Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 57
Filtrar
1.
J Invest Dermatol ; 141(10): 2436-2448, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-33864770

RESUMEN

Many inflammatory skin diseases are characterized by altered epidermal differentiation. Whether this altered differentiation promotes inflammatory responses has been unknown. Here, we show that IRAK2, a member of the signaling complex downstream of IL-1 and IL-36, correlates positively with disease severity in both atopic dermatitis and psoriasis. Inhibition of epidermal IRAK2 normalizes differentiation and inflammation in two mouse models of psoriasis- and atopic dermatitis-like inflammation. Specifically, we demonstrate that IRAK2 ties together proinflammatory and differentiation-dependent responses and show that this function of IRAK2 is specific to keratinocytes and acts through the differentiation-associated transcription factor ZNF750. Taken together, our findings suggest that IRAK2 has a critical role in promoting feed-forward amplification of inflammatory responses in skin through modulation of differentiation pathways and inflammatory responses.


Asunto(s)
Epidermis/patología , Inflamación/etiología , Quinasas Asociadas a Receptores de Interleucina-1/fisiología , Diferenciación Celular , Células Cultivadas , Dermatitis Atópica/etiología , Humanos , FN-kappa B/fisiología , Psoriasis/etiología , Índice de Severidad de la Enfermedad , Transducción de Señal , Factores de Transcripción/fisiología , Proteínas Supresoras de Tumor/fisiología
2.
Biosci Biotechnol Biochem ; 85(3): 545-552, 2021 Feb 24.
Artículo en Inglés | MEDLINE | ID: mdl-33590831

RESUMEN

Interleukin-1 receptor-associated kinase-3 (IRAK3) has a distinctive role in regulating inflammation. However, the functional role of IRAK3 and regulatory mechanism underlying the pathogenesis of osteoarthritis (OA) remain unclear. Here, we first found that IRAK3 was upregulated, while miR-33b-3p was downregulated in the cartilage of OA patients and IL-1ß-induced CHON-001 cells. IRAK3 was confirmed as the direct target of miR-33b-3p and negatively regulated by miR-33b-3p. There was an inverse correlation between IRAK3 mRNA expression and miR-33b-3p expression in OA cartilage tissues. The in vitro functional experiments showed that miR-33b-3p overexpression caused a remarkable increase in viability, a significant decrease in inflammatory mediators (IL-1ß and TNF-α), and apoptosis in IL-1ß-induced CHON-001 cells. Importantly, IRAK3 knockdown imitated, while overexpression reversed the effects of miR-33b-3p on IL-1ß-induced inflammation and apoptosis in CHON-001 cells. Collectively, miR-33b-3p significantly alleviated IL-1ß-induced inflammation and apoptosis by downregulating IRAK3, which may serve as a promising target for OA.


Asunto(s)
Apoptosis/fisiología , Condrocitos/patología , Regulación hacia Abajo , Quinasas Asociadas a Receptores de Interleucina-1/fisiología , Modelos Biológicos , Osteoartritis/patología , Línea Celular , Condrocitos/metabolismo , Humanos , Mediadores de Inflamación/metabolismo , Quinasas Asociadas a Receptores de Interleucina-1/genética , Interleucina-1beta/metabolismo , MicroARNs/fisiología , Osteoartritis/metabolismo , ARN Mensajero/genética , Factor de Necrosis Tumoral alfa/metabolismo
3.
Cell Signal ; 48: 69-80, 2018 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-29753111

RESUMEN

The endoplasmic reticulum (ER) is a cellular organelle with central roles in maintaining proteostasis. The accumulation of misfolded proteins in the ER lumen causes ER stress. Cells evoke an evolutionarily conserved adaptive signaling network "unfolded protein response" to restore ER homeostasis, however, how the signaling network is delicately orchestrated remains largely unrevealed. Meanwhile, the HECT type E3 ligase Smad ubiquitylation regulatory factor 1 (Smurf1) has been reported to play critical roles in several important biological pathways by targeting distinct substrates for ubiquitylation, including WFS1, a critical mediator of ER stress, whereas the regulation of Smurf1 activity and abundance upon ER stress are poorly understood. Here, we identified Interleukin-1 Receptor Associated Kinase 2 (IRAK2) as a novel modulator of Smurf1 in response to ER stress induced cell death. Mechanistically, IRAK2 phosphorylates Smurf1 at threonine residues to promote its self-degradation by ubiquitylation, resulting in altered cascade of ER effectors to induce apoptosis. Reduced IRAK2 expression correlates with increased Smurf1 abundance in human colorectal cancer cells. Taken together, these findings demonstrate a novel mechanism of interplays for the different branches of ER stress signaling network and highlight IRAK2 as a potential tumor suppressor to counterbalance oncogenic Smurf1.


Asunto(s)
Carcinogénesis/metabolismo , Neoplasias Colorrectales/metabolismo , Estrés del Retículo Endoplásmico/fisiología , Quinasas Asociadas a Receptores de Interleucina-1/fisiología , Ubiquitina-Proteína Ligasas/metabolismo , Respuesta de Proteína Desplegada/fisiología , Apoptosis , Células HCT116 , Células HEK293 , Células HT29 , Humanos , Fosforilación , Ubiquitinación
4.
PLoS One ; 12(10): e0186614, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-29088270

RESUMEN

Burkitt's lymphoma (BL) is the most common childhood cancer in equatorial Africa, and is endemic to areas where people are chronically co-infected with Epstein-Barr virus (EBV) and the malaria pathogen Plasmodium falciparum. The contribution of these pathogens in the oncogenic process remains poorly understood. We showed earlier that the activation of Toll-like receptor (TLR) 9 by hemozoin, a disposal product formed from the digestion of blood by P. falciparum, suppresses the lytic reactivation of EBV in BL cells. EBV lytic reactivation is regulated by the expression of transcription factor Zta (ZEBRA), encoded by the EBV gene BZLF1. Here, we explore in the BL cell line Akata, the mechanism involved in repression by TLR9 of expression of BZLF1. We show that BZLF1 repression is mediated upon TLR9 engagement by a mechanism that is largely independent of de novo protein synthesis. By CRISPR/Cas9-induced inactivation of TLR9, MyD88, IRAK4 and IRAK1 we confirm that BZLF1 repression is dependent on functional TLR9 and MyD88 signaling, and identify IRAK4 as an essential element for TLR9-induced repression of BZLF1 expression upon BCR cross-linking. Our results unprecedentedly show that TLR9-mediated inhibition of lytic EBV is largely independent of new protein synthesis and demonstrate the central roles of MyD88 and IRAK4 in this process contributing to EBV's persistence in the host's B-cell pool.


