Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Elife ; 102021 07 27.
Artículo en Inglés | MEDLINE | ID: mdl-34313226

RESUMEN

Upon viral RNA recognition, the RIG-I signalosome continuously generates IFNs and cytokines, leading to neutrophil recruitment and inflammation. Thus, attenuation of excessive immune and inflammatory responses is crucial to restore immune homeostasis and prevent unwarranted damage, yet few resolving mediators have been identified. In the present study, we demonstrated that RTN3 is strongly upregulated during RNA viral infection and acts as an inflammation-resolving regulator. Increased RTN3 aggregates on the endoplasmic reticulum and interacts with both TRIM25 and RIG-I, subsequently impairing K63-linked polyubiquitination and resulting in both IRF3 and NF-κB inhibition. Rtn3 overexpression in mice causes an obvious inflammation resolving phenomenon when challenged with VSV, Rtn3-overexpressing mice display significantly decreased neutrophil numbers and inflammatory cell infiltration, which is accompanied by reduced tissue edema in the liver and thinner alveolar interstitium. Taken together, our findings identify RTN3 as a conserved negative regulator of immune and inflammatory responses and provide insights into the negative feedback that maintains immune and inflammatory homeostasis.


Asunto(s)
Proteínas Portadoras/metabolismo , Proteína 58 DEAD Box/metabolismo , Proteínas de la Membrana/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Receptores Inmunológicos/metabolismo , Transducción de Señal/efectos de los fármacos , Factores de Transcripción/metabolismo , Proteínas de Motivos Tripartitos/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Animales , Antivirales/farmacología , Proteínas Portadoras/genética , Proteínas Portadoras/inmunología , Proteína 58 DEAD Box/genética , Femenino , Células HEK293 , Humanos , Inmunidad Innata , Factor 3 Regulador del Interferón/efectos de los fármacos , Proteínas de la Membrana/genética , Proteínas de la Membrana/inmunología , Ratones , Ratones Endogámicos C57BL , FN-kappa B/efectos de los fármacos , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/inmunología , Receptores Inmunológicos/genética , Factores de Transcripción/genética , Proteínas de Motivos Tripartitos/genética , Ubiquitina-Proteína Ligasas/genética , Ubiquitinación/efectos de los fármacos
2.
Toxicol In Vitro ; 62: 104698, 2020 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-31669364

RESUMEN

Both PM2.5 and respiratory viruses are part of the atmospheric constituents. Respiratory viruses are often associated with PM2.5 exposure, but the mechanism of toxicity remains to be explored. The vitro models that adequately reproduce healthy cells or diseased cells exposing to PM2.5 and infecting VSV can provide a useful tool for studying innate immune mechanisms and investigating new therapeutic focus. In the environment of PM2.5, an infection model in which VSV infected A549 cells was established, that mimics the state in which the antiviral innate immune pathways are activated after the respiratory system is infected with RNA viruses. Subsequently, the model was exposed to PM2.5 for 24 h. PM2.5 could be ingested by A549 cells and synergize with VSV to inhibit cell viability and promote apoptosis. The expression of VSV-G were more abundant after VSV-infected A549 cells were exposed to PM2.5. Furthermore, PM2.5 inhibits VSV-induced IFN-ß expression in A549 cells. ISG15, CCL-5, and CXCL-10 had the same expression tendency with IFN-ß mRNA, consistently. Interestingly, when MG132 was applied, the expression of p-IRF-3 and IFN-ß proteins reduced by PM2.5 were refreshed. Conversely, the expression of VSV-G proteins were decreased. PM2.5 could degrade p-IRF-3 proteins by ubiquitination pathway to inhibit VSV-induced IFN-ß expression in A549 cells. Therefore, replication of the VSV viruses was promoted.


