , Proteína Tirosina Quinasa CSK , Células Cultivadas , Células Dendríticas , Endosomas/fisiología , Activación Enzimática/fisiología , Células HeLa , Humanos , Factor 3 Regulador del Interferón/metabolismo , Ratones , Ratones Noqueados , Proteínas Tirosina Quinasas/metabolismo , ARN Bicatenario/fisiología , Rhinovirus/genética , Factor de Transcripción STAT1/metabolismo , Receptor Toll-Like 3/metabolismo , Familia-src Quinasas
19.
J Lipid Res ; 46(2): 201-10, 2005 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-15576846

RESUMEN

Atherosclerosis is a chronic inflammatory disease of the vessel wall characterized by the accumulation of lipid-laden macrophages and fibrotic material. The initiation of the disease is accompanied by the accumulation of modified lipoproteins in the vessel wall. Group IIa secretory phospholipase A2 (sPLA2 IIa) is a key candidate player in the enzymatic modification of low density lipoproteins. To study the role of sPLA2 IIa in macrophages during atherogenesis, transgenic mice were generated using the human sPLA2 IIa gene and the CD11b promoter. Bone marrow transplantation to LDL receptor-deficient mice was performed to study sPLA2 IIa in atherosclerosis. After 10 weeks of high-fat diet, mice overexpressing sPLA2 IIa in macrophages showed 2.3-fold larger lesions compared with control mice. Pathological examination revealed that sPLA2 IIa-expressing mice had increased collagen in their lesions, independent of lesion size. However, smooth muscle cells or fibroblasts in the lesions were not affected. Other parameters studied, including T-cells and cell turnover, were not significantly affected by overexpression of sPLA2 IIa in macrophages. These data clearly show that macrophage sPLA2 IIa is a proatherogenic factor and suggest that the enzyme regulates collagen production in the plaque and thus fibrotic cap development.


Asunto(s)
Arteriosclerosis/metabolismo , Colágeno/metabolismo , Macrófagos/metabolismo , Fosfolipasas A/biosíntesis , Animales , Northern Blotting , Trasplante de Médula Ósea , Antígeno CD11b/genética , Colesterol/metabolismo , Fibroblastos/metabolismo , Células Espumosas/metabolismo , Fosfolipasas A2 Grupo II , Humanos , Etiquetado Corte-Fin in Situ , Lipoproteínas LDL/metabolismo , Ratones , Ratones Transgénicos , Modelos Genéticos , Miocitos del Músculo Liso/metabolismo , Necrosis , Fenotipo , Fosfolipasas/química , Fosfolipasas A2 , Regiones Promotoras Genéticas , Receptores de LDL/genética , Receptores de LDL/fisiología , Factores de Tiempo
20.
J Lipid Res ; 45(5): 831-8, 2004 May.
Artículo en Inglés | MEDLINE | ID: mdl-14754906

RESUMEN

Oxidation and lipolytic remodeling of LDL are believed to stimulate LDL entrapment in the arterial wall, expanding the inflammatory response and promoting atherosclerosis. However, the cellular responses and molecular mechanisms underlying the atherogenic effects of lipolytically modified LDL are incompletely understood. Human THP-1 monocytes were prelabeled with [(3)H]arachidonic acid (AA) before incubation with LDL or LDL lipolytically modified by secretory PLA(2) (sPLA(2)) or bacterial sphingomyelinase (SMase). LDL elicited rapid and dose-dependent extracellular release of AA in monocytes. Interestingly, LDL modified by sPLA(2) or SMase displayed a marked increase in AA mobilization relative to native LDL, and this increase correlated with enhanced activity of cytosolic PLA(2) (cPLA(2)) assayed in vitro as well as increased monocyte tumor necrosis factor-alpha secretion. The AA liberation was attenuated by inhibitors toward cPLA(2) and sPLA(2), indicating that both PLA(2) enzymes participate in LDL-induced AA release. In conclusion, these results demonstrate that LDL lipolytically modified by sPLA(2) or SMase potentiates cellular AA release and cPLA(2) activation in human monocytes. From our results, we suggest novel atherogenic properties for LDL modified by sPLA(2) and SMase in AA release and signaling, which could contribute to the inflammatory gene expression observed in atherosclerosis.


Asunto(s)
Ácido Araquidónico/metabolismo , Lipoproteínas LDL/metabolismo , Lipoproteínas LDL/farmacología , Monocitos/efectos de los fármacos , Fosfolipasas A/metabolismo , Esfingomielina Fosfodiesterasa/metabolismo , Técnicas de Cultivo de Célula , Humanos , Lipoproteínas LDL/química , Monocitos/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...