Asunto(s)
Linfoma de Burkitt/patología , Herpesvirus Humano 4/genética , Quinasas Asociadas a Receptores de Interleucina-1/fisiología , Receptor Toll-Like 9/fisiología , Transactivadores/genética , Linfoma de Burkitt/virología , Línea Celular Tumoral , Repeticiones Palindrómicas Cortas Agrupadas y Regularmente Espaciadas , Herpesvirus Humano 4/fisiología , Humanos , Quinasas Asociadas a Receptores de Interleucina-1/metabolismo , Factor 88 de Diferenciación Mieloide/metabolismo , Activación Viral
5.
Hepatology ; 64(6): 1978-1993, 2016 12.
Artículo en Inglés | MEDLINE | ID: mdl-27628766

RESUMEN

Lipopolysaccharide (LPS)-mediated activation of Toll-like receptors (TLRs) in hepatic macrophages and injury to hepatocytes are major contributors to the pathogenesis of alcoholic liver disease. However, the mechanisms by which TLR-dependent inflammatory responses and alcohol-induced hepatocellular damage coordinately lead to alcoholic liver disease are not completely understood. In this study, we found that mice deficient in interleukin-1 receptor-associated kinase M (IRAKM), a proximal TLR pathway molecule typically associated with inhibition of TLR signaling, were actually protected from chronic ethanol-induced liver injury. In bone marrow-derived macrophages challenged with low concentrations of LPS, which reflect the relevant pathophysiological levels of LPS in both alcoholic patients and ethanol-fed mice, the IRAKM Myddosome was preferentially formed. Further, the IRAKM Myddosome mediated the up-regulation of Mincle, a sensor for cell death. Mincle-deficient mice were also protected from ethanol-induced liver injury. The endogenous Mincle ligand spliceosome-associated protein 130 (SAP130) is a danger signal released by damaged cells; culture of hepatocytes with ethanol increased the release of SAP130. Ex vivo studies in bone marrow-derived macrophages suggested that SAP130 and LPS synergistically activated inflammatory responses, including inflammasome activation. CONCLUSION: This study reveals a novel IRAKM-Mincle axis that contributes to the pathogenesis of ethanol-induced liver injury. (Hepatology 2016;64:1978-1993).


Asunto(s)
Muerte Celular/fisiología , Inflamación/etiología , Quinasas Asociadas a Receptores de Interleucina-1/fisiología , Lectinas Tipo C/fisiología , Hepatopatías Alcohólicas/fisiopatología , Proteínas de la Membrana/fisiología , Animales , Enfermedad Crónica , Femenino , Lipopolisacáridos/administración & dosificación , Ratones , FN-kappa B/fisiología
6.
Bioessays ; 38(7): 591-604, 2016 07.
Artículo en Inglés | MEDLINE | ID: mdl-27270491

RESUMEN

Interleukin-1 receptor-associated kinase-1 (IRAK1) is linked to the pathogenesis of atherosclerosis; however, its role in macrophage foam cell formation is not known. Therefore, the present study investigated the role of IRAK1 in lipid uptake, biosynthesis, and efflux in THP-1 derived macrophages and human monocyte-derived macrophages (HMDMs). Ox-LDL (40 µg/mL, 15 minutes-48 hours) treatment induced time-dependent increase in IRAK1, IRAK4, and Stat1 activation in THP-1 derived macrophages. IRAK1/4 inhibitor (INH) or IRAK1 siRNA significantly attenuated cholesterol accumulation, DiI-Ox-LDL binding, and uptake while cholesterol efflux to apoAI and HDL was enhanced in THP-1 derived macrophages and HMDMs. Ox-LDL treatment significantly increased the mRNA expression of CD36, LOX-1, SR-A, ABCA1, ABCG1, Caveolin-1, CYP27A1 while that of SR-BI was decreased. IRAK1/4 inhibition or IRAK1 knockdown, however, attenuated Ox-LDL-induced CD36 expression; augmented ABCA1 and ABCG1 expression while expression of others was unaffected in THP-1 derived macrophages and HMDMs. Moreover, IRAK1/4 inhibition had no significant effect on genes involved in lipid biosynthesis. In IRAK1/4 INH pre-treated THP-1 derived macrophages Ox-LDL-induced Stat1 phosphorylation and its binding to CD36 promoter was significantly attenuated while LXRα expression and its binding to the ABCA1/ABCG1 locus, NFATc2 activation and its binding to ABCA1 locus was enhanced. The present study thus demonstrates that IRAK regulates lipid accumulation by modulating CD36-mediated uptake and ABCA1-, ABCG1-dependent cholesterol efflux. Therefore, IRAK1 can be a potential target for preventing macrophage foam cell formation.