Asunto(s)
Contaminantes Atmosféricos/toxicidad , Factor 3 Regulador del Interferón/metabolismo , Material Particulado/toxicidad , Ubiquitinación/efectos de los fármacos , Vesiculovirus/efectos de los fármacos , Células A549 , Apoptosis/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Humanos , Factor 3 Regulador del Interferón/efectos de los fármacos , Interferón beta/metabolismo , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Estomatitis Vesicular/prevención & control , Estomatitis Vesicular/virología
3.
J Neuroinflammation ; 16(1): 52, 2019 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-30823934

RESUMEN

BACKGROUND: Amlexanox (ALX), a TBK1 inhibitor, can modulate immune responses and has anti-inflammatory properties. To investigate its role in regulating the progression of experimental autoimmune encephalomyelitis (EAE), we studied the effect of ALX on the maturation of dendritic cells (DCs) and the responses of effector and regulatory T cells (Tregs). METHODS: In vitro, bone marrow-derived DCs (BMDCs) were cultured and treated with ALX. Their proliferation, maturation, and their stimulatory function to induce T cells responses were detected. In vivo, the development of EAE from different groups was recorded. At the peak stage of disease, HE, LFB, and electronic microscope (EM) were used to evaluate inflammation and demyelination. Maturation of splenic DC and Th1/Th17/Treg response in the CNS and peripheral were also detected. To further explore the mechanism underlying the action of ALX in DC maturation, the activation of TBK1, IRF3, and AKT was analyzed. RESULTS: Our data indicated that ALX significantly inhibited the proliferation and maturation of BMDCs, characterized by the reduced MHCII, a co-stimulatory molecule, IL12, and IL-23 expression, along with morphological alterations. Co-culture of ALX-treated BMDCs inhibited allogeneic T cell proliferation and MOG-specific T cell response. In EAE mice, ALX significantly attenuated the EAE development by decreasing inflammatory infiltration and demyelination in the spinal cords, accompanied by reduced frequency of splenic pathogenic Th1 and Th17 cells and increased Tregs. Moreover, ALX treatment decreased Th1 and Th17 cytokines, but increased Treg cytokines in the CNS and spleen. Notably, ALX treatment reduced the frequency and expression of CD80 and CD86 on splenic DCs and lowered IL-12 and IL-23 secretion, further supporting an impaired maturation of splenic DCs. In addition, ALX potently reduced the phosphorylation of IRF3 and AKT in BMDC and splenic DCs, both of which are substrates of TBK1 and associated with DC maturation. CONCLUSIONS: ALX, a TBK1 inhibitor, mitigated EAE development by inhibiting DC maturation and subsequent pathogenic Th1 and Th17 responses while increasing Treg responses through attenuating the TBK1/AKT and TBK1/IRF3 signaling.


Asunto(s)
Aminopiridinas/farmacología , Células Dendríticas/efectos de los fármacos , Encefalomielitis Autoinmune Experimental/inmunología , Linfocitos T/efectos de los fármacos , Animales , Diferenciación Celular/efectos de los fármacos , Diferenciación Celular/inmunología , Factor 3 Regulador del Interferón/efectos de los fármacos , Factor 3 Regulador del Interferón/metabolismo , Activación de Linfocitos/efectos de los fármacos , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Proteínas Serina-Treonina Quinasas/efectos de los fármacos , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/efectos de los fármacos , Proteínas Proto-Oncogénicas c-akt/metabolismo , Transducción de Señal/efectos de los fármacos , Linfocitos T/inmunología , Linfocitos T Reguladores/efectos de los fármacos , Linfocitos T Reguladores/inmunología , Células TH1/efectos de los fármacos , Células TH1/inmunología , Células Th17/efectos de los fármacos , Células Th17/inmunología
4.
Arthritis Rheumatol ; 71(1): 50-62, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-30009417

RESUMEN

OBJECTIVE: The production of class-switched high-affinity autoantibodies derived from organized germinal centers (GCs) is a hallmark of many autoimmune inflammatory diseases, including rheumatoid arthritis (RA). TANK-binding kinase 1 (TBK-1) is a serine/threonine kinase involved in the maturation of GC follicular helper T (Tfh) cells downstream of inducible costimulator signaling. We undertook this study to assess the therapeutic potential of TBK-1 inhibition using the small-molecule inhibitor WEHI-112 in antibody-dependent models of inflammatory arthritis. METHODS: Using the models of collagen-induced arthritis (CIA), antigen-induced arthritis (AIA), and K/BxN serum-transfer-induced arthritis (STIA), we determined the effectiveness of WEHI-112 at inhibiting clinical and histologic features of arthritis in C57BL/6 and DBA/1 mice. We used immunohistochemistry to characterize GC populations during CIA development, and we used enzyme-linked immunosorbent assays to determine levels of Ig autoantibodies in WEHI-112-treated mice compared to vehicle-treated mice. RESULTS: WEHI-112, a tool compound that is semiselective for TBK-1 but that also has activity against IKKε and JAK2, abolished TBK-1-dependent activation of interferon (IFN) regulatory factor 3 and inhibited type I IFN responses in vitro. In vivo, treatment with WEHI-112 selectively abrogated clinical and histologic features of established, antibody-dependent CIA, but had minimal effects on an antibody-independent model of AIA or on K/BxN STIA. In keeping with these findings, WEHI-112 reduced arthritogenic type II collagen-specific IgG1 and IgG2b antibody production. Furthermore, WEHI-112 altered the GC Tfh cell phenotype and GC B cell function in CIA. CONCLUSION: We report that TBK-1 inhibition using WEHI-112 abrogated antibody-dependent CIA. As WEHI-112 failed to inhibit non-antibody-driven joint inflammation, we conclude that the major effect of this compound was most likely the targeting of TBK-1-mediated mechanisms in the GC reaction. This approach may have therapeutic potential in RA and in other GC-associated autoantibody-driven inflammatory diseases.


Asunto(s)
Artritis Experimental/inmunología , Artritis Reumatoide/inmunología , Autoanticuerpos/efectos de los fármacos , Centro Germinal/efectos de los fármacos , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Linfocitos T Colaboradores-Inductores/efectos de los fármacos , Animales , Autoanticuerpos/inmunología , Azetidinas/farmacología , Colágeno Tipo II , Ciclobutanos/farmacología , Modelos Animales de Enfermedad , Ensayo de Inmunoadsorción Enzimática , Adyuvante de Freund , Centro Germinal/inmunología , Inmunohistoquímica , Factores Inmunológicos , Técnicas In Vitro , Factor 3 Regulador del Interferón/efectos de los fármacos , Factor 3 Regulador del Interferón/inmunología , Interferón Tipo I/efectos de los fármacos , Interferón Tipo I/inmunología , Inhibidores de las Cinasas Janus/farmacología , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos DBA , Morfolinas/farmacología , Purinas , Pirazoles , Albúmina Sérica Bovina , Sulfonamidas/farmacología , Linfocitos T Colaboradores-Inductores/inmunología
5.
Nat Commun ; 9(1): 493, 2018 02 05.
Artículo en Inglés | MEDLINE | ID: mdl-29402958

RESUMEN

The innate immune system processes pathogen-induced signals into cell fate decisions. How information is turned to decision remains unknown. By combining stochastic mathematical modelling and experimentation, we demonstrate that feedback interactions between the IRF3, NF-κB and STAT pathways lead to switch-like responses to a viral analogue, poly(I:C), in contrast to pulse-like responses to bacterial LPS. Poly(I:C) activates both IRF3 and NF-κB, a requirement for induction of IFNß expression. Autocrine IFNß initiates a JAK/STAT-mediated positive-feedback stabilising nuclear IRF3 and NF-κB in first responder cells. Paracrine IFNß, in turn, sensitises second responder cells through a JAK/STAT-mediated positive feedforward pathway that upregulates the positive-feedback components: RIG-I, PKR and OAS1A. In these sensitised cells, the 'live-or-die' decision phase following poly(I:C) exposure is shorter-they rapidly produce antiviral responses and commit to apoptosis. The interlinked positive feedback and feedforward signalling is key for coordinating cell fate decisions in cellular populations restricting pathogen spread.


Asunto(s)
Inmunidad Innata/inmunología , Factor 3 Regulador del Interferón/inmunología , Interferón beta/inmunología , Quinasas Janus/inmunología , FN-kappa B/inmunología , Factores de Transcripción STAT/inmunología , 2',5'-Oligoadenilato Sintetasa , Animales , Línea Celular , Proteína 58 DEAD Box/inmunología , Retroalimentación Fisiológica , Técnicas de Inactivación de Genes , Inmunidad Innata/efectos de los fármacos , Inductores de Interferón/farmacología , Factor 3 Regulador del Interferón/efectos de los fármacos , Ratones , FN-kappa B/efectos de los fármacos , Poli I-C/farmacología , Factor de Transcripción STAT1/genética , Transducción de Señal , Factor de Transcripción ReIA/genética , Regulación hacia Arriba , eIF-2 Quinasa/inmunología
6.
Arthritis Rheumatol ; 69(7): 1495-1501, 2017 07.
Artículo en Inglés | MEDLINE | ID: mdl-28426911

RESUMEN

OBJECTIVE: Gain-of-function mutations in TMEM173, encoding the stimulator of interferon (IFN) genes (STING) protein, underlie a novel type I interferonopathy that is minimally responsive to conventional immunosuppressive therapies and associated with high frequency of childhood morbidity and mortality. STING gain-of-function causes constitutive oversecretion of IFN. This study was undertaken to determine the effects of a TANK-binding kinase 1 (TBK-1)/IKKɛ inhibitor (BX795) on secretion and signaling of IFN in primary peripheral blood mononuclear cells (PBMCs) from patients with mutations in STING. METHODS: PBMCs from 4 patients with STING-associated disease were treated with BX795. The effect of BX795 on IFN pathways was assessed by Western blotting and an IFNß reporter assay, as well as by quantification of IFNα in cell lysates, staining for STAT-1 phosphorylation, and measurement of IFN-stimulated gene (ISG) messenger RNA (mRNA) expression. RESULTS: Treatment of PBMCs with BX795 inhibited the phosphorylation of IFN regulatory factor 3 and IFNß promoter activity induced in HEK 293T cells by cyclic GMP-AMP or by genetic activation of STING. In vitro exposure to BX795 inhibited IFNα production in PBMCs of patients with STING-associated disease without affecting cell survival. In addition, BX795 decreased STAT-1 phosphorylation and ISG mRNA expression independent of IFNα blockade. CONCLUSION: These findings demonstrate the effect of BX795 on reducing type I IFN production and IFN signaling in cells from patients with gain-of-function mutations in STING. A combined inhibition of TBK-1 and IKKɛ therefore holds potential for the treatment of patients carrying STING mutations, and may also be relevant in other type I interferonopathies.


Asunto(s)
Factor 3 Regulador del Interferón/efectos de los fármacos , Subunidad gamma del Factor 3 de Genes Estimulados por el Interferón/efectos de los fármacos , Interferón-alfa/efectos de los fármacos , Interferón beta/efectos de los fármacos , Leucocitos Mononucleares/efectos de los fármacos , Proteínas de la Membrana/efectos de los fármacos , Pirimidinas/farmacología , Tiofenos/farmacología , Western Blotting , Niño , Células HEK293 , Humanos , Quinasa I-kappa B/antagonistas & inhibidores , Técnicas In Vitro , Factor 3 Regulador del Interferón/genética , Factor 3 Regulador del Interferón/metabolismo , Factores Reguladores del Interferón/efectos de los fármacos , Factores Reguladores del Interferón/genética , Subunidad gamma del Factor 3 de Genes Estimulados por el Interferón/genética , Subunidad gamma del Factor 3 de Genes Estimulados por el Interferón/metabolismo , Interferón-alfa/inmunología , Interferón beta/inmunología , Leucocitos Mononucleares/inmunología , Proteínas de la Membrana/genética , Proteínas de la Membrana/inmunología , Mutación , Nucleótidos Cíclicos/farmacología , Fosforilación/efectos de los fármacos , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , ARN Mensajero/efectos de los fármacos , ARN Mensajero/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Factor de Transcripción STAT1/efectos de los fármacos , Factor de Transcripción STAT1/metabolismo
7.
Pharmazie ; 66(4): 293-300, 2011 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-21612158

RESUMEN

Resveratrol, a stilbene type compound identified in wine and fruit juice, has been found to exhibit various pharmacological activities such as anti-oxidative, anti-cancerous, anti-inflammatory and anti-aging effects. Although numerous papers have explored the pharmacology of resveratrol in one particular cellular action, how this compound can have multiple effects simultaneously has not been fully addressed. In this study, therefore, we explored its broad-spectrum inhibitory mechanism using lipopolysaccharide (LPS)-mediated inflammatory responses and reporter gene assays involving overexpression of toll like receptor (TLR) adaptor molecules. Co-transfection of adaptor molecules such as (1) myeloid differentiation primary response gene 88 (MyD88), (2) Toll/4ll-1 Receptor-domain-containing adapter-inducing interferon-beta (TRIF), (3) TRIF-related adaptor molecule (TRAM), or (4) TANK-binding kinase (TBK) 1 strongly enhanced luciferase activity mediated by transcription factors including nuclear factor (NF)-KB, activator protein (AP)-1, and interferon regulatory factor (IRF)-3. Of the adaptor proteins, TRIF and TBK1 but not MyD88 and IKK enhanced luciferase activity mediated by these transcription factors. Resveratrol dose-dependently suppressed LPS-induced NO production in macrophages. It also blocked the increases in levels of mRNA for IFN-1, tumor necrosis factor (TNF)-alpha, and inducible nitric oxide synthase (iNOS) that were induced by LPS. Resveratrol diminished the translocation or activation of IRF-3 at 90min, c-Jun, a subunit of AP-1, and STAT-1 at 120 min, and p50, a subunit of NF-KB, at 60 and 90 min. Resveratrol strongly suppressed the up-regulation of luciferase activity induced by these adaptor molecules with IC50 values of 5 to 65 microM. In particular, higher inhibitory effects of resveratrol were when TRIF or TBK1 were overexpressed following cotransfection of luciferase constructs with IRF-3 binding sequences. Taken together, our data suggest that the suppression of TRIF and TBK1, which mediates transcriptional activation of NF-kappaB, AP-1, and IRF-3, contributes to resveratrol's broad-spectrum inhibitory activity, and that this compound can be further developed as a lead anti-inflammatory compound.


Asunto(s)
Proteínas Adaptadoras del Transporte Vesicular/efectos de los fármacos , Proteínas Adaptadoras del Transporte Vesicular/fisiología , Antiinflamatorios no Esteroideos/farmacología , Factor 3 Regulador del Interferón/efectos de los fármacos , Factor 3 Regulador del Interferón/fisiología , Proteínas Serina-Treonina Quinasas/efectos de los fármacos , Proteínas Serina-Treonina Quinasas/fisiología , Estilbenos/farmacología , Animales , Western Blotting , Núcleo Celular/efectos de los fármacos , Células Cultivadas , Colorantes , Genes Reporteros/efectos de los fármacos , Inflamación/inducido químicamente , Inflamación/prevención & control , Lipopolisacáridos/farmacología , Macrófagos Peritoneales/efectos de los fármacos , Masculino , Ratones , Ratones Endogámicos C57BL , Óxido Nítrico/metabolismo , Plásmidos/efectos de los fármacos , Plásmidos/genética , ARN Mensajero/biosíntesis , ARN Mensajero/genética , Resveratrol , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal/efectos de los fármacos , Sales de Tetrazolio , Tiazoles
8.
Mol Immunol ; 48(1-3): 294-304, 2010.
Artículo en Inglés | MEDLINE | ID: mdl-20728939

RESUMEN

Recent studies implicate the mammalian target of rapamycin (mTOR) pathway in the control of inflammatory responses following Toll-like receptor (TLR) stimulation in myeloid cells but its role in non-myeloid cells such as human keratinocytes is unknown. Here we show that TLR3 signaling can induce robust cytokine secretion including interleukin 1 beta (IL-1ß), tumor necrosis factor alpha (TNFα), IL-12p70 and interferon beta (IFN-ß), and our data reveal for the first time that inhibiting mTOR with rapamycin, suppresses these TLR3 induced responses but actually enhances bioactive IL-12p70 production in human oral keratinocytes. Rapamycin inhibited the phosphorylation of the 70-kDa ribosomal protein S6 kinase (p70S6K) and the 4E binding protein 1 (4EBP-1), and suppressed the mitogen activated protein kinase (MAPK) pathway by decreasing phosphorylation of c-Jun N-terminal kinase (JNK). We also show that TLR3 induces interferon regulatory factor 3 (IRF3) activation by Akt via an mTOR-p70S6K-4EBP1 pathway. Furthermore, we provide evidence that Poly I:C induced expression of IL-1ß, TNFα, IL-12p70 and IFN-ß was blocked by JNK inhibitor SP600125. TLR3 preferentially phosphorylated IKKα through mTOR to activate nuclear factor kappa beta (NF-κB) in human oral keratinocytes. Taken together, these data demonstrate p70S6K, p4EBP1, JNK, NF-κB and IRF3 are involved in the regulation of inflammatory mediators by TLR3 via the mTOR pathway. mTOR is a novel pathway modulating TLR3 induced inflammatory and antiviral responses in human oral keratinocytes.


Asunto(s)
Queratinocitos/inmunología , Mucosa Bucal/inmunología , Transducción de Señal/inmunología , Serina-Treonina Quinasas TOR/inmunología , Receptor Toll-Like 3/inmunología , Western Blotting , Citocinas/biosíntesis , Citocinas/efectos de los fármacos , Citocinas/inmunología , Humanos , Inmunosupresores/farmacología , Factor 3 Regulador del Interferón/efectos de los fármacos , Factor 3 Regulador del Interferón/inmunología , Factor 3 Regulador del Interferón/metabolismo , Proteínas Quinasas JNK Activadas por Mitógenos/efectos de los fármacos , Proteínas Quinasas JNK Activadas por Mitógenos/inmunología , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Queratinocitos/efectos de los fármacos , Queratinocitos/metabolismo , Proteínas Quinasas Activadas por Mitógenos/efectos de los fármacos , Proteínas Quinasas Activadas por Mitógenos/inmunología , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Mucosa Bucal/efectos de los fármacos , Mucosa Bucal/metabolismo , Poli I-C/farmacología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal/efectos de los fármacos , Sirolimus/farmacología , Serina-Treonina Quinasas TOR/efectos de los fármacos , Serina-Treonina Quinasas TOR/metabolismo , Receptor Toll-Like 3/efectos de los fármacos , Receptor Toll-Like 3/metabolismo
9.
J Leukoc Biol ; 86(2): 411-21, 2009 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-19406830

RESUMEN

M-CSF and GM-CSF are mediators involved in regulating the numbers and function of macrophage lineage populations and have been shown to contribute to macrophage heterogeneity. Type I IFN is an important mediator produced by macrophages and can have profound regulatory effects on their properties. In this study, we compared bone marrow-derived macrophages (BMM) and GM-CSF-induced BMM (GM-BMM) from wild-type and IFNAR1(-/-) mice to assess the contribution of endogenous type I IFN to the phenotypic differences between BMM and GM-BMM. BMM were capable of higher constitutive IFN-beta production, which contributed significantly to their basal transcriptome. Microarray analysis found that of the endogenous type I IFN-regulated genes specific to either BMM or GM-BMM, 488 of these gene alterations were unique to BMM, while only 50 were unique to GM-BMM. Moreover, BMM displayed enhanced basal mRNA levels, relative to GM-BMM, of a number of genes identified as being dependent on type I IFN signaling, including Stat1, Stat2, Irf7, Ccl5, Ccl12, and Cxcl10. As a result of prior type I IFN "priming," upon LPS stimulation BMM displayed increased activation of the MyD88-independent IRF-3/STAT1 pathways compared with GM-BMM, which correlated with the distinct cytokine/chemokine profiles of the two macrophage subsets. Furthermore, the autocrine type I IFN signaling loop regulated the production of the M1 and M2 signature cytokines, IL-12p70 and IL-10. Collectively, these findings demonstrate that constitutive and LPS-induced type I IFN play significant roles in regulating the differences in phenotype and function between BMM and GM-BMM.


Asunto(s)
Factor Estimulante de Colonias de Granulocitos y Macrófagos/farmacología , Interferón Tipo I/efectos de los fármacos , Factor Estimulante de Colonias de Macrófagos/farmacología , Macrófagos/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Animales , Comunicación Autocrina/efectos de los fármacos , Comunicación Autocrina/inmunología , Linaje de la Célula , Células Cultivadas , Citocinas/metabolismo , Femenino , Regulación de la Expresión Génica/efectos de los fármacos , Regulación de la Expresión Génica/inmunología , Factor 3 Regulador del Interferón/efectos de los fármacos , Factor 3 Regulador del Interferón/metabolismo , Interferón Tipo I/metabolismo , Interferón beta/metabolismo , Macrófagos/inmunología , Macrófagos/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Análisis de Secuencia por Matrices de Oligonucleótidos , Fenotipo , ARN Mensajero/efectos de los fármacos , ARN Mensajero/metabolismo , Receptor de Interferón alfa y beta/genética , Receptores de Quimiocina/efectos de los fármacos , Receptores de Quimiocina/metabolismo , Transducción de Señal/inmunología , Factores de Transcripción/efectos de los fármacos , Factores de Transcripción/metabolismo
10.
Arterioscler Thromb Vasc Biol ; 28(5): 871-7, 2008 May.
Artículo en Inglés | MEDLINE | ID: mdl-18323514

RESUMEN

OBJECTIVE: Macrophage-mediated chronic inflammation of adipose tissue is causally linked to insulin resistance in obesity. The beneficial effects of 3-hydroxy-3-methylglutaryl (HMG) coenzyme A (CoA) reductase inhibitors (statins) on glucose metabolism have been suggested, but the effects of these agents on adipose tissue inflammation are unclear. The aim of the present study is to define the effects of statins on adipose tissue inflammation and macrophages. METHODS AND RESULTS: Pravastatin or pitavastatin treatment of obese mice attenuated an increase in mRNA expressions of proinflammatory genes, including MCP1 and IL6, in adipose tissue. The supernatant of TLR4-stimulated RAW264 macrophages strongly induced the expression of these genes in 3T3-L1 adipocytes, which was inhibited by pretreatment of macrophages with either statin. Statins inhibited TLR4-mediated activation of interferon (IFN) regulatory factor (IRF)3 by either lipopolysaccharide (LPS) or palmitic acid, resulting in suppression of IFN-beta expression, but not that of NF-kappaB or JNK. Moreover, statins strongly downregulated TLR3-mediated gene expressions by poly(I:C), but not TLR2-stimulation by zymosan A. Neutralization of IFN-beta attenuated proinflammatory activities of the macrophage supernatant. CONCLUSIONS: Statins partially attenuated the development of adipose tissue inflammation in obese mice, which might be associated with an inhibitory effect of statins on TLR4-triggered expression of IFN-beta via MyD88-independent signaling pathway in macrophages.


Asunto(s)
Tejido Adiposo/efectos de los fármacos , Inhibidores de Hidroximetilglutaril-CoA Reductasas/farmacología , Inflamación/prevención & control , Factor 3 Regulador del Interferón/metabolismo , Interferón beta/metabolismo , Macrófagos/metabolismo , Factor 88 de Diferenciación Mieloide/metabolismo , Células 3T3-L1 , Tejido Adiposo/metabolismo , Tejido Adiposo/patología , Animales , Femenino , Inflamación/metabolismo , Inflamación/patología , Factor 3 Regulador del Interferón/efectos de los fármacos , Interferón beta/efectos de los fármacos , Interleucina-6/metabolismo , Macrófagos/efectos de los fármacos , Macrófagos/patología , Masculino , Ratones , Ratones Obesos , Factor 88 de Diferenciación Mieloide/efectos de los fármacos , Obesidad/metabolismo , Obesidad/patología , Pravastatina/farmacología , Quinolinas/farmacología , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología , Receptor Toll-Like 4/metabolismo
11.
J Invest Dermatol ; 126(7): 1574-81, 2006 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-16628196

RESUMEN

Toll-like receptor (TLR)3 is a receptor for virus-associated double-stranded RNA, and triggers antiviral immune responses during viral infection. Epidermal keratinocytes express TLR3 and provide an innate immune defense against viral infection. Since the intracellular regulatory mechanism is unknown, we hypothesized that the signal transducers and activators of transcription (STAT)-suppressors of cytokine signaling (SOCS) system regulates the innate immune response of keratinocytes. Treatment with polyinosinic-polycytidylic acid (poly(I:C)) resulted in the rapid translocation of IFN regulatory factor (IRF)-3 into the nucleus, followed by phosphorylation of STAT1 and STAT3. The activation of STATs by poly(I:C) probably occurs in an indirect fashion, through poly(I:C)-induced IFN. We infected cells with the dominant-negative forms of STAT1 (STAT1F), STAT3 (STAT3F), and SOCS1 using adenovirus vectors. Infection with STAT1F suppressed the induction of macrophage inflammatory protein (MIP)-1alpha by poly(I:C), whereas STAT3F had a minimal effect, which indicates that STAT1 mediates MIP-1alpha induction. SOCS1, which is a negative feedback regulator of STAT1 signaling, was induced by treatment with poly(I:C). SOCS1 infection inhibited the phosphorylation of STAT1 and significantly reduced poly(I:C)-induced MIP-1alpha production. Furthermore, STAT1-SOCS1 regulated poly(I:C)-induced TLR3 and IRF-7 expression. However, SOCS1 did not affect NF-kappaB signaling. Thus, the STAT1-SOCS1 pathway regulates the innate immune response via TLR3 signaling in epidermal keratinocytes.


Asunto(s)
Retroalimentación Fisiológica/fisiología , Inmunidad Innata/efectos de los fármacos , Péptidos y Proteínas de Señalización Intracelular/fisiología , Queratinocitos/inmunología , ARN Bicatenario/farmacología , Proteínas Represoras/fisiología , Factor de Transcripción STAT1/fisiología , Transducción de Señal/fisiología , Proteínas Supresoras de la Señalización de Citocinas/fisiología , Adenovirus Humanos/genética , Células Cultivadas , Quimiocina CCL3 , Quimiocina CCL4 , Regulación de la Expresión Génica/efectos de los fármacos , Vectores Genéticos/genética , Homeostasis/fisiología , Humanos , Inmunidad Innata/inmunología , Factor 3 Regulador del Interferón/análisis , Factor 3 Regulador del Interferón/efectos de los fármacos , Factor 3 Regulador del Interferón/fisiología , Factor 7 Regulador del Interferón/análisis , Factor 7 Regulador del Interferón/genética , Factor 7 Regulador del Interferón/fisiología , Péptidos y Proteínas de Señalización Intracelular/análisis , Péptidos y Proteínas de Señalización Intracelular/genética , Queratinocitos/química , Queratinocitos/fisiología , Proteínas Inflamatorias de Macrófagos/metabolismo , FN-kappa B/fisiología , Fosforilación/efectos de los fármacos , Poli I-C/farmacología , Proteínas Represoras/análisis , Proteínas Represoras/genética , Factor de Transcripción STAT1/análisis , Factor de Transcripción STAT1/efectos de los fármacos , Factor de Transcripción STAT1/genética , Factor de Transcripción STAT3/análisis , Factor de Transcripción STAT3/efectos de los fármacos , Factor de Transcripción STAT3/genética , Factor de Transcripción STAT3/fisiología , Transducción de Señal/efectos de los fármacos , Proteína 1 Supresora de la Señalización de Citocinas , Proteínas Supresoras de la Señalización de Citocinas/análisis , Proteínas Supresoras de la Señalización de Citocinas/genética , Receptor Toll-Like 3/análisis , Receptor Toll-Like 3/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...