Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
1.
Nat Immunol ; 21(10): 1219-1231, 2020 10.
Artigo em Inglês | MEDLINE | ID: mdl-32778760

RESUMO

Chronic inflammation is a common feature of obesity, with elevated cytokines such as interleukin-1 (IL-1) in the circulation and tissues. Here, we report an unconventional IL-1R-MyD88-IRAK2-PHB/OPA1 signaling axis that reprograms mitochondrial metabolism in adipocytes to exacerbate obesity. IL-1 induced recruitment of IRAK2 Myddosome to mitochondria outer membranes via recognition by TOM20, followed by TIMM50-guided translocation of IRAK2 into mitochondria inner membranes, to suppress oxidative phosphorylation and fatty acid oxidation, thereby attenuating energy expenditure. Adipocyte-specific MyD88 or IRAK2 deficiency reduced high-fat-diet-induced weight gain, increased energy expenditure and ameliorated insulin resistance, associated with a smaller adipocyte size and increased cristae formation. IRAK2 kinase inactivation also reduced high-fat diet-induced metabolic diseases. Mechanistically, IRAK2 suppressed respiratory super-complex formation via interaction with PHB1 and OPA1 upon stimulation of IL-1. Taken together, our results suggest that the IRAK2 Myddosome functions as a critical link between inflammation and metabolism, representing a novel therapeutic target for patients with obesity.


Assuntos
Adipócitos/imunologia , Inflamação/imunologia , Quinases Associadas a Receptores de Interleucina-1/metabolismo , Interleucina-1/metabolismo , Membranas Mitocondriais/metabolismo , Obesidade/imunologia , Adipócitos/patologia , Animais , Células Cultivadas , Humanos , Quinases Associadas a Receptores de Interleucina-1/genética , Masculino , Camundongos , Camundongos Knockout , Fator 88 de Diferenciação Mieloide/genética , Fator 88 de Diferenciação Mieloide/metabolismo , Fosforilação Oxidativa , Proibitinas , Transporte Proteico , Receptores de Interleucina-1/metabolismo , Transdução de Sinais
2.
Emerg Infect Dis ; 26(12): 2951-2955, 2020 12.
Artigo em Inglês | MEDLINE | ID: mdl-32809930

RESUMO

An ongoing outbreak of multidrug-resistant Salmonella enterica serovar Anatum began in Taiwan in 2015. Pork and poultry were identified as vehicles for transmission. Contaminated meat contributed to the high rate of infections among children. Nearly identical Salmonella Anatum strains have been identified in the United Kingdom, the United States, and the Philippines.


Assuntos
Antibacterianos , Salmonella enterica , Salmonella , Animais , Antibacterianos/farmacologia , Criança , Surtos de Doenças , Farmacorresistência Bacteriana Múltipla/efeitos dos fármacos , Humanos , Carne , Testes de Sensibilidade Microbiana , Filipinas , Salmonella/efeitos dos fármacos , Salmonella/genética , Salmonella/isolamento & purificação , Salmonella enterica/efeitos dos fármacos , Salmonella enterica/genética , Taiwan/epidemiologia , Reino Unido , Estados Unidos
3.
J Hepatol ; 73(6): 1470-1481, 2020 12.
Artigo em Inglês | MEDLINE | ID: mdl-32682051

RESUMO

BACKGROUNDS & AIMS: Alcohol-related liver disease (ALD) is a major cause of chronic liver disease worldwide with limited therapeutic options. Interleukin-1 receptor associated kinase 4 (IRAK4), the master kinase of Toll-like receptor (TLR)/IL-1R-mediated signalling activation, is considered a novel therapeutic target in inflammatory diseases, but has not been investigated in the context of ALD. METHODS: IRAK4 phosphorylation and IRAK1 protein were analysed in liver from alcohol-related hepatitis patients and healthy controls. IRAK4 kinase activity-inactive knock-in (Irak4 KI) mice and bone marrow chimeric mice were exposed to chronic ethanol-induced liver injury. IL-1ß-induced IRAK4-mediated signalling and acute phase response were investigated in cultured hepatocytes. IRAK1/4 inhibitor was used to test the therapeutic potential for ethanol-induced liver injury in mice. RESULTS: Increased IRAK4 phosphorylation and reduced IRAK1 protein were found in livers of patients with alcoholic hepatitis. In the chronic ethanol-induced liver injury mouse model, hepatic inflammation and hepatocellular damage were attenuated in Irak4 KI mice. IRAK4 kinase activity promotes expression of acute phase proteins in response to ethanol exposure, including C-reactive protein and serum amyloid A1 (SAA1). SAA1 and IL-1ß synergistically exacerbate ethanol-induced cell death ex vivo. Pharmacological blockage of IRAK4 kinase abrogated ethanol-induced liver injury, inflammation, steatosis, as well as acute phase gene expression and protein production in mice. CONCLUSIONS: Our data elucidate the critical role of IRAK4 kinase activity in the pathogenesis of ethanol-induced liver injury in mice and provide preclinical validation for use of an IRAK1/4 inhibitor as a new potential therapeutic strategy for the treatment of ALD. LAY SUMMARY: Herein, we have identified the role of IRAK4 kinase activity in the development of alcohol-induced liver injury in mice. Hepatocyte-specific IRAK4 is associated with an acute phase response and release of proinflammatory cytokines/chemokines, which synergistically exacerbate alcohol-induced hepatocyte cell death ex vivo. Pharmacological inhibition of IRAK4 kinase activity effectively attenuates alcohol-induced liver injury in mice and could have therapeutic implications.