Asunto(s)
Aterosclerosis/metabolismo , Colesterol/metabolismo , Células Espumosas/metabolismo , Quinasas Asociadas a Receptores de Interleucina-1/metabolismo , Transportador 1 de Casete de Unión a ATP/genética , Transportador 1 de Casete de Unión a ATP/metabolismo , Transportador de Casetes de Unión a ATP, Subfamilia G, Miembro 1/genética , Transportador de Casetes de Unión a ATP, Subfamilia G, Miembro 1/metabolismo , Animales , Aterosclerosis/patología , Transporte Biológico , Antígenos CD36/genética , Antígenos CD36/metabolismo , Células Espumosas/patología , Regulación de la Expresión Génica , Humanos , Quinasas Asociadas a Receptores de Interleucina-1/fisiología , Fosforilación , Factor de Transcripción STAT1/metabolismo
7.
Proc Natl Acad Sci U S A ; 113(20): 5670-5, 2016 May 17.
Artículo en Inglés | MEDLINE | ID: mdl-27147605

RESUMEN

Genetic polymorphisms in coding genes play an important role when using mouse inbred strains as research models. They have been shown to influence research results, explain phenotypical differences between inbred strains, and increase the amount of interesting gene variants present in the many available inbred lines. SPRET/Ei is an inbred strain derived from Mus spretus that has ∼1% sequence difference with the C57BL/6J reference genome. We obtained a listing of all SNPs and insertions/deletions (indels) present in SPRET/Ei from the Mouse Genomes Project (Wellcome Trust Sanger Institute) and processed these data to obtain an overview of all transcripts having nonsynonymous coding sequence variants. We identified 8,883 unique variants affecting 10,096 different transcripts from 6,328 protein-coding genes, which is about 28% of all coding genes. Because only a subset of these variants results in drastic changes in proteins, we focused on variations that are nonsense mutations that ultimately resulted in a gain of a stop codon. These genes were identified by in silico changing the C57BL/6J coding sequences to the SPRET/Ei sequences, converting them to amino acid (AA) sequences, and comparing the AA sequences. All variants and transcripts affected were also stored in a database, which can be browsed using a SPRET/Ei M. spretus variants web tool (www.spretus.org), including a manual. We validated the tool by demonstrating the loss of function of three proteins predicted to be severely truncated, namely Fas, IRAK2, and IFNγR1.


Asunto(s)
Codón sin Sentido , Ratones Endogámicos/genética , Polimorfismo de Nucleótido Simple , Animales , Ontología de Genes , Quinasas Asociadas a Receptores de Interleucina-1/fisiología , Ratones , Ratones Endogámicos C57BL , Receptores de Interferón/fisiología , Receptor fas/fisiología , Receptor de Interferón gamma
8.
Cell Death Dis ; 6: e1949, 2015 Oct 29.
Artículo en Inglés | MEDLINE | ID: mdl-26512959

RESUMEN

The activation of Toll-like receptor 4 (TLR4) signaling has an important role in promoting lipid accumulation and pro-inflammatory effects in vascular smooth muscle cells (VSMCs), which facilitate atherosclerosis development and progression. Previous studies have demonstrated that excess lipid accumulation in VSMCs is due to an inhibition of the expression of ATP-binding cassette transporter A1 (ABCA1), an important molecular mediator of lipid efflux from VSMCs. However, the underlying molecular mechanisms of this process are unclear. The purpose of this study was to disclose the underlying molecular mechanisms of TLR4 signaling in regulating ABCA1 expression. Primary cultured VSMCs were stimulated with 50 µg/ml oxidized low-density lipoprotein (oxLDL). We determined that enhancing TLR4 signaling using oxLDL significantly downregulated ABCA1 expression and induced lipid accumulation in VSMCs. However, TLR4 knockout significantly rescued oxLDL-induced ABCA1 downregulation and lipid accumulation. In addition, IL-1R-associated kinase 1 (IRAK1) was involved in the effects of TLR4 signaling on ABCA1 expression and lipid accumulation. Silencing IRAK1 expression using a specific siRNA reversed TLR4-induced ABCA1 downregulation and lipid accumulation in vitro. These results were further confirmed by our in vivo experiments. We determined that enhancing TLR4 signaling by administering a 12-week-long high-fat diet (HFD) to mice significantly increased IRAK1 expression, which downregulated ABCA1 expression and induced lipid accumulation. In addition, TLR4 knockout in vivo reversed the effects of the HFD on IRAK1 and ABCA1 expression, as well as on lipid accumulation. In conclusion, IRAK1 is involved in TLR4-mediated downregulation of ABCA1 expression and lipid accumulation in VSMCs.


Asunto(s)
Transportador 1 de Casete de Unión a ATP/genética , Quinasas Asociadas a Receptores de Interleucina-1/fisiología , Metabolismo de los Lípidos , Miocitos del Músculo Liso/metabolismo , Receptor Toll-Like 4/fisiología , Transportador 1 de Casete de Unión a ATP/metabolismo , Animales , Regulación hacia Abajo , Eliminación de Gen , Quinasas Asociadas a Receptores de Interleucina-1/genética , Quinasas Asociadas a Receptores de Interleucina-1/metabolismo , Ratones , Transducción de Señal , Receptor Toll-Like 4/genética , Receptor Toll-Like 4/metabolismo
9.
J Clin Invest ; 125(11): 4239-54, 2015 Nov 02.
Artículo en Inglés | MEDLINE | ID: mdl-26457732

RESUMEN

Hepatocellular carcinoma (HCC) is frequently associated with pathogen infection-induced chronic inflammation. Large numbers of innate immune cells are present in HCCs and can influence disease outcome. Here, we demonstrated that the tumor suppressor serine/threonine-protein kinase 4 (STK4) differentially regulates TLR3/4/9-mediated inflammatory responses in macrophages and thereby is protective against chronic inflammation-associated HCC. STK4 dampened TLR4/9-induced proinflammatory cytokine secretion but enhanced TLR3/4-triggered IFN-ß production via binding to and phosphorylating IL-1 receptor-associated kinase 1 (IRAK1), leading to IRAK1 degradation. Notably, macrophage-specific Stk4 deletion resulted in chronic inflammation, liver fibrosis, and HCC in mice treated with a combination of diethylnitrosamine (DEN) and CCl4, along with either LPS or E. coli infection. STK4 expression was markedly reduced in macrophages isolated from human HCC patients and was inversely associated with the levels of IRAK1, IL-6, and phospho-p65 or phospho-STAT3. Moreover, serum STK4 levels were specifically decreased in HCC patients with high levels of IL-6. In STK4-deficient mice, treatment with an IRAK1/4 inhibitor after DEN administration reduced serum IL-6 levels and liver tumor numbers to levels similar to those observed in the control mice. Together, our results suggest that STK4 has potential as a diagnostic biomarker and therapeutic target for inflammation-induced HCC.


Asunto(s)
Carcinoma Hepatocelular/inmunología , Neoplasias Hepáticas/inmunología , Proteínas Serina-Treonina Quinasas/fisiología , Receptores Toll-Like/inmunología , Animales , Tetracloruro de Carbono/toxicidad , Carcinoma Hepatocelular/química , Carcinoma Hepatocelular/etiología , Citocinas/metabolismo , Dietilnitrosamina , Infecciones por Escherichia coli/complicaciones , Femenino , Células HEK293 , Hepatitis Animal/inducido químicamente , Hepatitis Animal/inmunología , Humanos , Inmunidad Innata , Interferón beta/biosíntesis , Interferón beta/genética , Quinasas Asociadas a Receptores de Interleucina-1/fisiología , Interleucina-6/análisis , Péptidos y Proteínas de Señalización Intracelular , Lipopolisacáridos/toxicidad , Neoplasias Hepáticas/química , Neoplasias Hepáticas/etiología , Neoplasias Hepáticas Experimentales/etiología , Neoplasias Hepáticas Experimentales/genética , Neoplasias Hepáticas Experimentales/inmunología , Neoplasias Hepáticas Experimentales/prevención & control , Pulmón/inmunología , Pulmón/patología , Macrófagos/inmunología , Macrófagos/metabolismo , Masculino , Ratones , Proteínas de Neoplasias/análisis , Fosforilación , Procesamiento Proteico-Postraduccional , Proteínas Serina-Treonina Quinasas/sangre , Proteínas Serina-Treonina Quinasas/deficiencia , Factor de Transcripción STAT3/análisis , Transducción de Señal , Organismos Libres de Patógenos Específicos , Factor de Transcripción ReIA/análisis
10.
Hepatology ; 62(5): 1375-87, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26250868

RESUMEN

UNLABELLED: Patients carrying very rare loss-of-function mutations in interleukin-1 receptor-associated kinase 4 (IRAK4), a critical signaling mediator in Toll-like receptor signaling, are severely immunodeficient, highlighting the paramount role of IRAK kinases in innate immunity. We discovered a comparatively frequent coding variant of the enigmatic human IRAK2, L392V (rs3844283), which is found homozygously in ∼15% of Caucasians, to be associated with a reduced ability to induce interferon-alpha in primary human plasmacytoid dendritic cells in response to hepatitis C virus (HCV). Cytokine production in response to purified Toll-like receptor agonists was also impaired. Additionally, rs3844283 was epidemiologically associated with a chronic course of HCV infection in two independent HCV cohorts and emerged as an independent predictor of chronic HCV disease. Mechanistically, IRAK2 L392V showed intact binding to, but impaired ubiquitination of, tumor necrosis factor receptor-associated factor 6, a vital step in signal transduction. CONCLUSION: Our study highlights IRAK2 and its genetic variants as critical factors and potentially novel biomarkers for human antiviral innate immunity.


Asunto(s)
Hepatitis C Crónica/inmunología , Quinasas Asociadas a Receptores de Interleucina-1/genética , Genotipo , Células HEK293 , Humanos , Interferón-alfa/biosíntesis , Interferones , Quinasas Asociadas a Receptores de Interleucina-1/fisiología , Interleucinas/genética , Polimorfismo de Nucleótido Simple , Factor 6 Asociado a Receptor de TNF/metabolismo , Receptores Toll-Like/fisiología , Ubiquitinación
11.
Br J Cancer ; 112(2): 232-7, 2015 Jan 20.
Artículo en Inglés | MEDLINE | ID: mdl-25290089

RESUMEN

Innate immune signalling has an essential role in inflammation, and the dysregulation of signalling components of this pathway is increasingly being recognised as an important mediator in cancer initiation and progression. In some malignancies, dysregulation of inflammatory toll-like receptor (TLR) and interleukin-1 receptor (IL1R) signalling is typified by increased NF-κB activity, and it occurs through somatic mutations, chromosomal deletions, and/or transcriptional deregulation. Interleukin-1 receptor-associated kinase (IRAK) family members are mediators of TLR/IL1R superfamily signalling, and mounting evidence implicates these kinases as viable cancer targets. Although there have been previous efforts aimed at the development of IRAK kinase inhibitors, this is currently an area of renewed interest for cancer drug development.


Asunto(s)
Quinasas Asociadas a Receptores de Interleucina-1/fisiología , Neoplasias/metabolismo , Transducción de Señal , Animales , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Ensayos de Selección de Medicamentos Antitumorales , Regulación Neoplásica de la Expresión Génica , Humanos , Inmunidad Innata/genética , Quinasas Asociadas a Receptores de Interleucina-1/antagonistas & inhibidores , Terapia Molecular Dirigida , Neoplasias/tratamiento farmacológico , Neoplasias/genética , Neoplasias/inmunología
12.
Proc Natl Acad Sci U S A ; 111(14): 5295-300, 2014 Apr 08.
Artículo en Inglés | MEDLINE | ID: mdl-24706909

RESUMEN

Endotoxemia is caused by excessive inflammation, but the immune system has various mechanisms to avoid collateral organ damage in endotoxemia. A handful of reports have shown that innate immune responses are suppressed by the adaptive immune system. However, the molecular mechanism by which adaptive immune cells suppress innate inflammatory responses is not clear. Here, we report that T cells are shown to interact with macrophages at the early stage of enodotoxemia and to prolong survival of mice through controlling TNF and IL-10 levels by macrophage CD40 stimulation. The cross-talk between CD40 and toll-like receptor (TLR4) signaling first mediates IL-1 receptor-associated kinase 1 (IRAK1) nuclear translocation and its binding to the IL-10 gene promoter in macrophages, without interfering with the NFκB pathway. IL-10 is then detected by macrophages in an autocrine fashion to destabilize Tnfa mRNA. To induce IRAK1-mediated IL-10 expression, signals from both CD40 and TLR4 are essential. CD40 signaling induces IRAK1 sumoylation in the presence of TNF receptor-associated factor 2 (TRAF2) and intracellular isoform of osteopontin (iOPN) whereas TLR4 signaling provides IFN regulatory factor 5 (IRF5) as a chaperone for sumoylated IRAK1 nuclear translocation. Interaction of T cells with macrophages was observed in the spleen in vivo after endotoxemia induction with LPS injection. Our study demonstrates a mechanistic basis for the immunosuppressive role of macrophage CD40 in LPS endotoxemia.


Asunto(s)
Regulación hacia Abajo , Inflamación/inmunología , Quinasas Asociadas a Receptores de Interleucina-1/fisiología , Interleucina-10/fisiología , Macrófagos/inmunología , Linfocitos T/inmunología , Factor de Necrosis Tumoral alfa/biosíntesis , Animales , Antígenos CD/inmunología , Núcleo Celular/metabolismo , Ratones , Transporte de Proteínas , Transducción de Señal , Sumoilación , Receptor Toll-Like 4/metabolismo
13.
Dev Comp Immunol ; 45(1): 190-7, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24631582

RESUMEN

Interleukin 1 (IL-1) receptor-associated kinase (IRAK) family members are crucial signal transducer in the Toll-like receptor/IL-1R signal pathway, which mediates downstream signal cascades involved in the innate and adaptive immune responses. In this study, we identified an IRAK-4 protein (EcIRAK-4) in the orange-spotted grouper (Epinephelus coioides), with an N-terminal death domain, a proST domain, and a central kinase domain, similar to that of other fishes and mammals. A sequence alignment and phylogenic analysis demonstrated that full-length EcIRAK-4 shares a high degree of sequence identity with those of other fishes, especially the roughskin sculpin, and their death domains and kinase domains share greater identity than their proST domains. A conservation analysis indicated that most of the functional sites in mammalian IRAK-4 are conserved in IRAK-4 of the grouper and other fishes, with the exception of the sites of interaction with IRAK-2 and one autophosphorylation site within the activation loop. EcIRAK-4 is broadly expressed in all the tissues examined, with highest expression in the head kidney and liver. After infection with Cryptocaryon irritans, EcIRAK-4 expression was significantly upregulated, especially in the skin, which suggests that this molecule is involved in the host's defense against parasitic infection. Surprisingly, after cotransfection with grouper MyD88, EcIRAK-4 significantly impaired the NF-κB activity induced by MyD88. EcIRAK-4 was uniformly distributed throughout the cytoplasm in HeLa cells. These findings suggest that although IRAK-4 is evolutionarily conserved between fish and mammals, its signal transduction function is markedly different.


Asunto(s)
Infecciones por Cilióforos/veterinaria , Enfermedades de los Peces/enzimología , Proteínas de Peces/fisiología , Quinasas Asociadas a Receptores de Interleucina-1/fisiología , Factor 88 de Diferenciación Mieloide/metabolismo , FN-kappa B/metabolismo , Perciformes/metabolismo , Secuencia de Aminoácidos , Animales , Secuencia de Bases , Infecciones por Cilióforos/enzimología , Infecciones por Cilióforos/inmunología , Enfermedades de los Peces/inmunología , Enfermedades de los Peces/parasitología , Expresión Génica , Células HEK293 , Células HeLa , Humanos , Datos de Secuencia Molecular , Especificidad de Órganos , Perciformes/inmunología , Perciformes/parasitología , Filogenia , Transducción de Señal , Activación Transcripcional/inmunología
14.
J Am Soc Nephrol ; 25(2): 292-304, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24309188

RESUMEN

The mechanisms that determine full recovery versus subsequent progressive CKD after AKI are largely unknown. Because macrophages regulate inflammation as well as epithelial recovery, we investigated whether macrophage activation influences AKI outcomes. IL-1 receptor-associated kinase-M (IRAK-M) is a macrophage-specific inhibitor of Toll-like receptor (TLR) and IL-1 receptor signaling that prevents polarization toward a proinflammatory phenotype. In postischemic kidneys of wild-type mice, IRAK-M expression increased for 3 weeks after AKI and declined thereafter. However, genetic depletion of IRAK-M did not affect immunopathology and renal dysfunction during early postischemic AKI. Regarding long-term outcomes, wild-type kidneys regenerated completely within 5 weeks after AKI. In contrast, IRAK-M(-/-) kidneys progressively lost up to two-thirds of their original mass due to tubule loss, leaving atubular glomeruli and interstitial scarring. Moreover, M1 macrophages accumulated in the renal interstitial compartment, coincident with increased expression of proinflammatory cytokines and chemokines. Injection of bacterial CpG DNA induced the same effects in wild-type mice, and TNF-α blockade with etanercept partially prevented renal atrophy in IRAK-M(-/-) mice. These results suggest that IRAK-M induction during the healing phase of AKI supports the resolution of M1 macrophage- and TNF-α-dependent renal inflammation, allowing structural regeneration and functional recovery of the injured kidney. Conversely, IRAK-M loss-of-function mutations or transient exposure to bacterial DNA may drive persistent inflammatory mononuclear phagocyte infiltrates, which impair kidney regeneration and promote CKD. Overall, these results support a novel role for IRAK-M in the regulation of wound healing and tissue regeneration.


Asunto(s)
Lesión Renal Aguda/patología , Macrófagos/fisiología , Regeneración/fisiología , Daño por Reperfusión/patología , Lesión Renal Aguda/inmunología , Lesión Renal Aguda/fisiopatología , Animales , Atrofia , Etanercept , Femenino , Perfilación de la Expresión Génica , Inmunoglobulina G/farmacología , Quinasas Asociadas a Receptores de Interleucina-1/deficiencia , Quinasas Asociadas a Receptores de Interleucina-1/genética , Quinasas Asociadas a Receptores de Interleucina-1/fisiología , Riñón/fisiología , Túbulos Renales/patología , Activación de Macrófagos , Macrófagos/clasificación , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Fenotipo , Receptores Tipo I de Interleucina-1/antagonistas & inhibidores , Receptores del Factor de Necrosis Tumoral , Receptores Tipo I de Factores de Necrosis Tumoral/antagonistas & inhibidores , Daño por Reperfusión/inmunología , Daño por Reperfusión/fisiopatología , Receptores Toll-Like/fisiología , Factor de Necrosis Tumoral alfa/fisiología
15.
Circulation ; 128(14): 1542-54, 2013 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-24030499

RESUMEN

BACKGROUND: Viral myocarditis follows a fatal course in ≈30% of patients. Interleukin-1 receptor-associated kinase 4 (IRAK4), a major nodal signal transducer in innate immunity, can play a pivotal role in host inflammatory response. We sought to determine how IRAK4 modulates inflammation and outcome in a mouse model of viral myocarditis. METHODS AND RESULTS: Myocarditis was induced after intraperitoneal inoculation of coxsackievirus B3 into C57Bl/6 IRAK4-deficient mice and their littermate controls. Mortality and viral proliferation were markedly reduced in IRAK4(-/-) mice compared with their IRAK4(+/+) littermates. Disease resistance of IRAK4(-/-) mice paralleled increased amounts of protective heart-infiltrating CCR5(+) monocytes/macrophages and enhanced interferon-α and interferon-γ production 2 days after infection. Competitive bone marrow chimera demonstrated that intact IRAK4 function inhibited heart-specific migration of bone marrow-derived CCR5(+) cells. Mechanistically, lack of IRAK4 resulted in interferon regulatory factor 5 homodimerization via reduced melanoma differentiation-associated protein 5 degradation and enhanced Stat1 and Stat5 phosphorylation. Consequently, antiviral interferon-α and interferon-γ production, as well as CCR5(+) cell recruitment, increased, whereas the overall proinflammatory response was drastically reduced in the absence of IRAK4. CONCLUSIONS: Innate immunity signal transducer IRAK4 exacerbates viral myocarditis through inhibition of interferon production and reduced mobilization of protective CCR5(+) monocytes/macrophages to the heart. The combination of IRAK4 inhibitors and antiviral adjuvants may become an attractive therapeutic approach against viral myocarditis in the future.


Asunto(s)
Antígeno CD11b/análisis , Infecciones por Coxsackievirus/inmunología , Interferones/biosíntesis , Quinasas Asociadas a Receptores de Interleucina-1/fisiología , Monocitos/fisiología , Miocarditis/inmunología , Receptores CCR5/análisis , Traslado Adoptivo , Animales , Movimiento Celular/fisiología , Quimiocina CCL5/deficiencia , Quimiocina CCL5/fisiología , Infecciones por Coxsackievirus/fisiopatología , Infecciones por Coxsackievirus/virología , ARN Helicasas DEAD-box/metabolismo , Dimerización , Resistencia a la Enfermedad , Enterovirus Humano B/fisiología , Síndromes de Inmunodeficiencia/genética , Síndromes de Inmunodeficiencia/fisiopatología , Factores Reguladores del Interferón/química , Factores Reguladores del Interferón/metabolismo , Helicasa Inducida por Interferón IFIH1 , Quinasas Asociadas a Receptores de Interleucina-1/deficiencia , Quinasas Asociadas a Receptores de Interleucina-1/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Miocarditis/fisiopatología , Miocarditis/virología , Enfermedades de Inmunodeficiencia Primaria , Procesamiento Proteico-Postraduccional , Quimera por Radiación , Receptores CCR5/deficiencia , Receptores CCR5/fisiología , Factor de Transcripción STAT1/metabolismo , Factor de Transcripción STAT5/metabolismo , Replicación Viral
16.
Cancer Cell ; 24(1): 90-104, 2013 Jul 08.
Artículo en Inglés | MEDLINE | ID: mdl-23845443

RESUMEN

Myelodysplastic syndromes (MDSs) arise from a defective hematopoietic stem/progenitor cell. Consequently, there is an urgent need to develop targeted therapies capable of eliminating the MDS-initiating clones. We identified that IRAK1, an immune-modulating kinase, is overexpressed and hyperactivated in MDSs. MDS clones treated with a small molecule IRAK1 inhibitor (IRAK1/4-Inh) exhibited impaired expansion and increased apoptosis, which coincided with TRAF6/NF-κB inhibition. Suppression of IRAK1, either by RNAi or with IRAK1/4-Inh, is detrimental to MDS cells, while sparing normal CD34(+) cells. Based on an integrative gene expression analysis, we combined IRAK1 and BCL2 inhibitors and found that cotreatment more effectively eliminated MDS clones. In summary, these findings implicate IRAK1 as a drugable target in MDSs.


Asunto(s)
Quinasas Asociadas a Receptores de Interleucina-1/antagonistas & inhibidores , Síndromes Mielodisplásicos/tratamiento farmacológico , Animales , Apoptosis , Línea Celular , Perfilación de la Expresión Génica , Humanos , Quinasas Asociadas a Receptores de Interleucina-1/genética , Quinasas Asociadas a Receptores de Interleucina-1/fisiología , Ratones , Síndromes Mielodisplásicos/patología , FN-kappa B/metabolismo , Fosforilación , Proteínas Proto-Oncogénicas c-bcl-2/antagonistas & inhibidores , Factor 6 Asociado a Receptor de TNF/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto
17.
J Leukoc Biol ; 94(2): 291-300, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23695305

RESUMEN

Prior exposure to LPS induces "endotoxin tolerance" that reprograms TLR4 responses to subsequent LPS challenge by altering expression of inflammatory mediators. Endotoxin tolerance is thought to limit the excessive cytokine storm and prevent tissue damage during sepsis but renders the host immunocompromised and susceptible to secondary infections. Tolerance initiated via one TLR can affect cellular responses to challenge via the same TLR ("homotolerance") or through different TLRs ("heterotolerance"). IRAK4, an essential component of the MyD88-dependent pathway, functions as a kinase and an adapter, activating subsets of divergent signaling pathways. In this study, we addressed mechanistically the role of IRAK4 kinase activity in TLR4- and TLR2-induced tolerance using macrophages from WT versus IRAK4(KDKI) mice. Whereas IRAK4 kinase deficiency decreased LPS signaling, it did not prevent endotoxin tolerance, as endotoxin pretreatment of WT and IRAK4(KDKI) macrophages inhibited LPS-induced MAPK phosphorylation, degradation of IκB-α and recruitment of p65 to the TNF-α promoter, expression of proinflammatory cytokines, and increased levels of A20 and IRAK-M. Pretreatment of WT macrophages with Pam3Cys, a TLR2-TLR1 agonist, ablated p-p38 and p-JNK in response to challenge with Pam3Cys and LPS, whereas IRAK4(KDKI) macrophages exhibited attenuated TLR2-elicited homo- and heterotolerance at the level of MAPK activation. Thus, IRAK4 kinase activity is not required for the induction of endotoxin tolerance but contributes significantly to TLR2-elicited homo- and heterotolerance.


Asunto(s)
Endotoxinas/inmunología , Quinasas Asociadas a Receptores de Interleucina-1/fisiología , Lipopolisacáridos/toxicidad , Macrófagos Peritoneales/inmunología , Receptor Toll-Like 2/fisiología , Receptor Toll-Like 4/fisiología , Animales , Cisteína Endopeptidasas , Citocinas/biosíntesis , Citocinas/genética , Proteínas de Unión al ADN/biosíntesis , Proteínas de Unión al ADN/genética , Tolerancia a Medicamentos , Endotoxinas/toxicidad , Activación Enzimática , Femenino , Regulación de la Expresión Génica/efectos de los fármacos , Regulación de la Expresión Génica/inmunología , Síndromes de Inmunodeficiencia/genética , Síndromes de Inmunodeficiencia/inmunología , Quinasas Asociadas a Receptores de Interleucina-1/biosíntesis , Quinasas Asociadas a Receptores de Interleucina-1/genética , Quinasas Asociadas a Receptores de Interleucina-1/inmunología , Péptidos y Proteínas de Señalización Intracelular/biosíntesis , Péptidos y Proteínas de Señalización Intracelular/genética , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Lipoproteínas/farmacología , Sistema de Señalización de MAP Quinasas/fisiología , Macrófagos Peritoneales/efectos de los fármacos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Factor 88 de Diferenciación Mieloide/fisiología , FN-kappa B/metabolismo , Enfermedades de Inmunodeficiencia Primaria , Transducción de Señal , Receptor Toll-Like 2/agonistas , Proteína 3 Inducida por el Factor de Necrosis Tumoral alfa , Ubiquitina-Proteína Ligasas/biosíntesis , Ubiquitina-Proteína Ligasas/genética , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
18.
Inflamm Bowel Dis ; 19(6): 1266-77, 2013 May.
Artículo en Inglés | MEDLINE | ID: mdl-23567778

RESUMEN

BACKGROUND: Microbial sensing by Toll-like receptors (TLR) and its negative regulation have an important role in the pathogenesis of inflammation-related cancer. In this study, we investigated the role of negative regulation of Toll-like receptors signaling and gut microbiota in the development of colitis-associated cancer in mouse model. METHODS: Colitis-associated cancer was induced by azoxymethane and dextran sodium sulfate in wild-type and in interleukin-1 receptor-associated kinase M (IRAK-M)-deficient mice with or without antibiotic (ATB) treatment. Local cytokine production was analyzed by multiplex cytokine assay or enzyme-linked immunosorbent assay, and regulatory T cells were analyzed by flow cytometry. Changes in microbiota composition during tumorigenesis were analyzed by pyrosequencing, and ß-glucuronidase activity was measured in intestinal content by fluorescence assay. RESULTS: ATB treatment of wild-type mice reduced the incidence and severity of tumors. Compared with nontreated mice, ATB-treated mice had significantly lower numbers of regulatory T cells in colon, altered gut microbiota composition, and decreased ß-glucuronidase activity. However, the ß-glucuronidase activity was not as low as in germ-free mice. IRAK-M-deficient mice not only developed invasive tumors, but ATB-induced decrease in ß-glucuronidase activity did not rescue them from severe carcinogenesis phenotype. Furthermore, IRAK-M-deficient mice had significantly increased levels of proinflammatory cytokines in the tumor tissue. CONCLUSIONS: We conclude that gut microbiota promotes tumorigenesis by increasing the exposure of gut epithelium to carcinogens and that IRAK-M-negative regulation is essential for colon cancer resistance even in conditions of altered microbiota. Therefore, gut microbiota and its metabolic activity could be potential targets for colitis-associated cancer therapy.


Asunto(s)
Colitis/complicaciones , Neoplasias del Colon/etiología , Tracto Gastrointestinal/microbiología , Quinasas Asociadas a Receptores de Interleucina-1/fisiología , Metagenoma , Animales , Azoximetano/toxicidad , Western Blotting , Carcinógenos/toxicidad , Colitis/inducido químicamente , Neoplasias del Colon/metabolismo , Neoplasias del Colon/patología , Citocinas/genética , Citocinas/metabolismo , Sulfato de Dextran/toxicidad , Femenino , Citometría de Flujo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , ARN Mensajero/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Receptores de Interleucina-1/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal , Linfocitos T Reguladores/inmunología , Linfocitos T Reguladores/metabolismo , Linfocitos T Reguladores/patología , Receptores Toll-Like/genética , Receptores Toll-Like/metabolismo
19.
J Immunol ; 190(6): 2844-56, 2013 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-23396947

RESUMEN

IRAK1 is involved in the regulation of type I IFN production downstream of TLR3. Previous work indicated that IRAK1 negatively regulates TRIF-mediated activation of IRF3 and IRF7. We report that IRAK1 limits the activation of the TLR3-NF-κB pathway. Following TLR3 stimulation, IRAK1-deficient macrophages produced increased levels of IL-6 and IFN-ß compared with wild type macrophages. Pharmacological inhibition of TAK1 reduced this increase in IFN-ß, together with the heightened activation of IRF3 and p65 found in TLR3-ligand stimulated IRAK1-deficient macrophages. Recently, IKKε and TANK-binding kinase 1 (TBK1) were reported to limit activation of the NF-κB pathway downstream of IL-1R, TNFR1, and TLRs. We show that TBK1 has a positive role in the TLR3-NF-κB pathway, because we detected reduced levels of IL-6 and reduced activation of p65 in TBK1-deficient macrophages. In contrast, we show that IKKε limits the activation of the TLR3-NF-κB pathway. Furthermore, we show that IRAK1 is required for the activation of IKKε downstream of TLR3. We report impaired activation of ERK1/2 in IRAK1- and IKKε-deficient macrophages, a novel finding for both kinases. Importantly, this work provides novel mechanistic insight into the regulation of the TLR3-signaling pathway, providing strong evidence that an IRAK1-IKKε-signaling axis acts to limit the production of both type I IFNs and proinflammatory cytokines by regulating TAK1 activity.


Asunto(s)
Proteínas Portadoras/metabolismo , Regulación hacia Abajo/inmunología , Quinasa I-kappa B/fisiología , Quinasas Asociadas a Receptores de Interleucina-1/fisiología , Quinasas Quinasa Quinasa PAM/metabolismo , Sistema de Señalización de MAP Quinasas/inmunología , Receptor Toll-Like 3/metabolismo , Animales , Proteínas Portadoras/antagonistas & inhibidores , Línea Celular , Regulación hacia Abajo/genética , Células HEK293 , Humanos , Quinasa I-kappa B/genética , Inflamación/enzimología , Inflamación/genética , Inflamación/inmunología , Quinasas Asociadas a Receptores de Interleucina-1/genética , Péptidos y Proteínas de Señalización Intracelular , Quinasas Quinasa Quinasa PAM/antagonistas & inhibidores , Quinasas Quinasa Quinasa PAM/genética , Sistema de Señalización de MAP Quinasas/genética , Ratones , Microglía/enzimología , Microglía/inmunología , Microglía/patología , Mapeo de Interacción de Proteínas , Receptor Toll-Like 3/genética , Factores de Elongación Transcripcional
20.
Uirusu ; 62(1): 39-45, 2012 Jun.
Artículo en Japonés | MEDLINE | ID: mdl-23189823

RESUMEN

The innate immune system detects pathogen-derived nucleic acids (DNA and RNA) and induces type I interferon (IFN) and other cytokines, resulting in the host defense against pathogen. We identified interferon-inducible tripartite-motif (TRIM) 56 as a regulator of double-stranded DNA-mediated type I interferon induction. TRIM56 interacted with STING and targeted it for lysine 63-linked ubiquitination. This modification induced STING dimerization, which was a prerequisite for recruitment of the antiviral kinase TBK1 and subsequent induction of IFN-beta. Taken together, these results show that TRIM56 is an interferon-inducible E3 ubiquitin ligase that modulates STING to confer double-stranded DNA-mediated innate immune responses. It is well known that Toll-like receptor 7 (TLR7) and TLR9 sense viral nucleic acids and induce production of type I interferon (IFN) by plasmacytoid dendritic cells (pDCs) to protect the host from virus infection. We showed that the IFN-inducible antiviral protein Viperin promoted TLR7- and TLR9-mediated production of type I IFN by pDCs. Viperin expression was potently induced after TLR7 or TLR9 stimulation and Viperin localized to the cytoplasmic lipid-enriched compartments, lipid bodies, in pDCs. Viperin interacted with the signal mediators IRAK1 and TRAF6 to recruit them to the lipid bodies and facilitated K63-linked ubiquitination of IRAK1 to induce the nuclear translocation of transcription factor IRF7. Thus, besides direct inhibition of viral replication, this finding reveals that Viperin mediates its antiviral function via the regulation of the TLR7 and TLR9-IRAK1 signaling axis in pDCs.


Asunto(s)
ADN Bacteriano/inmunología , ADN Viral/inmunología , Inmunidad Innata/inmunología , ARN Bacteriano/inmunología , ARN Viral/inmunología , Ubiquitina-Proteína Ligasas/inmunología , Animales , Humanos , Factor 7 Regulador del Interferón , Interferón Tipo I/biosíntesis , Quinasas Asociadas a Receptores de Interleucina-1/fisiología , Proteínas de la Membrana/metabolismo , Oxidorreductasas actuantes sobre Donantes de Grupo CH-CH , Proteínas/inmunología , Transducción de Señal , Receptor Toll-Like 7/inmunología , Receptor Toll-Like 9/inmunología , Proteínas de Motivos Tripartitos , Ubiquitinación
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA