Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Front Immunol ; 15: 1279920, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38495890

RESUMEN

Type I interferons play a fundamental role in innate host defense against viral infections by eliciting the induction of an antiviral gene program that serves to inhibit viral replication. Activation of type I interferon is regulated by the IRF3 transcription factor, which undergoes phosphorylation-dependent activation by the upstream kinase, TBK1, during viral infection. However, the mechanisms by which TBK1 achieves activation to support signaling to IRF3 remain incompletely understood. Here we identified the E3 ubiquitin ligase, tripartite motif containing 28 (TRIM28), as a positive regulator of type I interferon activation by facilitating TBK1 signaling. Genetic deletion of TRIM28 via CRISPR-Cas9 editing resulted in impaired type I interferon activation upon both RNA and DNA virus challenge, corresponding with increased susceptibility to virus infections in TRIM28 knockout cells. Mechanistically, TRIM28 interacted with TBK1 and mediated the assembly of K63-linked ubiquitin chains onto TBK1, a post-translational modification shown to augment TBK1 signal transmission events. TRIM28 knockout cells further displayed defective TBK1 phosphorylation and complex assembly with IRF3, resulting in impaired IRF3 phosphorylation. Altogether, our data demonstrate TBK1 to be a novel substrate for TRIM28 and identify TRIM28 as an essential regulatory factor in controlling innate antiviral immune responses.


Asunto(s)
Interferón Tipo I , Proteínas Serina-Treonina Quinasas , Proteínas Serina-Treonina Quinasas/metabolismo , Transducción de Señal , Fosforilación , Interferón beta
2.
Front Physiol ; 13: 887702, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36479348

RESUMEN

The rising prevalence of obesity presents a world-wide challenge as it is associated with numerous comorbidities including cardiovascular disease, insulin resistance and hypertension. Obesity-associated illnesses are estimated to cause nearly 4 million deaths globally per year, therefore there is a critical need to better understand associated pathogenesis, identify new therapeutic targets, and develop new interventions. Emerging data identify a key role for chronic inflammation in mediating obesity related disease states and reveal higher incidence of autoimmune disease development. Of the multiple potential mechanisms linking obesity and autoimmunity, the strongest link has been shown for leptin, a hormone secreted at high levels from obese white adipose tissue. Numerous studies have demonstrated that leptin enhances activation of both arms of the immune system, while its absence protects against development of autoimmunity. Other potential newly discovered mechanisms that contribute to autoimmune pathogenesis are not directly connected but also associated with obesity including sustained platelet activation, gut dysbiosis, and aging. Here we review how obesity instigates autoimmunity, particularly in the context of immune cell activations and adipokine secretion.

3.
Acta Pharm Sin B ; 12(4): 1624-1635, 2022 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-35251918

RESUMEN

SARS-CoV-2 is an emerging viral pathogen and a major global public health challenge since December of 2019, with limited effective treatments throughout the pandemic. As part of the innate immune response to viral infection, type I interferons (IFN-I) trigger a signaling cascade that culminates in the activation of hundreds of genes, known as interferon stimulated genes (ISGs), that collectively foster an antiviral state. We report here the identification of a group of type I interferon suppressed genes, including fatty acid synthase (FASN), which are involved in lipid metabolism. Overexpression of FASN or the addition of its downstream product, palmitate, increased viral infection while knockout or knockdown of FASN reduced infection. More importantly, pharmacological inhibitors of FASN effectively blocked infections with a broad range of viruses, including SARS-CoV-2 and its variants of concern. Thus, our studies not only suggest that downregulation of metabolic genes may present an antiviral strategy by type I interferon, but they also introduce the potential for FASN inhibitors to have a therapeutic application in combating emerging infectious diseases such as COVID-19.

4.
Front Cell Infect Microbiol ; 11: 789462, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-35083167

RESUMEN

Corona virus disease 2019 (COVID-19) pathogenesis is intimately linked to the severe acute respiratory syndrome corona virus 2 (SARS-CoV-2) and disease severity has been associated with compromised induction of type I interferon (IFN-I) cytokines which coordinate the innate immune response to virus infections. Here we identified the SARS-CoV-2 encoded protein, Spike, as an inhibitor of IFN-I that antagonizes viral RNA pattern recognition receptor RIG-I signaling. Ectopic expression of SARS-CoV-2 Spike blocked RIG-I mediated activation of IFNß and downstream induction of interferon stimulated genes. Consequently, SARS-CoV-2 Spike expressing cells harbored increased RNA viral burden compared to control cells. Co-immunoprecipitation experiments revealed SARS-CoV-2 Spike associated with interferon regulatory factor 3 (IRF3), a key transcription factor that governs IFN-I activation. Co-expression analysis via immunoassays further indicated Spike specifically suppressed IRF3 expression as NF-κB and STAT1 transcription factor levels remained intact. Further biochemical experiments uncovered SARS-CoV-2 Spike potentiated proteasomal degradation of IRF3, implicating a novel mechanism by which SARS-CoV-2 evades the host innate antiviral immune response to facilitate COVID-19 pathogenesis.


Asunto(s)
Factores de Restricción Antivirales/inmunología , COVID-19 , Inmunidad Innata , Factor 3 Regulador del Interferón , Glicoproteína de la Espiga del Coronavirus/inmunología , COVID-19/inmunología , Células HEK293 , Humanos , Factor 3 Regulador del Interferón/metabolismo , Interferón Tipo I/metabolismo , SARS-CoV-2
5.
Antimicrob Agents Chemother ; 63(12)2019 09 09.
Artículo en Inglés | MEDLINE | ID: mdl-31527024

RESUMEN

Azithromycin (AZM) is a widely used antibiotic, with additional antiviral and anti-inflammatory properties that remain poorly understood. Although Zika virus (ZIKV) poses a significant threat to global health, there are currently no vaccines or effective therapeutics against it. Herein, we report that AZM effectively suppresses ZIKV infection in vitro by targeting a late stage in the viral life cycle. Besides that, AZM upregulates the expression of host type I and III interferons and several of their downstream interferon-stimulated genes (ISGs) in response to ZIKV infection. In particular, we found that AZM upregulates the expression of MDA5 and RIG-I, pathogen recognition receptors (PRRs) induced by ZIKV infection, and increases the levels of phosphorylated TBK1 and IRF3. Interestingly, AZM treatment upregulates phosphorylation of TBK1, without inducing phosphorylation of IRF3 by itself. These findings highlight the potential use of AZM as a broad antiviral agent to combat viral infection and prevent ZIKV associated devastating clinical outcomes, such as congenital microcephaly.

6.
J Virol ; 93(23)2019 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-31511375

RESUMEN

In spite of several decades of research focused on understanding the biology of human herpes simplex virus 1 (HSV-1), no tool has been developed to study its genome in a high-throughput fashion. Here, we describe the creation of a transposon insertion mutant library of the HSV-1 genome. Using this tool, we aimed to identify novel viral regulators of type I interferon (IFN-I). HSV-1 evades the host immune system by encoding viral proteins that inhibit the type I interferon response. Applying differential selective pressure, we identified the three strongest viral IFN-I regulators in HSV-1. We report that the viral polymerase processivity factor UL42 interacts with the host transcription factor IFN regulatory factor 3 (IRF-3), inhibiting its phosphorylation and downstream beta interferon (IFN-ß) gene transcription. This study represents a proof of concept for the use of high-throughput screening of the HSV-1 genome in investigating viral biology and offers new targets both for antiviral therapy and for oncolytic vector design.IMPORTANCE This work is the first to report the use of a high-throughput mutagenesis method to study the genome of HSV-1. We report three novel viral proteins potentially involved in regulating the host type I interferon response. We describe a novel mechanism by which the viral protein UL42 is able to suppress the production of beta interferon. The tool we introduce in this study can be used to study the HSV-1 genome in great detail to better understand viral gene functions.


Asunto(s)
ADN Polimerasa Dirigida por ADN/metabolismo , Exodesoxirribonucleasas/metabolismo , Herpesvirus Humano 1/genética , Interferón Tipo I/metabolismo , Mutagénesis , Proteínas Virales/metabolismo , Células A549 , Antivirales/farmacología , ADN Polimerasa Dirigida por ADN/genética , Exodesoxirribonucleasas/genética , Células HEK293 , Herpesvirus Humano 1/fisiología , Humanos , Factor 3 Regulador del Interferón/metabolismo , Interferón beta/genética , Receptor de Interferón alfa y beta/genética , Proteínas Virales/genética
7.
J Clin Invest ; 129(5): 1926-1939, 2019 04 02.
Artículo en Inglés | MEDLINE | ID: mdl-30939123

RESUMEN

IL-26 is an antimicrobial protein secreted by Th17 cells that has the ability to directly kill extracellular bacteria. To ascertain whether IL-26 contributes to host defense against intracellular bacteria, we studied leprosy, caused by the obligate intracellular pathogen Mycobacterium leprae, as a model. Analysis of leprosy skin lesions by gene expression profiling and immunohistology revealed that IL-26 was more strongly expressed in lesions from the self-limited tuberculoid compared with expression in progressive lepromatous patients. IL-26 directly bound to M. leprae in axenic culture and reduced bacteria viability. Furthermore, IL-26, when added to human monocyte-derived macrophages infected with M. leprae, entered the infected cell, colocalized with the bacterium, and reduced bacteria viability. In addition, IL-26 induced autophagy via the cytoplasmic DNA receptor stimulator of IFN genes (STING), as well as fusion of phagosomes containing bacilli with lysosomal compartments. Altogether, our data suggest that the Th17 cytokine IL-26 contributes to host defense against intracellular bacteria.


Asunto(s)
Interleucinas/inmunología , Lepra Lepromatosa/microbiología , Lepra Tuberculoide/microbiología , Células Th17/inmunología , Autofagia , Citocinas/inmunología , Perfilación de la Expresión Génica , Humanos , Lisosomas/inmunología , Lisosomas/microbiología , Macrófagos/inmunología , Monocitos/citología , Mycobacterium leprae , Mycobacterium tuberculosis , Fagosomas/inmunología , Proteínas Recombinantes/inmunología , Transducción de Señal
8.
JCI Insight ; 4(8)2019 04 18.
Artículo en Inglés | MEDLINE | ID: mdl-30996142

RESUMEN

DC, through the uptake, processing, and presentation of antigen, are responsible for activation of T cell responses to defend the host against infection, yet it is not known if they can directly kill invading bacteria. Here, we studied in human leprosy, how Langerhans cells (LC), specialized DC, contribute to host defense against bacterial infection. IFN-γ treatment of LC isolated from human epidermis and infected with Mycobacterium leprae (M. leprae) activated an antimicrobial activity, which was dependent on the upregulation of the antimicrobial peptide cathelicidin and induction of autophagy. IFN-γ induction of autophagy promoted fusion of phagosomes containing M. leprae with lysosomes and the delivery of cathelicidin to the intracellular compartment containing the pathogen. Autophagy enhanced the ability of M. leprae-infected LC to present antigen to CD1a-restricted T cells. The frequency of IFN-γ labeling and LC containing both cathelicidin and autophagic vesicles was greater in the self-healing lesions vs. progressive lesions, thus correlating with the effectiveness of host defense against the pathogen. These data indicate that autophagy links the ability of DC to kill and degrade an invading pathogen, ensuring cell survival from the infection while facilitating presentation of microbial antigens to resident T cells.


Asunto(s)
Presentación de Antígeno , Autofagia , Células de Langerhans/inmunología , Lepra/inmunología , Mycobacterium leprae/inmunología , Antígenos Bacterianos/inmunología , Péptidos Catiónicos Antimicrobianos/inmunología , Péptidos Catiónicos Antimicrobianos/metabolismo , Autofagosomas/inmunología , Autofagosomas/metabolismo , Autofagosomas/microbiología , Biopsia , Células Cultivadas , Epidermis/inmunología , Epidermis/microbiología , Epidermis/patología , Humanos , Interferón gamma/inmunología , Células de Langerhans/microbiología , Células de Langerhans/ultraestructura , Lepra/microbiología , Lepra/patología , Lisosomas/inmunología , Lisosomas/metabolismo , Lisosomas/microbiología , Microscopía Electrónica de Transmisión , Mycobacterium leprae/aislamiento & purificación , Cultivo Primario de Células , Proteínas Recombinantes/inmunología , Linfocitos T/inmunología , Regulación hacia Arriba/inmunología , Catelicidinas
9.
Nat Commun ; 9(1): 2770, 2018 07 17.
Artículo en Inglés | MEDLINE | ID: mdl-30018345

RESUMEN

Detection of viral genomes by the innate immune system elicits an antiviral gene program mediated by type I interferons (IFNs). While viral RNA and DNA species induce IFN via separate pathways, the mechanisms by which these pathways are differentially modulated are unknown. Here we show that the positive regulator of IFN in the RNA pathway, TRAF3, has an inhibitory function in the DNA pathway. Loss of TRAF3 coincides with increased expression of the alternative NF-κB-inducing molecule, NIK, which interacts with the DNA pathway adaptor, STING, to enhance IFN induction. Cells lacking NIK display defective IFN activation in the DNA pathway due to impaired STING signaling, and NIK-deficient mice are more susceptible to DNA virus infection. Mechanistically, NIK operates independently from alternative NF-κB signaling components and instead requires autophosphorylation and oligomerization to activate STING. Thus a previously undescribed pathway for NIK exists in activating IFN in the DNA pathway.


Asunto(s)
ADN Viral/genética , Herpesvirus Humano 1/genética , Interacciones Huésped-Patógeno , Proteínas Serina-Treonina Quinasas/genética , ARN Viral/genética , Factor 3 Asociado a Receptor de TNF/genética , Virus de la Estomatitis Vesicular Indiana/genética , Células A549 , Animales , ADN Viral/inmunología , Femenino , Regulación de la Expresión Génica , Genes Reporteros , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/inmunología , Células HEK293 , Herpesvirus Humano 1/inmunología , Humanos , Inmunidad Innata , Interferón-alfa/genética , Interferón-alfa/inmunología , Interferón beta/genética , Interferón beta/inmunología , Proteínas de la Membrana/genética , Proteínas de la Membrana/inmunología , Ratones , Ratones Noqueados , FN-kappa B/genética , FN-kappa B/inmunología , Proteínas Serina-Treonina Quinasas/inmunología , ARN Viral/inmunología , Transducción de Señal , Células THP-1 , Factor 3 Asociado a Receptor de TNF/inmunología , Virus de la Estomatitis Vesicular Indiana/inmunología , Quinasa de Factor Nuclear kappa B
10.
Mol Cell Biol ; 37(1)2017 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-27736772

RESUMEN

The host response to RNA virus infection consists of an intrinsic innate immune response and the induction of apoptosis as mechanisms to restrict viral replication. The mitochondrial adaptor molecule MAVS plays critical roles in coordinating both virus-induced type I interferon production and apoptosis; however, the regulation of MAVS-mediated apoptosis is poorly understood. Here, we show that the adaptor protein TAX1BP1 functions as a negative regulator of virus-induced apoptosis. TAX1BP1-deficient cells are highly sensitive to apoptosis in response to infection with the RNA viruses vesicular stomatitis virus and Sendai virus and to transfection with poly(I·C). TAX1BP1 undergoes degradation during RNA virus infection, and loss of TAX1BP1 is associated with apoptotic cell death. TAX1BP1 deficiency augments virus-induced activation of proapoptotic c-Jun N-terminal kinase (JNK) signaling. Virus infection promotes the mitochondrial localization of TAX1BP1 and concomitant interaction with the mitochondrial adaptor MAVS. TAX1BP1 recruits the E3 ligase Itch to MAVS to trigger its ubiquitination and degradation, and loss of TAX1BP1 or Itch results in increased MAVS protein expression. Together, these results indicate that TAX1BP1 functions as an adaptor molecule for Itch to target MAVS during RNA virus infection and thus restrict virus-induced apoptosis.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Mitocondrias/metabolismo , Proteínas de Neoplasias/metabolismo , Virus ARN/patogenicidad , Proteínas Represoras/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Animales , Apoptosis , Células HEK293 , Células HeLa , Humanos , Ratones , Virus Sendai/patogenicidad , Ubiquitinación , Vesiculovirus/patogenicidad
11.
Mol Cell Biol ; 36(12): 1776-92, 2016 06 15.
Artículo en Inglés | MEDLINE | ID: mdl-27090639

RESUMEN

We and others have shown that the cystatin E/M gene is inactivated in primary human tumors, pointing to its role as a tumor suppressor gene. However, the molecular mechanism of tumor suppression is not yet understood. Using plasmid-directed cystatin E/M gene overexpression, a lentivirus-mediated tetracycline-inducible vector system, and human papillomavirus 16 (HPV 16) E6 and E7 gene-immortalized normal human epidermal keratinocytes, we demonstrated intracellular and non-cell-autonomous apoptotic growth inhibition of tumor cell lines and that growth inhibition is associated with cytoplasmic retention of NF-κB. We further demonstrated decreased phosphorylation of IκB kinase (IKKß) and IκBα in the presence of tumor necrosis factor alpha (TNF-α), confirming the role of cystatin E/M in the regulation of the NF-κB signaling pathway. Growth suppression of nude mouse xenograft tumors carrying a tetracycline-inducible vector system was observed with the addition of doxycycline in drinking water, confirming that the cystatin E/M gene is a tumor suppressor gene. Finally, immunohistochemical analyses of cervical carcinoma in situ and primary tumors have shown a statistically significant inverse relationship between the expression of cystatin E/M and cathepsin L and a direct relationship between the loss of cystatin E/M expression and nuclear expression of NF-κB. We therefore propose that the cystatin E/M suppressor gene plays an important role in the regulation of NF-κB.


Asunto(s)
Cistatina M/metabolismo , Citoplasma/metabolismo , Proteínas I-kappa B/metabolismo , FN-kappa B/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo , Neoplasias del Cuello Uterino/patología , Animales , Catepsina L/metabolismo , Línea Celular Tumoral , Proliferación Celular , Cistatina M/genética , Doxiciclina/administración & dosificación , Femenino , Regulación Neoplásica de la Expresión Génica , Vectores Genéticos/farmacología , Células HeLa , Humanos , Lentivirus/genética , Ratones , Ratones Desnudos , Trasplante de Neoplasias , Fosforilación , Transducción de Señal , Neoplasias del Cuello Uterino/genética , Neoplasias del Cuello Uterino/metabolismo
12.
PLoS One ; 8(1): e53717, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23308279

RESUMEN

A key feature of the innate antiviral immune response is a rapid nonspecific response to virus infection largely mediated by the induction and extracellular secretion of type I interferons (IFNs) that restrict virus replication. Cytoplasmic sensors such as RIG-I recognize viral RNA and trigger antiviral signaling pathways that upregulate IFN transcription. However, it remains largely unknown how antiviral signaling is negatively regulated to maintain homeostasis after the elimination of virus. In this report, we have identified the RING domain-containing protein RING finger 11 (RNF11) as a novel negative regulator of innate antiviral signaling. Overexpression of RNF11 downregulated IFN-ß expression and enhanced viral replication whereas siRNA-mediated knockdown of RNF11 suppressed viral replication. RNF11 interacted with the noncanonical IKK kinases TBK1/IKKi and attenuated their Lys63-linked polyubiquitination by blocking interactions with the E3 ligase TRAF3. The inhibitory function of RNF11 was dependent on the ubiquitin-binding adaptor molecule TAX1BP1 which was required for RNF11 to target TBK1/IKKi. Collectively, these results indicate that RNF11 functions together with TAX1BP1 to restrict antiviral signaling and IFN-ß production.


Asunto(s)
Proteínas Portadoras/genética , Fibroblastos/inmunología , Regulación de la Expresión Génica , Inmunidad Innata , Péptidos y Proteínas de Señalización Intracelular/genética , Proteínas de Neoplasias/genética , Transducción de Señal , Animales , Proteínas Portadoras/antagonistas & inhibidores , Proteínas Portadoras/inmunología , Línea Celular , Proteínas de Unión al ADN , Fibroblastos/virología , Células HEK293 , Humanos , Interferón beta/biosíntesis , Interferón beta/inmunología , Péptidos y Proteínas de Señalización Intracelular/inmunología , Ratones , Proteínas de Neoplasias/inmunología , Proteínas Serina-Treonina Quinasas/genética , Proteínas Serina-Treonina Quinasas/inmunología , ARN Interferente Pequeño/genética , Factor 3 Asociado a Receptor de TNF/genética , Factor 3 Asociado a Receptor de TNF/inmunología , Ubiquitinación , Vesiculovirus/inmunología , Replicación Viral
13.
Nat Immunol ; 13(12): 1155-61, 2012 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-23142775

RESUMEN

The induction of type I interferons by the bacterial secondary messengers cyclic di-GMP (c-di-GMP) or cyclic di-AMP (c-di-AMP) is dependent on a signaling axis that involves the adaptor STING, the kinase TBK1 and the transcription factor IRF3. Here we identified the heliase DDX41 as a pattern-recognition receptor (PRR) that sensed both c-di-GMP and c-di-AMP. DDX41 specifically and directly interacted with c-di-GMP. Knockdown of DDX41 via short hairpin RNA in mouse or human cells inhibited the induction of genes encoding molecules involved in the innate immune response and resulted in defective activation of STING, TBK1 and IRF3 in response to c-di-GMP or c-di-AMP. Our results suggest a mechanism whereby c-di-GMP and c-di-AMP are detected by DDX41, which forms a complex with STING to signal to TBK1-IRF3 and activate the interferon response.


Asunto(s)
GMP Cíclico/análogos & derivados , ARN Helicasas DEAD-box/metabolismo , Fosfatos de Dinucleósidos/metabolismo , Interferón Tipo I/inmunología , Listeria monocytogenes/inmunología , Listeria monocytogenes/metabolismo , Receptores de Reconocimiento de Patrones/metabolismo , Animales , Línea Celular , GMP Cíclico/metabolismo , ARN Helicasas DEAD-box/genética , Humanos , Inmunidad Innata , Factor 3 Regulador del Interferón/metabolismo , Macrófagos/inmunología , Proteínas de la Membrana/metabolismo , Ratones , Proteínas Serina-Treonina Quinasas/metabolismo , Interferencia de ARN , ARN Interferente Pequeño , Receptores de Reconocimiento de Patrones/genética , Sistemas de Mensajero Secundario , Transducción de Señal
14.
Immunity ; 36(6): 1073-86, 2012 Jun 29.
Artículo en Inglés | MEDLINE | ID: mdl-22579474

RESUMEN

STING is an essential signaling molecule for DNA and cyclic di-GMP (c-di-GMP)-mediated type I interferon (IFN) production via TANK-binding kinase 1 (TBK1) and interferon regulatory factor 3 (IRF3) pathway. It contains an N-terminal transmembrane region and a cytosolic C-terminal domain (CTD). Here, we describe crystal structures of STING CTD alone and complexed with c-di-GMP in a unique binding mode. The strictly conserved aa 153-173 region was shown to be cytosolic and participated in dimerization via hydrophobic interactions. The STING CTD functions as a dimer and the dimerization was independent of posttranslational modifications. Binding of c-di-GMP enhanced interaction of a shorter construct of STING CTD (residues 139-344) with TBK1. This suggests an extra TBK1 binding site, other than serine 358. This study provides a glimpse into the unique architecture of STING and sheds light on the mechanism of c-di-GMP-mediated TBK1 signaling.


Asunto(s)
GMP Cíclico/análogos & derivados , Proteínas de la Membrana/química , Secuencia de Aminoácidos , Secuencia Conservada , Cristalografía por Rayos X , GMP Cíclico/metabolismo , Dimerización , Células HEK293 , Humanos , Interacciones Hidrofóbicas e Hidrofílicas , Proteínas de la Membrana/metabolismo , Modelos Moleculares , Datos de Secuencia Molecular , Fragmentos de Péptidos/química , Fragmentos de Péptidos/metabolismo , Unión Proteica , Conformación Proteica , Mapeo de Interacción de Proteínas , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Recombinantes de Fusión/química , Alineación de Secuencia , Homología de Secuencia de Aminoácido , Relación Estructura-Actividad
15.
Immunity ; 34(6): 821-2, 2011 Jun 24.
Artículo en Inglés | MEDLINE | ID: mdl-21703534

RESUMEN

In this issue of Immunity, Allen et al. (2011) and Xia et al. (2011) provide in vivo and biochemical evidence that NLRX1, a member of the nucleotide binding domain and leucine-rich-repeat-containing protein family, functions as a negative regulator of RIG-I and Toll-like receptors.

16.
Microbes Infect ; 13(3): 209-15, 2011 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-21111841

RESUMEN

The deubiquitinating enzyme A20 (also known as TNFAIP3) is essential for maintaining immune homeostasis. A20 is a key regulator of inflammatory, antiviral and apoptotic signaling pathways. Here, we review recent advances illustrating the role of A20 as an essential negative regulator of inflammatory and antiviral signaling.


Asunto(s)
Inflamación/inmunología , Péptidos y Proteínas de Señalización Intracelular/fisiología , Proteínas Nucleares/fisiología , Virosis/inmunología , Animales , Muerte Celular , Proteínas de Unión al ADN , Humanos , Inflamación/virología , Péptidos y Proteínas de Señalización Intracelular/inmunología , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Proteínas Nucleares/inmunología , Proteínas Nucleares/metabolismo , Transducción de Señal , Proteína 3 Inducida por el Factor de Necrosis Tumoral alfa
18.
J Biol Chem ; 285(20): 14999-15009, 2010 May 14.
Artículo en Inglés | MEDLINE | ID: mdl-20304918

RESUMEN

Induction of type I interferons by the transcription factor IRF3 is essential in the initiation of antiviral innate immunity. Activation of IRF3 requires C-terminal phosphorylation by the upstream kinases TBK1-IKKi, where IRF3 phosphorylation promotes dimerization, and subsequent nuclear translocation to the IFNbeta promoter. Recent studies have described the ubiquitin-editing enzyme A20 as a negative regulator of IRF3 signaling by associating with TBK1-IKKi; however, the regulatory mechanism of A20 inhibition remains unclear. Here we describe the adaptor protein, TAX1BP1, as a key regulator of A20 function in terminating signaling to IRF3. Murine embryonic fibroblasts (MEFs) deficient in TAX1BP1 displayed increased amounts of IFNbeta production upon viral challenge compared with WT MEFs. TAX1BP1 inhibited virus-mediated activation of IRF3 at the level of TBK1-IKKi. TAX1BP1 and A20 blocked antiviral signaling by disrupting Lys(63)-linked polyubiquitination of TBK1-IKKi independently of the A20 deubiquitination domain. Furthermore, TAX1BP1 was required for A20 effector function because A20 was defective for the targeting and inactivation of TBK1 and IKKi in Tax1bp1(-)(/)(-) MEFs. Additionally, we found the E3 ubiquitin ligase TRAF3 to play a critical role in promoting TBK1-IKKi ubiquitination. Collectively, our results demonstrate TBK1-IKKi to be novel substrates for A20 and further identify a novel mechanism whereby A20 and TAX1BP1 restrict antiviral signaling by disrupting a TRAF3-TBK1-IKKi signaling complex.


Asunto(s)
Péptidos y Proteínas de Señalización Intracelular/fisiología , Proteínas de Neoplasias/fisiología , Proteínas Nucleares/fisiología , Proteínas Quinasas/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Transducción de Señal , Vesiculovirus/patogenicidad , Animales , Línea Celular , Proteínas de Unión al ADN , Ensayo de Inmunoadsorción Enzimática , Humanos , Ratones , Proteína 3 Inducida por el Factor de Necrosis Tumoral alfa
19.
EMBO J ; 28(5): 513-22, 2009 Mar 04.
Artículo en Inglés | MEDLINE | ID: mdl-19131965

RESUMEN

The RING domain protein RNF11 is overexpressed in breast cancers and promotes tumour growth factor-beta (TGF-beta) signalling. RNF11 has been proposed to regulate TGF-beta signalling by interacting with HECT- and SCF-type E3 ligases; however, the role of RNF11 in other signalling pathways is poorly understood. Here, we demonstrate a novel function of RNF11 as a negative regulator of NF-kappaB and jun N-terminal kinase (JNK) signalling pathways. Knockdown of RNF11 with siRNA resulted in persistent tumour necrosis factor (TNF)- and lipopolysaccharide (LPS)-mediated NF-kappaB and JNK signalling. RNF11 interacted with the NF-kappaB inhibitor A20 and its regulatory protein TAX1BP1 in a stimulus-dependent manner. RNF11 negatively regulated RIP1 and TRAF6 ubiquitination upon stimulation with TNF and LPS, respectively. Furthermore, RNF11 was required for A20 to interact with and inactivate RIP1 to inhibit TNF-mediated NF-kappaB activation. Our studies reveal that RNF11, together with TAX1BP1 and Itch, is an essential component of an A20 ubiquitin-editing protein complex that ensures transient activation of inflammatory signalling pathways.


Asunto(s)
Proteínas Portadoras/metabolismo , Péptidos y Proteínas de Señalización Intracelular/fisiología , FN-kappa B/fisiología , Proteínas Nucleares/fisiología , Ubiquitina-Proteína Ligasas/fisiología , Proteínas Portadoras/genética , Línea Celular , Proteínas de Unión al ADN , Regulación hacia Abajo , Técnicas de Silenciamiento del Gen , Humanos , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Proteínas Quinasas JNK Activadas por Mitógenos/fisiología , Lipopolisacáridos/farmacología , Monocitos/metabolismo , Proteínas de Neoplasias/metabolismo , Proteínas de Complejo Poro Nuclear/metabolismo , Unión Proteica , Estructura Terciaria de Proteína , ARN Interferente Pequeño/genética , Proteínas de Unión al ARN/metabolismo , Transducción de Señal/fisiología , Factor 6 Asociado a Receptor de TNF/metabolismo , Factor de Crecimiento Transformador beta/farmacología , Proteína 3 Inducida por el Factor de Necrosis Tumoral alfa , Ubiquitinación
20.
Nat Immunol ; 9(3): 254-62, 2008 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-18246070

RESUMEN

The ubiquitin-editing enzyme A20 is a critical negative regulator of inflammation and cytokine-mediated activation of the transcription factor NF-kappaB; however, little is known about the mechanisms of A20-mediated inactivation of signaling intermediates such as RIP1. Here we demonstrate that the regulatory molecule TAX1BP1 recruited the E3 ligase Itch to A20 via two 'PPXY' motifs. Itch was essential for the termination of tumor necrosis factor receptor signaling by controlling A20-mediated recruitment and inactivation of RIP1. Furthermore, the Tax oncoprotein of human T cell leukemia virus type I targeted this complex for inactivation by disrupting the interaction among TAX1BP1, A20 and Itch. Thus, our studies show a previously unappreciated complexity of A20 substrate recognition and inactivation whereby TAX1BP1 and Itch function as essential subunits of an A20 ubiquitin-editing complex.


Asunto(s)
Regulación hacia Abajo/inmunología , Péptidos y Proteínas de Señalización Intracelular/fisiología , Proteínas Nucleares/fisiología , Proteínas Represoras/fisiología , Transducción de Señal/inmunología , Ubiquitina-Proteína Ligasas/fisiología , Animales , Células Cultivadas , Proteínas de Unión al ADN , Eliminación de Gen , Proteínas de Homeodominio/genética , Humanos , MAP Quinasa Quinasa 4/metabolismo , Ratones , Ratones Noqueados , Proteínas de Neoplasias , Proteínas Serina-Treonina Quinasas/metabolismo , Proteína 3 Inducida por el Factor de Necrosis Tumoral alfa , Quinasa de Factor Nuclear kappa B
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...