Assuntos
Doença Hepática Induzida por Substâncias e Drogas , Inibidores de Checkpoint Imunológico/farmacologia , Quinases Associadas a Receptores de Interleucina-1/antagonistas & inibidores , Quinases Associadas a Receptores de Interleucina-1/metabolismo , Hepatopatias Alcoólicas/metabolismo , Proteínas de Fase Aguda/metabolismo , Animais , Células Cultivadas , Doença Hepática Induzida por Substâncias e Drogas/tratamento farmacológico , Doença Hepática Induzida por Substâncias e Drogas/metabolismo , Humanos , Interleucina-1beta/metabolismo , Hepatopatias Alcoólicas/patologia , Camundongos , Camundongos Endogâmicos C57BL , Fragmentos de Peptídeos/metabolismo , Fosforilação , Receptores de Interleucina-1/metabolismo , Transdução de Sinais/efeitos dos fármacos , Receptores Toll-Like/metabolismo , Resultado do Tratamento
4.
Pediatr Neonatol ; 61(4): 432-438, 2020 08.
Artigo em Inglês | MEDLINE | ID: mdl-32340872

RESUMO

BACKGROUND: The epidemiology of nontyphoidal Salmonella (NTS) resistant to ciprofloxacin or ceftriaxone and its impact on patients' clinical course are rarely reported. METHODS: Children with culture-proven salmonellosis treated in a medical center in northern Taiwan in 2017 were enrolled. To trace the source of Salmonella, Salmonella isolated from food samples were collected from markets. Antimicrobial susceptibility and serotypes were determined. RESULTS: Among the 453 isolates, 122 (26.9%) were highly antimicrobial-resistant, as defined by resistance to ciprofloxacin or ceftriaxone or both. The most prevalent highly resistant serotype was S. Anatum (66, 54.1%). Salmonella was detected in 94.1%, 66.7%, and 8.6% of examined pork, chicken, and vegetables examined, respectively. S. Anatum (6, 21.4%) and S. Derby (6, 21.4%) were the major serotypes isolated. Majority of the S. Anatum (5, 83.3%) were highly antimicrobial-resistant. More patients infected by highly resistant Salmonella required carbapenem treatment (OR = 23.5, 95% confidence interval [CI] 2.8-192.7, P < 0.001). Patients with ceftriaxone-resistant NTS infections had a significantly longer hospital stay than others (P < 0.001). Totally, 34 (7.5%) presented with invasive diseases (31 bacteremia, 1 intestinal perforation, 1 toxic megacolon and 1 septic arthritis). Risk factors for invasive disease included prolonged fever for ≧5 days and infection occurring in warmer season from May to October. The rise of ambient temperature in northern Taiwan was associated with increasing Salmonella infections. CONCLUSIONS: Retail meats were the main source of highly antimicrobial-resistant NTS in northern Taiwan. Highly antimicrobial resistance significantly impacted the clinical course and treatment of children with NTS infection.


Assuntos
Farmacorresistência Bacteriana , Carne/microbiologia , Intoxicação Alimentar por Salmonella/tratamento farmacológico , Intoxicação Alimentar por Salmonella/epidemiologia , Adolescente , Animais , Antibacterianos/uso terapêutico , Artrite Infecciosa/etiologia , Bacteriemia/etiologia , Carbapenêmicos/uso terapêutico , Bovinos , Galinhas , Criança , Pré-Escolar , Feminino , Humanos , Lactente , Perfuração Intestinal/etiologia , Masculino , Megacolo Tóxico/etiologia , Testes de Sensibilidade Microbiana , Salmonella/isolamento & purificação , Suínos , Taiwan/epidemiologia
5.
J Biol Chem ; 295(15): 4849-4857, 2020 04 10.
Artigo em Inglês | MEDLINE | ID: mdl-32107314

RESUMO

Monocytes are rapidly recruited to sites of diabetic complications and differentiate into macrophages. Previously, we showed that rat kidney mesangial cells dividing during hyperglycemic stress abnormally synthesize hyaluronan (HA) in intracellular compartments. This initiates a stress response, resulting in an extracellular HA matrix after division that recruits inflammatory cells. Cell-cell communication among macrophages that are recruited into the glomeruli and the damaged rat mesangial cells leads to diabetic nephropathy, fibrosis, and proteinurea, which are inhibited in heparin-treated diabetic rats. In this study, we found that murine bone marrow-derived macrophages (BMDMs) and a human leukemic cell line, U937 cells, dividing in hyperglycemia also accumulate intracellular HA and that heparin inhibits the HA accumulation. Both cell types expressed increased levels of proinflammatory markers: inducible nitric-oxide synthase and tumor necrosis factor-α, when cultured under hyperglycemic stress, which was inhibited by heparin. Furthermore, the abnormal intracellular HA was also observed in peripheral blood monocytes derived from three different hyperglycemic diabetic mouse models: streptozotocin-treated, high-fat fed, and Ins2Akita. Moreover, peripheral blood monocytes in humans with type 2 diabetes and poorly controlled blood glucose levels (hemoglobin A1c (HbA1c) levels of >7) also had intracellular HA, whereas those with HbA1c of <7, did not. Of note, heparin increased the anti-inflammatory markers arginase 1 and interleukin-10 in murine BMDMs. We conclude that heparin treatment of high glucose-exposed dividing BMDMs promotes an anti-inflammatory tissue-repair phenotype in these cells.


Assuntos
Anti-Inflamatórios/farmacologia , Diabetes Mellitus Experimental/fisiopatologia , Diabetes Mellitus Tipo 2/fisiopatologia , Heparina/farmacologia , Hiperglicemia/patologia , Inflamação/prevenção & controle , Macrófagos/imunologia , Animais , Anticoagulantes/farmacologia , Arginase/metabolismo , Matriz Extracelular/metabolismo , Feminino , Glucose/metabolismo , Humanos , Hiperglicemia/imunologia , Hiperglicemia/metabolismo , Inflamação/etiologia , Inflamação/metabolismo , Inflamação/patologia , Mediadores da Inflamação/imunologia , Mediadores da Inflamação/metabolismo , Interleucina-10/metabolismo , Macrófagos/efeitos dos fármacos , Macrófagos/metabolismo , Camundongos , Monócitos/efeitos dos fármacos , Monócitos/imunologia , Monócitos/metabolismo
6.
Open Forum Infect Dis ; 6(12): ofz447, 2019 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-31858016

RESUMO

BACKGROUND: Nontyphoidal Salmonella (NTS) is an important foodborne pathogen worldwide. We investigated a 2018 outbreak of highly antimicrobial-resistant Salmonella enterica serotype Goldcoast in northern Taiwan. METHODS: We collected 30 clinical isolates and 2 meat isolates from this outbreak in New Taipei and Taoyuan, Taiwan in 2018. The clinical manifestations and the treatment of the patients were reviewed. To trace the source, we examined NTS isolated from food samples collected from the markets in northern Taiwan. All of the isolates along with an additional human isolate from China were sequenced and compared with the sequences of Salmonella Goldcoast reported by other countries. RESULTS: The outbreak involved 14 pediatric patients (<5 years old) and 16 adults (36 to 83 years old). Nine patients with invasive or severe disease required carbapenem treatment. The MIC90 of ceftriaxone and ciprofloxacin for the outbreak isolates was >256 µg/mL and 1 µg/mL, respectively, and a conjugative 278-kilobase plasmid harboring bla CTX-M-55 and qnrS1 contributed towards the resistance. Whole-genome sequencing revealed a clonal relationship among the outbreak isolates and the 2 collected from the retail meats. The outbreak clone was phylogenetically close to that of Salmonella Goldcoast reported in the United Kingdom, Poland, and China, whereas similar resistance plasmids were found in China and Cambodia. CONCLUSIONS: The clinical spectrum of the high-level cephalosporin-resistant Salmonella Goldcoast is similar to that of other NTS serotypes, but severe cases required carbapenem treatment. The study confirmed the emergence of a highly antimicrobial-resistant clone of Salmonella Goldcoast, highlighting the importance of surveillance for food safety.

7.
Elife ; 62017 10 09.
Artigo em Inglês | MEDLINE | ID: mdl-28990926

RESUMO

Expression of inflammatory genes is determined in part by post-transcriptional regulation of mRNA metabolism but how stimulus- and transcript-dependent nuclear export influence is poorly understood. Here, we report a novel pathway in which LPS/TLR4 engagement promotes nuclear localization of IRAK2 to facilitate nuclear export of a specific subset of inflammation-related mRNAs for translation in murine macrophages. IRAK2 kinase activity is required for LPS-induced RanBP2-mediated IRAK2 sumoylation and subsequent nuclear translocation. Array analysis showed that an SRSF1-binding motif is enriched in mRNAs dependent on IRAK2 for nuclear export. Nuclear IRAK2 phosphorylates SRSF1 to reduce its binding to target mRNAs, which promotes the RNA binding of the nuclear export adaptor ALYREF and nuclear export receptor Nxf1 loading for the export of the mRNAs. In summary, LPS activates a nuclear function of IRAK2 that facilitates the assembly of nuclear export machinery to export selected inflammatory mRNAs to the cytoplasm for translation.


Assuntos
Transporte Ativo do Núcleo Celular , Quinases Associadas a Receptores de Interleucina-1/metabolismo , Macrófagos/imunologia , RNA Mensageiro/metabolismo , Animais , Lipopolissacarídeos/metabolismo , Macrófagos/efeitos dos fármacos , Camundongos , Proteínas de Transporte Nucleocitoplasmático/metabolismo , Fosforilação , Proteínas de Ligação a RNA/metabolismo , Fatores de Processamento de Serina-Arginina/metabolismo , Sumoilação
8.
Hepatology ; 64(6): 1978-1993, 2016 12.
Artigo em Inglês | MEDLINE | ID: mdl-27628766

RESUMO

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).


Assuntos
Morte Celular/fisiologia , Inflamação/etiologia , Quinases Associadas a Receptores de Interleucina-1/fisiologia , Lectinas Tipo C/fisiologia , Hepatopatias Alcoólicas/fisiopatologia , Proteínas de Membrana/fisiologia , Animais , Doença Crônica , Feminino , Lipopolissacarídeos/administração & dosagem , Camundongos , NF-kappa B/fisiologia
9.
Gastroenterology ; 149(7): 1860-1871.e8, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26344057

RESUMO

BACKGROUND & AIMS: Single immunoglobulin and toll-interleukin 1 receptor (SIGIRR), a negative regulator of the Toll-like and interleukin-1 receptor (IL-1R) signaling pathways, controls intestinal inflammation and suppresses colon tumorigenesis in mice. However, the importance of SIGIRR in human colorectal cancer development has not been determined. We investigated the role of SIGIRR in development of human colorectal cancer. METHODS: We performed RNA sequence analyses of pairs of colon tumor and nontumor tissues, each collected from 68 patients. Immunoblot and immunofluorescence analyses were used to determine levels of SIGIRR protein in primary human colonic epithelial cells, tumor tissues, and colon cancer cell lines. We expressed SIGIRR and mutant forms of the protein in Vaco cell lines. We created and analyzed mice that expressed full-length (control) or a mutant form of Sigirr (encoding SIGIRR(N86/102S), which is not glycosylated) specifically in the intestinal epithelium. Some mice were given azoxymethane (AOM) and dextran sulfate sodium to induce colitis-associated cancer. Intestinal tissues were collected and analyzed by immunohistochemical and gene expression profile analyses. RESULTS: RNA sequence analyses revealed increased expression of a SIGIRR mRNA isoform, SIGIRR(ΔE8), in colorectal cancer tissues compared to paired nontumor tissues. SIGIRR(ΔE8) is not modified by complex glycans and is therefore retained in the cytoplasm-it cannot localize to the cell membrane or reduce IL1R signaling. SIGIRR(ΔE8) interacts with and has a dominant-negative effect on SIGIRR, reducing its glycosylation, localization to the cell surface, and function. Most SIGIRR detected in human colon cancer tissues was cytoplasmic, whereas in nontumor tissues it was found at the cell membrane. Mice that expressed SIGIRR(N86/102S) developed more inflammation and formed larger tumors after administration of azoxymethane and dextran sulfate sodium than control mice; colon tissues from these mutant mice expressed higher levels of the inflammatory cytokines IL-17A and IL-6 had activation of the transcription factors STAT3 and NFκB. SIGIRR(N86/102S) expressed in colons of mice did not localize to the epithelial cell surface. CONCLUSION: Levels of SIGIRR are lower in human colorectal tumors, compared with nontumor tissues; tumors contain the dominant-negative isoform SIGIRR(ΔE8). This mutant protein blocks localization of full-length SIGIRR to the surface of colon epithelial cells and its ability to downregulate IL1R signaling. Expression of SIGIRR(N86/102S) in the colonic epithelium of mice increases expression of inflammatory cytokines and formation and size of colitis-associated tumors.


Assuntos
Colite/metabolismo , Colo/metabolismo , Neoplasias do Colo/metabolismo , Genes Dominantes , Mucosa Intestinal/metabolismo , Receptores de Interleucina-1/metabolismo , Animais , Azoximetano , Membrana Celular/metabolismo , Colite/induzido quimicamente , Colite/genética , Colite/patologia , Colo/patologia , Neoplasias do Colo/induzido quimicamente , Neoplasias do Colo/genética , Neoplasias do Colo/patologia , Citosol/metabolismo , Sulfato de Dextrana , Perfilação da Expressão Gênica/métodos , Regulação Neoplásica da Expressão Gênica , Predisposição Genética para Doença , Glicosilação , Células HeLa , Humanos , Mediadores da Inflamação/metabolismo , Mucosa Intestinal/patologia , Camundongos da Linhagem 129 , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Mutação , Análise de Sequência com Séries de Oligonucleotídeos , Fenótipo , Processamento de Proteína Pós-Traducional , RNA Mensageiro/metabolismo , Receptores de Interleucina-1/genética , Transdução de Sinais , Técnicas de Cultura de Tecidos , Transfecção
10.
J Exp Med ; 211(5): 887-907, 2014 May 05.
Artigo em Inglês | MEDLINE | ID: mdl-24752299

RESUMO

Low-grade systemic inflammation is often associated with metabolic syndrome, which plays a critical role in the development of the obesity-associated inflammatory diseases, including insulin resistance and atherosclerosis. Here, we investigate how Toll-like receptor-MyD88 signaling in myeloid and endothelial cells coordinately participates in the initiation and progression of high fat diet-induced systemic inflammation and metabolic inflammatory diseases. MyD88 deficiency in myeloid cells inhibits macrophage recruitment to adipose tissue and their switch to an M1-like phenotype. This is accompanied by substantially reduced diet-induced systemic inflammation, insulin resistance, and atherosclerosis. MyD88 deficiency in endothelial cells results in a moderate reduction in diet-induced adipose macrophage infiltration and M1 polarization, selective insulin sensitivity in adipose tissue, and amelioration of spontaneous atherosclerosis. Both in vivo and ex vivo studies suggest that MyD88-dependent GM-CSF production from the endothelial cells might play a critical role in the initiation of obesity-associated inflammation and development of atherosclerosis by priming the monocytes in the adipose and arterial tissues to differentiate into M1-like inflammatory macrophages. Collectively, these results implicate a critical MyD88-dependent interplay between myeloid and endothelial cells in the initiation and progression of obesity-associated inflammatory diseases.


Assuntos
Células Endoteliais/metabolismo , Inflamação/metabolismo , Inflamação/fisiopatologia , Insulina/metabolismo , Células Mieloides/metabolismo , Fator 88 de Diferenciação Mieloide/metabolismo , Obesidade/complicações , Análise de Variância , Animais , Antígeno CD11b/metabolismo , Citometria de Fluxo , Imuno-Histoquímica , Inflamação/etiologia , Camundongos , Reação em Cadeia da Polimerase em Tempo Real
11.
PLoS One ; 8(5): e64256, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23724040

RESUMO

Endoplasmic reticulum (ER) stress occurs when unfolded proteins accumulate in the lumen of the organelle, triggering signal transduction events that contribute either to cellular adaptation and recovery or alternatively to cellular dysfunction and death. ER stress has been implicated in numerous diseases. To identify novel modulators of ER stress, we undertook a siRNA library screen of the kinome, revealing Interleukin-1 Receptor-Associated Kinase-2 (IRAK2) as a contributor to unfolded protein response (UPR) signaling and ER stress-induced cell death. Knocking down expression of IRAK2 (but not IRAK1) in cultured mammalian cells suppresses ER stress-induced expression of the pro-apoptotic transcription factor CHOP and activation of stress kinases. Similarly, RNAi-mediated silencing of the IRAK family member Tube (but not Pelle) suppresses activation of stress kinase signaling induced by ER stress in Drosophila cells. The action of IRAK2 maps to the IRE1 pathway, rather than the PERK or ATF6 components of the UPR. Interestingly, ER stress also induces IRAK2 gene expression in an IRE1/XBP1-dependent manner, suggesting a mutually supporting amplification loop involving IRAK2 and IRE1. In vivo, ER stress induces Irak2 expression in mice. Moreover, Irak2 gene knockout mice display defects in ER stress-induced CHOP expression and IRE1 pathway signaling. These findings demonstrate an unexpected linkage of the innate immunity machinery to UPR signaling, revealing IRAK2 as a novel amplifier of the IRE1 pathway.


Assuntos
Estresse do Retículo Endoplasmático , Quinases Associadas a Receptores de Interleucina-1/metabolismo , Transdução de Sinais , Animais , Morte Celular , Linhagem Celular , Drosophila/citologia , Estresse do Retículo Endoplasmático/genética , Regulação da Expressão Gênica , Técnicas de Silenciamento de Genes , Humanos , Quinases Associadas a Receptores de Interleucina-1/genética , Proteínas de Membrana/metabolismo , Camundongos , Camundongos Knockout , Proteínas Serina-Treonina Quinases/metabolismo , RNA Interferente Pequeno/metabolismo , Homologia de Sequência de Aminoácidos , Resposta a Proteínas não Dobradas
12.
EMBO J ; 32(4): 583-96, 2013 Feb 20.
Artigo em Inglês | MEDLINE | ID: mdl-23376919

RESUMO

Toll-like receptors transduce their signals through the adaptor molecule MyD88 and members of the IL-1R-associated kinase family (IRAK-1, 2, M and 4). IRAK-1 and IRAK-2, known to form Myddosomes with MyD88-IRAK-4, mediate TLR7-induced TAK1-dependent NFκB activation. IRAK-M was previously known to function as a negative regulator that prevents the dissociation of IRAKs from MyD88, thereby inhibiting downstream signalling. However, we now found that IRAK-M was also able to interact with MyD88-IRAK-4 to form IRAK-M Myddosome to mediate TLR7-induced MEKK3-dependent second wave NFκB activation, which is uncoupled from post-transcriptional regulation. As a result, the IRAK-M-dependent pathway only induced expression of genes that are not regulated at the post-transcriptional levels (including inhibitory molecules SOCS1, SHIP1, A20 and IκBα), exerting an overall inhibitory effect on inflammatory response. On the other hand, through interaction with IRAK-2, IRAK-M inhibited TLR7-mediated production of cytokines and chemokines at translational levels. Taken together, IRAK-M mediates TLR7-induced MEKK3-dependent second wave NFκB activation to produce inhibitory molecules as a negative feedback for the pathway, while exerting inhibitory effect on translational control of cytokines and chemokines.


Assuntos
Citocinas/biossíntese , Quinases Associadas a Receptores de Interleucina-1/metabolismo , Glicoproteínas de Membrana/metabolismo , NF-kappa B/metabolismo , Receptores de Interleucina-1/metabolismo , Transdução de Sinais/fisiologia , Receptor 7 Toll-Like/metabolismo , Animais , Linhagem Celular , Citocinas/genética , Humanos , Quinases Associadas a Receptores de Interleucina-1/genética , MAP Quinase Quinase Quinase 3/genética , MAP Quinase Quinase Quinase 3/metabolismo , Glicoproteínas de Membrana/genética , Camundongos , NF-kappa B/genética , Receptores de Interleucina-1/genética , Receptor 7 Toll-Like/genética
13.
Immunity ; 37(5): 800-12, 2012 Nov 16.
Artigo em Inglês | MEDLINE | ID: mdl-23142783

RESUMO

Interleukin-1 (IL-1)-induced activation of the mTOR kinase pathway has major influences on Th17 cell survival, proliferation, and effector function. Via biochemical and genetic approaches, the kinases IKKi and GSK3α were identified as the critical intermediate signaling components for IL-1-induced AKT activation, which in turn activated mTOR. Although insulin-induced AKT activation is known to phosphorylate and inactivate GSK3α and GSK3ß, we found that GSK3α but not GSK3ß formed a constitutive complex to phosphorylate and suppress AKT activation, showing that a reverse action from GSK to AKT can take place. Upon IL-1 stimulation, IKKi was activated to mediate GSK3α phosphorylation at S21, thereby inactivating GSK3α to promote IL-1-induced AKT-mTOR activation. Thus, IKKi has a critical role in Th17 cell maintenance and/or proliferation through the GSK-AKT-mTOR pathway, implicating the potential of IKKi as a therapeutic target.


Assuntos
Quinase 3 da Glicogênio Sintase/metabolismo , Quinase I-kappa B/metabolismo , Interleucina-1/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Serina-Treonina Quinases TOR/metabolismo , Células Th17/metabolismo , Animais , Processos de Crescimento Celular/fisiologia , Ativação Enzimática , Quinase 3 da Glicogênio Sintase/imunologia , Glicogênio Sintase Quinase 3 beta , Insulina/imunologia , Insulina/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Fosforilação , Proteínas Serina-Treonina Quinases/imunologia , Proteínas Proto-Oncogênicas c-akt/imunologia , Transdução de Sinais , Serina-Treonina Quinases TOR/imunologia , Células Th17/citologia , Células Th17/enzimologia , Células Th17/imunologia
14.
Mol Cell Biol ; 32(19): 3990-4000, 2012 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-22851693

RESUMO

Interleukin-1 (IL-1) receptor-associated kinase (IRAK1) is phosphorylated, ubiquitinated, and degraded upon IL-1 stimulation. IRAK1 can be ubiquitinated through both K48- and K63-linked polyubiquitin chains upon IL-1 stimulation. While the Pellino proteins have been shown to meditate K63-linked polyubiquitination on IRAK1, the E3 ligase for K48-linked ubiquitination of IRAK1 has not been identified. In this study, we report that the SCF (Skp1-Cullin1-F-box)-ß-TrCP complex functions as the K48-linked ubiquitination E3 ligase for IRAK1. IL-1 stimulation induced the interaction of IRAK1 with Cullin1 and ß-TrCP. Knockdown of ß-TrCP1 and ß-TrCP2 attenuated the K48-linked ubiquitination and degradation of IRAK1. Importantly, ß-TrCP deficiency abolished the translocation TAK1-TRAF6 complex from the membrane to the cytosol, resulting in a diminishment of the IL-1-induced TAK1-dependent pathway. Taken together, these results implicate a positive role of ß-TrCP-mediated IRAK1 degradation in IL-1-induced TAK1 activation.


Assuntos
Quinases Associadas a Receptores de Interleucina-1/imunologia , Interleucina-1/imunologia , MAP Quinase Quinase Quinases/imunologia , NF-kappa B/imunologia , Fator 6 Associado a Receptor de TNF/imunologia , Ubiquitina-Proteína Ligases/imunologia , Proteínas Contendo Repetições de beta-Transducina/imunologia , Linhagem Celular , Citosol , Técnicas de Silenciamento de Genes , Humanos , Quinase I-kappa B/imunologia , Quinase I-kappa B/metabolismo , Quinases Associadas a Receptores de Interleucina-1/metabolismo , MAP Quinase Quinase 4/imunologia , MAP Quinase Quinase 4/metabolismo , MAP Quinase Quinase Quinases/metabolismo , Fosforilação , Transporte Proteico , Fator 6 Associado a Receptor de TNF/metabolismo , Ubiquitina-Proteína Ligases/genética , Ubiquitina-Proteína Ligases/metabolismo , Ubiquitinação , Proteínas Contendo Repetições de beta-Transducina/genética , Proteínas Contendo Repetições de beta-Transducina/metabolismo
15.
J Biol Chem ; 287(30): 25686-95, 2012 Jul 20.
Artigo em Inglês | MEDLINE | ID: mdl-22669975

RESUMO

Interleukin 1 receptor-associated kinase 1(IRAK1), a key molecule in TLR/IL-1R-mediated signaling, is phosphorylated, ubiquitinated, and degraded upon ligand stimulation. We and others have recently identified Pellino proteins as novel RING E3 ubiquitin ligases involved in IRAK1 polyubiquitination and degradation. However, it remains unclear how each Pellino member distinctly regulates TLR/IL-1R signaling by modulating IRAK1 ubiquitination. In this study we examined the role of Pellino 2 in IL-1- and LPS-mediated signaling and gene expression by knocking down Pellino 2 in human 293-IL-1R cells and primary bone marrow macrophages. Pellino 2 (but not Pellino 1) knockdown abolished IL-1- and LPS-induced Lys-63-linked IRAK1 ubiquitination with reduced Lys-48-linked IRAK1 ubiquitination. Furthermore, Pellino 2 is required for TAK1-dependent NFκB activation. However, because of the retained TAK1-independent NFκB activation, the levels of IL-1- and LPS-induced NFκB activation were not substantially affected in Pellino 2 knockdown 293-IL-1R cells and primary macrophages, respectively. On the other hand, Pellino 2 knockdown reduced the IL-1- and LPS-induced inflammatory gene expression at late time points, which was accompanied by increased decay rates of the mRNAs of the inflammatory genes. Importantly, IL-1- and LPS-mediated JNK and ERK activation were substantially attenuated in Pellino 2 knock-down cells, implicating MAPK activation in TLR/IL-1R-induced mRNA stabilization. Taken together, this study demonstrated that Pellino 2 plays a critical role for TLR/IL-1R-mediated post-transcriptional control.


Assuntos
Interleucina-1/metabolismo , Sistema de Sinalização das MAP Quinases/fisiologia , Macrófagos/metabolismo , Proteínas Nucleares/metabolismo , Receptores de Interleucina-1/metabolismo , Receptores Toll-Like/metabolismo , Animais , Ativação Enzimática/efeitos dos fármacos , Ativação Enzimática/fisiologia , Técnicas de Silenciamento de Genes , Células HEK293 , Humanos , Interleucina-1/genética , Lipopolissacarídeos/farmacologia , MAP Quinase Quinase Quinases/genética , MAP Quinase Quinase Quinases/metabolismo , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Camundongos , NF-kappa B/genética , NF-kappa B/metabolismo , Proteínas Nucleares/genética , Estabilidade de RNA/efeitos dos fármacos , Estabilidade de RNA/fisiologia , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Receptores de Interleucina-1/genética , Receptores Toll-Like/genética
16.
J Zhejiang Univ Sci B ; 12(8): 644-51, 2011 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-21796805

RESUMO

OBJECTIVE: To study whether Tongxinluo (TXL) can induce angiogenesis in bone marrow mesenchymal stem cells (MSCs), and to investigate the underlying mechanism. METHODS: Bone marrow MSCs were obtained from male Sprague-Dawley rats. We established an angiogenesis model in vitro via matrigel experiment. MSCs were seeded on matrigel coated 24-well plates, and treated by TXL 50 and 100 mg/L. After 24 h, we observed the tube formations of MSCs in the matrigel. Cell migration ability was examined by wound scratch test and transwell assay. Expressions of vascular endothelial growth factor (VEGF), fetal liver kinase-1 (Flk-1), matrix metalloproteinase-2 (MMP-2), MMP-9, and tissue inhibitor of metalloproteinase-2 (TIMP-2) were analyzed at the protein level by Western blot. Gelatin zymography assay was applied to investigating the MSC paracrine abilities of pro-MMP-2 and activated MMP-2. RESULTS: TXL promoted MSC tube formation in matrigel. The ratio of TXL 100 mg/L treated-MSC tubular length was increased 3.04-fold compared to the control group (P<0.05). Scratch test and transwell assay showed that TXL could improve the cell migration ability of MSCs. Western blot experiments showed that TXL promoted MSC synthesis of MMP-2, but it had no influence on the expressions of MMP-9 and TIMP-2. This effect was confirmed by gelatin zymography assay, which showed that TXL increased MSC secretion of pro-MMP-2 and activated MMP-2. VEGF expression of TXL treated-MSCs was increased compared to the control group. The expression of Flk-1 was not different among the groups. CONCLUSIONS: This study demonstrates that TXL can promote the tube formation of MSCs, and the underlying mechanisms are associated with increased migration ability of MSCs and the up-regulation of MMP-2 and VEGF expressions.


Assuntos
Medicamentos de Ervas Chinesas/farmacologia , Regulação da Expressão Gênica , Células-Tronco Mesenquimais/citologia , Animais , Movimento Celular , Colágeno/química , Combinação de Medicamentos , Técnicas In Vitro , Laminina/química , Masculino , Metaloproteinase 2 da Matriz/metabolismo , Células-Tronco Mesenquimais/efeitos dos fármacos , Neovascularização Patológica , Neovascularização Fisiológica , Proteoglicanas/química , Ratos , Ratos Sprague-Dawley , Fator A de Crescimento do Endotélio Vascular/metabolismo , Cicatrização
17.
J Immunol ; 186(5): 3006-14, 2011 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-21270393

RESUMO

Bone marrow-derived plasmacytoid dendritic cells (pDCs) from IL-1R-associated kinase (IRAK)2-deficient mice produced more IFNs than did wild-type pDCs upon stimulation with the TLR9 ligand CpG. Furthermore, in CpG-stimulated IRAK2-deficient pDCs there was increased nuclear translocation of IFN regulatory factor 7, the key transcription factor for IFN gene transcription in these cells. In IRAK2-deficient macrophages, enhanced NF-κB activation and increased expression of CpG-induced genes were detected within 2 h after treatment. However, at later times, NF-κB activation was decreased and, in contrast to the results with IFN, there was less secretion of other proinflammatory cytokines (such as TNF-α) and chemokines in CpG-stimulated IRAK2-deficient pDCs and macrophages. Therefore, although IRAK2 is a negative regulator of TLR9-mediated IFN production through its modulation of the transcriptional activity of IFN regulatory factor 7, it is also a positive regulator of TLR9-mediated proinflammatory cytokine and chemokine production at some level subsequent to transcription.


Assuntos
Citocinas/biossíntese , Mediadores da Inflamação/metabolismo , Interferon Tipo I/biossíntese , Interferon beta/biossíntese , Quinases Associadas a Receptores de Interleucina-1/fisiologia , Receptor Toll-Like 9/fisiologia , Animais , Células Cultivadas , Ilhas de CpG/imunologia , Citocinas/antagonistas & inibidores , Citocinas/fisiologia , Células Dendríticas/imunologia , Células Dendríticas/metabolismo , Regulação para Baixo/imunologia , Infecções por Herpesviridae/enzimologia , Infecções por Herpesviridae/imunologia , Infecções por Herpesviridae/patologia , Mediadores da Inflamação/antagonistas & inibidores , Mediadores da Inflamação/fisiologia , Fator Regulador 7 de Interferon/antagonistas & inibidores , Fator Regulador 7 de Interferon/fisiologia , Interferon Tipo I/antagonistas & inibidores , Interferon-alfa , Interferon beta/antagonistas & inibidores , Quinases Associadas a Receptores de Interleucina-1/deficiência , Ligantes , Camundongos , Camundongos Knockout , Proteínas Recombinantes , Rhadinovirus/imunologia , Receptor Toll-Like 9/metabolismo , Infecções Tumorais por Vírus/enzimologia , Infecções Tumorais por Vírus/imunologia , Infecções Tumorais por Vírus/patologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA