Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 27
Filtrar
1.
J Cell Mol Med ; 24(21): 12716-12725, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-32977368

RESUMEN

The role of corticosteroids in acute lung injury (ALI) remains uncertain. This study aims to determine the underlying mechanisms of corticosteroid treatment for lipopolysaccharide (LPS)-induced inflammation and ALI. We used corticosteroid treatment for LPS-induced murine ALI model to investigate the effect of corticosteroid on ALI in vivo. Moreover, LPS-stimulated macrophages were used to explore the specific anti-inflammatory effects of corticosteroids on NLRP3-inflammasome in vitro. We found corticosteroids attenuated LPS-induced ALI, which manifested in reduction of the alveolar structure destruction, the infiltration of neutrophils and the inflammatory cytokines release of interleukin-1ß (IL-1ß) and interleukin-18 (IL-18) in Lung. In vitro, when NLRP3-inflammasome was knocked out, inflammatory response of caspase-1 activation and IL-1ß secretion was obviously declined. Further exploration, our results showed that when corticosteroid preprocessed macrophages before LPS primed, it obviously inhibited the activation of caspase-1 and the maturation of IL-1ß, which depended on inhibiting the nuclear factor-κB (NF-κB) signal pathway activation. However, when corticosteroids intervened the LPS-primed macrophages, it also negatively regulated NLRP3-inflammasome activation through suppressing mitochondrial reactive oxygen species (mtROS) production. Our results revealed that corticosteroids played a protection role in LPS-induced inflammation and ALI by suppressing both NF-κB signal pathway and mtROS-dependent NLRP3 inflammasome activation.


Asunto(s)
Corticoesteroides/uso terapéutico , Inflamasomas/antagonistas & inhibidores , Inflamación/tratamiento farmacológico , Inflamación/metabolismo , Proteína con Dominio Pirina 3 de la Familia NLR/antagonistas & inhibidores , Lesión Pulmonar Aguda , Corticoesteroides/farmacología , Animales , Caspasa 1/metabolismo , Dexametasona/farmacología , Dexametasona/uso terapéutico , Activación Enzimática/efectos de los fármacos , Inflamasomas/metabolismo , Inflamación/inducido químicamente , Interleucina-18/metabolismo , Lipopolisacáridos , Ratones Endogámicos C57BL , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , Modelos Biológicos , FN-kappa B/metabolismo , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Transducción de Señal
2.
Nat Immunol ; 9(8): 898-907, 2008 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-18604210

RESUMEN

The inhibitory signaling of natural killer (NK) cells is crucial in the regulation of innate immune responses. Here we show that the association of KIR2DL1, an inhibitory receptor of NK cells, with beta-arrestin 2 mediated recruitment of the tyrosine phosphatases SHP-1 and SHP-2 to KIR2DL1 and facilitated 'downstream' inhibitory signaling. Consequently, the cytotoxicity of NK cells was higher in beta-arrestin 2-deficient mice but was inhibited in beta-arrestin 2-transgenic mice. Moreover, beta-arrestin 2-deficient mice were less susceptible than wild-type mice to mouse cytomegalovirus infection, an effect that was abolished by depletion of NK cells. Our findings identify a previously unknown mechanism by which the inhibitory signaling in NK cells is regulated.


Asunto(s)
Arrestinas/farmacología , Células Asesinas Naturales/efectos de los fármacos , Células Asesinas Naturales/inmunología , Receptores Inmunológicos/inmunología , Transducción de Señal/inmunología , Animales , Células Cultivadas , Péptidos y Proteínas de Señalización Intracelular , Ratones , Receptores Inmunológicos/química , Receptores Inmunológicos/genética , Receptores Inmunológicos/metabolismo , Arrestina beta 2 , beta-Arrestinas
3.
Am J Physiol Lung Cell Mol Physiol ; 313(4): L677-L686, 2017 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-28684545

RESUMEN

Macrolides antibiotics have been effectively used in many chronic diseases, especially with Pseudomonas aeruginosa (P. aeruginosa) infection. The mechanisms underlying the therapeutic effects of macrolides in these diseases remain poorly understood. We established a mouse model of chronic lung infection using P. aeruginosa agar-beads, with azithromycin treatment or placebo. Lung injury, bacterial clearance, and inflammasome-related proteins were measured. In vitro, the inflammasomes activation induced by flagellin or ATP were assessed in LPS-primed macrophages with or without macrolides treatment. Plasma IL-18 levels were determined from patients who were diagnosed with bronchiectasis isolated with or without P. aeruginosa and treated with azithromycin for 3-5 days. Azithromycin treatment enhanced bacterial clearance and attenuated lung injury in mice chronically infected with P. aeruginosa, which resulted from the inhibition of caspase-1-dependent IL-1ß and IL-18 secretion. In vitro, azithromycin and erythromycin inhibited NLRC4 and NLRP3 inflammasomes activation. Plasma IL-18 levels were higher in bronchiectasis patients with P. aeruginosa isolation compared with healthy controls. Azithromycin administration markedly decreased IL-18 secretion in bronchiectasis patients. The results of this study reveal that azithromycin and erythromycin exert a novel anti-inflammatory effect by attenuating inflammasomes activation, which suggests potential treatment options for inflammasome-related diseases.


Asunto(s)
Bronquiectasia/tratamiento farmacológico , Inflamasomas/antagonistas & inhibidores , Macrólidos/farmacología , Infecciones por Pseudomonas/prevención & control , Pseudomonas aeruginosa/efectos de los fármacos , Animales , Antibacterianos/farmacología , Azitromicina/farmacología , Bronquiectasia/microbiología , Células Cultivadas , Humanos , Inflamasomas/efectos de los fármacos , Macrófagos/citología , Macrófagos/efectos de los fármacos , Ratones , Ratones Endogámicos C57BL , Infecciones por Pseudomonas/microbiología
4.
PLoS Pathog ; 9(8): e1003545, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23990780

RESUMEN

Nuclear hormone receptors respond to small molecules such as retinoids or steroids and regulate development. Signaling in the conserved p38/PMK-1 MAP kinase pathway regulates innate immunity. In this study, we show that the Caenorhabditis elegans nuclear receptor DAF-12 negatively regulates the defense against pathogens via the downstream let-7 family of microRNAs, which directly target SKN-1, a gene downstream of PMK-1. These findings identify nuclear hormone receptors as components of innate immunity that crosstalk with the p38/PMK-1 MAP kinase pathway.


Asunto(s)
Proteínas de Caenorhabditis elegans/inmunología , Caenorhabditis elegans/inmunología , Inmunidad Innata/fisiología , Sistema de Señalización de MAP Quinasas/inmunología , MicroARNs/inmunología , Receptores Citoplasmáticos y Nucleares/inmunología , Animales , Caenorhabditis elegans/genética , Proteínas de Caenorhabditis elegans/genética , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/inmunología , Sistema de Señalización de MAP Quinasas/genética , MicroARNs/genética , Proteínas Quinasas Activadas por Mitógenos/genética , Proteínas Quinasas Activadas por Mitógenos/inmunología , Receptores Citoplasmáticos y Nucleares/genética , Factores de Transcripción/genética , Factores de Transcripción/inmunología
5.
Exp Mol Pathol ; 97(3): 590-9, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25446845

RESUMEN

The effect of IL-10 on macrophage migration was investigated, including the analysis of protein expression, cytokine secretion and activation of the MAPKs and NF-kB pathway. The expression of endogenic IL-10 was down-regulated in macrophage stimulated with oxLDL for indicated time. Exogenous IL-10 reversed oxLDL-inhibited chemotactic macrophage numbers by 48.18 ± 4.93% (3h), 64.8 ± 5.61% (6h) and 63.66 ± 3.05% (12h), and pretreated with SB203580 (P38 signaling inhibitor) in macrophage, oxLDL could not inhibit macrophage emigration. IL-10 significantly decreased oxLDL-mediated increase of SR-A expression, and pretreatment with ß-arrestin2 RNAi in macrophage could not change oxLDL-induced SR-A expression. IL-10 also significantly decreased oxLDL-mediated increase of ß-arrestin2 expression, and pre-treated with SR-A RNAi in macrophage, oxLDL could not induce the increase of ß-arrestin2 expression. However, IL-10 significantly reversed oxLDL-mediated inhibition of CCR7 expression about 95.7 8 ± 0.99% (mRNA) and 80.06 ± 19.46% (protein), and pretreated with P38 inhibitor SB203580 in macrophage, oxLDL could not decrease CCR7 expression. IL-10 inhibited oxLDL-mediated inhibition of MMP9 secretion about 74.02 ± 22.35%, and pretreated with P38 inhibitor SB203580 in macrophage, oxLDL could not decrease MMP9 secretion. Treatment with oxLDL increased P38-phosphorylation by 31.88 ± 2.79%, 40.24 ± 5.69% and 30.93 ± 4.66% at 15, 30 and 60 min, respectively, whereas the effect of IL-10 on the expression of phosphorylated P38 was reversed by 49.49 ± 12.12%, 70.93 ± 16.14% and 47.62 ± 6.00% in up-indicated time-points, respectively. From these data, we speculated oxLDL-SR-A-ß-arrestin2-P38-MMP9/CCR7 could play a critical role in the macrophages migration, which was blocked by IL-10 through inhibiting oxLDL uptake.


Asunto(s)
Movimiento Celular/fisiología , Interleucina-10/metabolismo , Lipoproteínas LDL/metabolismo , Sistema de Señalización de MAP Quinasas/fisiología , Macrófagos/metabolismo , Animales , Western Blotting , Ratones , Ratones Endogámicos C57BL , Reacción en Cadena en Tiempo Real de la Polimerasa
6.
J Immunol ; 185(12): 7435-42, 2010 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-21068409

RESUMEN

MAPK phosphatase-1 (MKP-1) is an archetypical member of the dual-specificity phosphatase family that deactivates MAPKs. Induction of MKP-1 has been implicated in attenuating the LPS- or peptidoglycan-induced biosynthesis of proinflammatory cytokines, but the role of noncoding RNA in the expression of the MKP-1 is still poorly understood. In this study, we show that MKP-1 is a direct target of microRNA-101 (miR-101). Transfection of miR-101 attenuates induction of MKP-1 by LPS as well as prolonged activation of p38 and JNK/stress-activated protein kinase, whereas inhibition of miR-101 enhances the expression of MKP-1 and shortens p38 and JNK activation. We also found that expression of miR-101 is induced by multiple TLR ligands, including LPS, peptidoglycan, or polyinosinic-polycytidylic acid, and that inhibition of PI3K/Akt by LY294002 or Akt RNA interference blocks the induction of miR-101 by LPS in RAW264.7 macrophage cells. Moreover, treatment of cells with dexamethasone, a widely used anti-inflammatory agent, markedly inhibits miR-101 expression and enhances the expression of MKP-1 in LPS-stimulated macrophages. Together, these results indicate that miR-101 regulates the innate immune responses of macrophages to LPS through targeting MKP-1.


Asunto(s)
Fosfatasa 1 de Especificidad Dual/inmunología , Inmunidad Innata/fisiología , Macrófagos/inmunología , MicroARNs/inmunología , Animales , Antiinflamatorios/farmacología , Línea Celular , Dexametasona/farmacología , Fosfatasa 1 de Especificidad Dual/biosíntesis , Activación Enzimática/efectos de los fármacos , Activación Enzimática/inmunología , Regulación Enzimológica de la Expresión Génica/efectos de los fármacos , Inmunidad Innata/efectos de los fármacos , Lipopolisacáridos/farmacología , MAP Quinasa Quinasa 4/inmunología , MAP Quinasa Quinasa 4/metabolismo , Macrófagos/metabolismo , Ratones , MicroARNs/biosíntesis , Receptores Toll-Like/agonistas , Receptores Toll-Like/inmunología , Receptores Toll-Like/metabolismo , Proteínas Quinasas p38 Activadas por Mitógenos/inmunología , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
7.
Microbiol Spectr ; 10(5): e0155022, 2022 10 26.
Artículo en Inglés | MEDLINE | ID: mdl-36190409

RESUMEN

Pseudomonas aeruginosa (PA) is known as one kind of extracellular pathogens. However, more evidence showed that PA encounters the intracellular environment in different mammalian cell types. Little is known of innate immune factors modulating intracellular PA survival. In the present study, we proposed that interferon-ß (IFN-ß) is beneficial to the survival of PA in the cytoplasm of macrophages. Furthermore, we found that interleukin-1ß (IL-1ß) induced by PA suppresses IFN-ß response driven by the cGAS-STING-TBK1 pathway. Mechanistically, IL-1ß decreased the production of cyclic GMP-AMP (cGAMP) by activating AKT kinase. cGAMP is necessarily sufficient to stimulate the transcription of IFN-ß via the STING adaptor-TBK1 kinase-IRF3 transcription factor axis. Thus, our findings uncovered a novel module for PA intracellular survival involving IFN-ß production restricted by IL-1ß and provided a strong rationale for a potential clinical strategy against pulmonary PA infection patients. IMPORTANCE The link between innate immunity and intracellular Pseudomonas aeruginosa is unclear. Our studies illuminated the role of interferon-ß (IFN-ß) in remote intracellular PA infection. Furthermore, our experimental evidence also indicated that IL-1ß is a negative regulator of IFN-ß production and, in particular, P. aeruginosa infection. The inhibition of IFN-ß may be used as a potential therapeutic method against pulmonary PA infection.


Asunto(s)
Proteínas Proto-Oncogénicas c-akt , Pseudomonas aeruginosa , Animales , Humanos , Pseudomonas aeruginosa/metabolismo , Interleucina-1beta/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Transducción de Señal , Proteínas de la Membrana/metabolismo , Nucleotidiltransferasas/metabolismo , Factor 3 Regulador del Interferón/metabolismo , Interferón beta/metabolismo , Inmunidad Innata , Mamíferos/metabolismo
8.
Emerg Microbes Infect ; 11(1): 2132-2146, 2022 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-35930458

RESUMEN

Airway microenvironment played an important role in the progression of chronic respiratory disease. Here we showed that standardized pondus hydrogenii (pH) of exhaled breath condensate (EBC) of bronchiectasis patients was significantly lower than that of controls and was significantly correlated with bronchiectasis severity index (BSI) scores and disease prognosis. EBC pH was lower in severe patients than that in mild and moderate patients. Besides, acidic microenvironment deteriorated Pseudomonas aeruginosa (P. aeruginosa) pulmonary infection in mice models. Mechanistically, acidic microenvironment increased P. aeruginosa outer membrane vesicles (PA_OMVs) released and boosted it induced the activation of interferon regulatory factor3 (IRF3)-interferonß (IFN-ß) signalling pathway, ultimately compromised the anti-bacteria immunity. Targeted knockout of IRF3 or type 1 interferon receptor (IFNAR1) alleviated lung damage and lethality of mice after P. aeruginosa infection that aggravated by acidic microenvironment. Together, these findings identified airway acidification impaired host resistance to P. aeruginosa infection by enhancing it induced the activation of IRF3-IFN-ß signalling pathway. Standardized EBC pH may be a useful biomarker of disease severity and a potential therapeutic target for the refractory P. aeruginosa infection. The study also provided one more reference parameter for drug selection and new drug discovery for bronchiectasis.


Asunto(s)
Bronquiectasia , Interferón Tipo I , Infecciones por Pseudomonas , Animales , Concentración de Iones de Hidrógeno , Interferón beta/genética , Ratones , Pseudomonas aeruginosa/genética
9.
Proc Natl Acad Sci U S A ; 105(30): 10553-8, 2008 Jul 29.
Artículo en Inglés | MEDLINE | ID: mdl-18650396

RESUMEN

RIG-I has been implicated in innate immunity by sensing intracellular viral RNAs and inducing type I IFN production. However, we have found a significant RIG-I induction in a biological setting without active viral infection-namely, during RA-induced terminal granulocytic differentiation of acute myeloid leukemias. Here, we present evidence that a significant Rig-I induction also occurs during normal myelopoiesis and that the disruption of the Rig-I gene in mice leads to the development of a progressive myeloproliferative disorder. The initiation of progressive myeloproliferative disorder is mainly due to an intrinsic defect of Rig-I(-/-) myeloid cells, which are characterized by a reduced expression of IFN consensus sequence binding protein, a major regulator of myeloid differentiation. Thus, our study reveals a critical regulatory role of Rig-I in modulating the generation and differentiation of granulocytes.


Asunto(s)
ARN Helicasas DEAD-box/fisiología , Regulación de la Expresión Génica , Granulocitos/citología , Receptores de Ácido Retinoico/fisiología , Animales , Diferenciación Celular , Proliferación Celular , Proteína 58 DEAD Box , Exones , Granulocitos/metabolismo , Células Madre Hematopoyéticas/citología , Humanos , Inmunidad Innata , Ratones , Ratones Noqueados , Ratones Transgénicos , Células Mieloides/citología , Trastornos Mieloproliferativos/metabolismo
10.
ACS Biomater Sci Eng ; 7(5): 1817-1826, 2021 05 10.
Artículo en Inglés | MEDLINE | ID: mdl-33966375

RESUMEN

Pseudomonas aeruginosa (PA) has emerged as a pressing challenge to pulmonary infection and lung damage. The LL37 peptide is an efficient antimicrobial agent against PA strains, but its application is limited because of fast clearance in vivo, biosafety concerns, and low bioavailability. Thus, an albumin-based nanodrug delivery system with reduction sensitivity was developed by forming intermolecular disulfide bonds to increase in vivo LL37 performance against PA. Cationic LL37 can be efficiently encapsulated via electrostatic interactions to exert improved antimicrobial effects. The LL37 peptide exhibits greater than 48 h of sustained released from LL37 peptide nanoparticles (LL37 PNP), and prolonged antimicrobial effects were noted as the incubation time increased. Levels of inflammatory cytokines secreted by peritoneal macrophages, including TNF-α and IL-6, were reduced significantly after LL37 PNP treatment following PA stimulation, indicating that LL37 PNP inhibits PA growth and exerts anti-inflammatory effects in vitro. In a murine model of acute PA lung infection, LL37 PNP significantly reduced TNF-α and IL-1ß expression and alleviated lung damage. The accelerated clearance of PA indicates that LL37 PNP could improve PA lung infection and the subsequent inflammation response more efficiently compared with free LL37 peptide. In conclusion, this excellent biocompatible LL37 delivery strategy may serve as an alternative approach for the application of new types of clinical treatment in future.


Asunto(s)
Nanopartículas , Pseudomonas aeruginosa , Albúminas , Animales , Péptidos Catiónicos Antimicrobianos , Preparaciones de Acción Retardada , Pulmón , Ratones
11.
Zhejiang Da Xue Xue Bao Yi Xue Ban ; 39(5): 464-9, 2010 09.
Artículo en Zh | MEDLINE | ID: mdl-20936719

RESUMEN

OBJECTIVE: To investigate the effect of isoflurane, sevoflurane and desflurane on the expression of ICAM-1 and VCAM-1 in LPS-induced rat lung microvascular endothelial cells (RLMVECs). METHODS: Cultured LPS-treated RLMVECs were exposed to 0.7, 1.0 or 2.0 minimum alveolar concentration (MAC) iosoflurane, sevoflurane or desflurane in 6 h. The protein expression of ICAM-1 and VCAM-1 was determined by Western blot analysis. The expression of ICAM-1 mRNA was detected by reverse-transcription polymerase chain reaction (RT-PCR). RESULT: Isoflurane at concentration of 1.0 MAC up-regulated the expression of ICAM-1 in LPS-induced RLMVECs (P <0.05); while same concentrations of sevoflurane and desflurane down-regulated the expression of ICAM-1 (P<0.05). Desflurane at concentration of 2.0 MAC up-regulated the expression of ICAM-1 in non-LPS-induced RLMVECs. All the volatile anesthetics down-regulated the expression of VCAM-1 in a dose-dependent manner. CONCLUSION: Compared to isoflurane, 1.0 MAC sevoflurane and desflurane down-regulate the expression of ICAM-1, which may be the molecule mechanism of their protective effect in acute lung injury.


Asunto(s)
Anestésicos por Inhalación/farmacología , Células Endoteliales/metabolismo , Endotelio Vascular/citología , Molécula 1 de Adhesión Intercelular/metabolismo , Molécula 1 de Adhesión Celular Vascular/metabolismo , Anestésicos por Inhalación/administración & dosificación , Animales , Células Cultivadas , Desflurano , Células Endoteliales/efectos de los fármacos , Isoflurano/administración & dosificación , Isoflurano/análogos & derivados , Isoflurano/farmacología , Lipopolisacáridos/farmacología , Pulmón/irrigación sanguínea , Éteres Metílicos/administración & dosificación , Éteres Metílicos/farmacología , Ratas , Ratas Sprague-Dawley , Sevoflurano
12.
Mol Immunol ; 125: 178-186, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32717666

RESUMEN

PM2.5, a major component of air pollutants, has caused severe health problems. It has been reported that PM2.5 index is closely associated with severity of influenza A virus (IAV) infection. However, the underlying mechanisms have not been addressed. NLRP3 inflammasome and type I interferon signaling regulate host defense against influenza infection. The present study investigated the potential effects of air pollutants on host defense against influenza infection in vitro and in vivo. In this study, different concentrations of PM2.5 were pre-exposed to macrophages and mice before IAV infection to assess the negative effects of air pollutants in virus infection. We found that exposure to PM2.5 deteriorated influenza virus infection via compromising innate immune responses manifested by a decrease IL-1ß and IFN-ß production in vitro. Meanwhile, mice exposed with PM2.5 were susceptible to PR8 virus infection due to down-regulation of IL-1ß and IFN-ß. Mechanistically, PM 2.5 exposure suppressed the NLRP3 inflammasome activation and the AHR-TIPARP signaling pathway, by which compromised the anti-influenza immunity. Thus, our study revealed that PM2.5 could alter macrophage inflammatory responses by suppressing LPS-induced activation of NLRP3 inflammasome and expression of IFN-ß during influenza infection. These findings provided us new insights in understanding that PM2.5 combining with influenza infection could enhance the severity of pneumonia.


Asunto(s)
Contaminantes Atmosféricos/toxicidad , Inflamasomas/efectos de los fármacos , Interferón beta/biosíntesis , Proteína con Dominio Pirina 3 de la Familia NLR/efectos de los fármacos , Infecciones por Orthomyxoviridae/inmunología , Material Particulado/toxicidad , Animales , Inflamasomas/inmunología , Inflamasomas/metabolismo , Subtipo H1N1 del Virus de la Influenza A , Interferón beta/inmunología , Macrófagos/efectos de los fármacos , Macrófagos/inmunología , Macrófagos/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteína con Dominio Pirina 3 de la Familia NLR/inmunología , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Infecciones por Orthomyxoviridae/metabolismo
13.
Cell Signal ; 20(11): 2002-12, 2008 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-18713649

RESUMEN

The adaptor protein paxillin plays an important role in cell migration. Although the c-Jun amino-terminal kinase (JNK) phosphorylation of paxillin on Ser 178 has been found to be critical for cell migration, the precise mechanism by which JNK regulates cell migration is still not very clear. Here, the migration of human corneal epithelial (HCE) cells was used to determine which signaling pathways are involved in EGF-induced paxillin phosphorylation. Paxillin was phosphorylated on Tyr 31 and Tyr 118 after induction of migration by EGF in HCE cells. Specific inhibition of JNK activation by inhibitor SP600125 or overexpression of a dominant-negative JNK mutant not only blocked EGF-induced cell migration, but also eliminated tyrosine phosphorylation of paxillin on Tyr 31 and Tyr 118. HCE cells overexpressing paxillin-S178A mutant also exhibited lower mobility, and reduced phosphorylation of Tyr 31 and Tyr 118. However, paxillin-S178A-inhibited cell migration can be rescued by overexpression of paxillin-Y31E/Y118E mutant. Importantly, inhibition of JNK by SP600125 or overexpression of paxillin-S178A mutant prevented the association of FAK with paxillin. Taken together, these results suggest that phosphorylation of paxillin on Ser 178 by JNK is required for the association of paxillin with FAK, and subsequent tyrosine phosphorylation of paxillin.


Asunto(s)
Movimiento Celular , Células Epiteliales/citología , Epitelio Corneal/citología , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Paxillin/metabolismo , Fosfotirosina/metabolismo , Línea Celular , Movimiento Celular/efectos de los fármacos , Activación Enzimática/efectos de los fármacos , Factor de Crecimiento Epidérmico/farmacología , Células Epiteliales/efectos de los fármacos , Células Epiteliales/enzimología , Proteína-Tirosina Quinasas de Adhesión Focal/metabolismo , Humanos , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Modelos Biológicos , Proteínas Mutantes/metabolismo , Fosforilación/efectos de los fármacos , Fosfoserina/metabolismo
14.
Cell Signal ; 20(7): 1329-37, 2008 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-18456458

RESUMEN

MAP (Mitogen-activated protein) kinases play an important role in regulating many critical cellular processes. The inactivation of MAP kinases is always accomplished by a family of dual-specificity phosphatases, termed MAPK phosphatases (MKPs). Here, we have identified a novel MKP-like protein, designated DMKP-4, from the Drosophila genome. DMKP-4 is a protein of 387 amino acids, with a dual-specificity phosphatase (DSP) catalytic domain. Recombinant protein DMKP-4 retains intrinsic phosphatase activity against chromogenic substrate pNPP. Overexpression of DMKP-4 inhibited the activation of ERK, JNK and p38 by H(2)O(2), sorbitol and heat shock in HEK293-T cells, and JNK activation in Drosophila S2 cells under PGN stimuli. "Knockdown" of DMKP-4 expression by RNAi significantly enhanced the PGN-stimulated activation of JNK, but not ERK nor p38. Further study revealed that DMKP-4 interacted specifically with JNK via its DSP domain. Mutation of Cys-126 to serine in the DSP domain of DMKP-4 not only eliminated its interaction with JNK, but also markedly reduced its phosphatase activity. Thus, DMKP-4 is a Drosophila homologue of mammalian MKPs, and may play important roles in the regulation of various developmental processes.


Asunto(s)
Proteínas de Drosophila/metabolismo , Drosophila melanogaster/enzimología , Fosfatasas de Especificidad Dual/metabolismo , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Transducción de Señal , Secuencia de Aminoácidos , Compuestos de Anilina/metabolismo , Animales , Secuencia de Bases , Línea Celular , Cisteína/genética , Proteínas de Drosophila/química , Proteínas de Drosophila/genética , Drosophila melanogaster/citología , Fosfatasas de Especificidad Dual/química , Fosfatasas de Especificidad Dual/genética , Activación Enzimática , Células HeLa , Humanos , Modelos Biológicos , Datos de Secuencia Molecular , Mutación/genética , Compuestos Organofosforados/metabolismo , Fosforilación , Unión Proteica , Estructura Terciaria de Proteína , Transfección
15.
Cell Signal ; 19(2): 393-400, 2007 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-16978838

RESUMEN

Mitogen-activated protein (MAP) kinases play a critical role in innate immune responses to microbial infection through eliciting the biosynthesis of proinflammatory cytokines. MAP phosphatases (MKP)-1 is an archetypical member of the dual-specificity phosphatase family that deactivates MAP kinases. Induction of MKP-1 has been implicated in attenuating the lipopolysaccharide (LPS) and Peptidoglycan (PGN) responses, but how the expression of the MKP-1 is regulated is still not fully understood. Here, we show that inhibition of p38 MAP kinase by specific inhibitor SB 203580 or RNA interference (RNAi) markedly reduced the expression of MKP-1 in LPS or PGN-treated macrophages, which is correlated with prolonged activation of p38 and JNK. Depletion of MAPKAP kinase 2 (MK2), a downstream substrate of p38, by RNAi also inhibited the expression of MKP-1. The mRNA level of MKP-1 is not affected by inhibition of p38, but the expression of MKP-1 is inhibited by treatment of cycloheximide. Thus, p38 MAPK plays a critical role in mediating expression of MKP-1 at a post-transcriptional level. Furthermore, inhibition of p38 by SB 203580 prevented the expression of MKP-1 in LPS-tolerized macrophages, restored the activation of MAP kinases after LPS restimulation. These results indicate a critical role of p38-MK2-dependent induction of MKP-1 in innate immune responses.


Asunto(s)
Proteínas de Ciclo Celular/metabolismo , Retroalimentación Fisiológica , Regulación Enzimológica de la Expresión Génica , Proteínas Inmediatas-Precoces/metabolismo , Macrófagos/fisiología , Fosfoproteínas Fosfatasas/metabolismo , Proteínas Tirosina Fosfatasas/metabolismo , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo , Animales , Células Cultivadas , Interacciones Farmacológicas , Tolerancia a Medicamentos , Fosfatasa 1 de Especificidad Dual , Activación Enzimática , Imidazoles/farmacología , Péptidos y Proteínas de Señalización Intracelular , Lipopolisacáridos/farmacología , MAP Quinasa Quinasa 4/metabolismo , Masculino , Ratones , Ratones Endogámicos BALB C , Modelos Biológicos , Peptidoglicano/farmacología , Biosíntesis de Proteínas , Proteínas Quinasas/genética , Proteínas Quinasas/metabolismo , Proteína Fosfatasa 1 , Proteínas Serina-Treonina Quinasas , Piridinas/farmacología , Interferencia de ARN , Transducción de Señal , Proteínas Quinasas p38 Activadas por Mitógenos/genética , Proteínas Quinasas p38 Activadas por Mitógenos/fisiología
16.
Int Immunopharmacol ; 8(7): 951-8, 2008 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-18486905

RESUMEN

Andrographis paniculata Nees is an official herbal medicine for treatment of infection and inflammation in China. Andrograpanin, the one of diterpene lactones in A. paniculata, is a hydrolysate from neoandrographolide in vivo and in vitro. The goal of the present study was to investigate andrograpanin which effects on over production of nitric oxide (NO) and pro-inflammatory cytokines (TNF-alpha, IL-6 and IL-12p70) and the key signaling pathways involved in lipopolysaccharide (LPS)-activated macrophage cells. The results showed that NO and all three pro-inflammatory cytokines were inhibited by andrograpanin (15-90 microM) in a dose-dependent manner. The RT-PCR and western blotting assays showed that andrograpanin inhibited productions of NO and pro-inflammatory cytokines through down-regulating iNOS and pro-inflammatory cytokines gene expression levels. Further studies suggested that down-regulation of p38 mitogen-activated protein kinase (MAPKs) signaling pathways were involved in the anti-inflammatory activities of andrograpanin. This study provided evidences that andrograpanin might be useful as a potential anti-inflammatory leading compound for inflammatory drug development.


Asunto(s)
Andrographis/química , Antiinflamatorios/farmacología , Diterpenos/farmacología , Lipopolisacáridos/farmacología , Macrófagos/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Proteínas Quinasas p38 Activadas por Mitógenos/antagonistas & inhibidores , Animales , Supervivencia Celular/efectos de los fármacos , Citocinas/biosíntesis , Regulación hacia Abajo , Macrófagos/metabolismo , Ratones , Ratones Endogámicos BALB C , FN-kappa B/metabolismo , Óxido Nítrico/biosíntesis , Óxido Nítrico Sintasa de Tipo II/genética , Fosforilación
17.
Cell Signal ; 18(4): 441-8, 2006 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-16014325

RESUMEN

The p38 mitogen-activated protein kinase (MAPK) signaling pathway plays an important role in cellular responses to inflammatory stimuli and environmental stress. Activation of p38 is mediated through phosphorylation by upstream MAPKK, which in turn is activated by MAPKKK. However, the mechanism of how different upstream MAP2Ks and MAP3Ks specifically contribute to p38 activation in response to different stimuli is still not clearly understood. By using double-stranded RNA-mediated interference (RNAi) in Drosophila cells, we demonstrate that D-MKK3 is a major MAP2K responsible for D-p38 activation by UV, heat shock, NaCl or peptiodglycan (PGN). Stimulation of UV and PGN activates D-p38 through D-MEKK1, heat shock-induced activation of D-p38 signals through both D-MEKK1 and D-ASK1. On the other hand, maximal activation of D-p38 by NaCl requires the expression of four MAP3Ks.


Asunto(s)
Drosophila/metabolismo , MAP Quinasa Quinasa 2/metabolismo , MAP Quinasa Quinasa 3/metabolismo , Sistema de Señalización de MAP Quinasas/fisiología , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo , Animales , Línea Celular , Drosophila/efectos de los fármacos , Drosophila/efectos de la radiación , Calor , MAP Quinasa Quinasa 1/efectos de los fármacos , MAP Quinasa Quinasa 1/metabolismo , MAP Quinasa Quinasa 1/efectos de la radiación , MAP Quinasa Quinasa 2/efectos de los fármacos , MAP Quinasa Quinasa 2/efectos de la radiación , MAP Quinasa Quinasa 3/efectos de los fármacos , MAP Quinasa Quinasa 3/efectos de la radiación , MAP Quinasa Quinasa Quinasa 5/metabolismo , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Sistema de Señalización de MAP Quinasas/efectos de la radiación , Peptidoglicano/farmacología , Interferencia de ARN , ARN Bicatenario/farmacología , Transducción de Señal , Cloruro de Sodio/farmacología , Rayos Ultravioleta , Proteínas Quinasas p38 Activadas por Mitógenos/efectos de los fármacos , Proteínas Quinasas p38 Activadas por Mitógenos/efectos de la radiación
18.
Cell Signal ; 18(7): 964-70, 2006 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-16311020

RESUMEN

The TAK1 plays a pivotal role in the innate immune response of Drosophila by controlling the activation of JNK and NF-kappaB. Activation of TAK1 in mammals is mediated by two TAK1-binding proteins, TAB1 and TAB2, but the role of the TAB proteins in the immune response of Drosophila has not yet been established. Here, we report the identification of a TAB2-like protein in Drosophila called dTAB2. dTAB2 can interact with dTAK1, and stimulate the activation of the JNK and NF-kB signaling pathway. Furthermore, we have found that silencing of dTAB2 expression by dsRNAi inhibits JNK activation by peptidoglycans (PGN), but not by NaCl or sorbitol. In addition, suppression of dTAB2 blocked PGN-induced expression of antibacterial peptide genes, a function normally mediated by the activation of NF-kappaB signaling pathway. No significant effect on p38 activation by dTAB2 was found. These results suggest that dTAB2 is specifically required for PGN-induced activation of JNK and NF-kappaB signaling pathways.


Asunto(s)
Proteínas de Drosophila/fisiología , Drosophila melanogaster/inmunología , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , FN-kappa B/metabolismo , Animales , Células Cultivadas , Proteínas de Drosophila/biosíntesis , Proteínas de Drosophila/genética , Drosophila melanogaster/metabolismo , Activación Enzimática , Inmunidad Innata , Proteínas de Insectos/fisiología , FN-kappa B/farmacología , Peptidoglicano/farmacología , Unión Proteica , Interferencia de ARN , Transducción de Señal , Cloruro de Sodio/farmacología , Sorbitol/farmacología , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
19.
Cell Signal ; 28(9): 1292-1303, 2016 09.
Artículo en Inglés | MEDLINE | ID: mdl-27234131

RESUMEN

CD36, a scavenger receptor, plays an important role in the progression of atherosclerosis through its interaction with oxidized low-density lipoprotein (ox-LDL). Porphyromonas gingivalis (P. gingivalis, Pg) has been shown to promote macrophage-derived foam cell formation by affecting the expression of CD36. However, the regulatory role of CD36 in macrophages infected with Pg remains largely unknown. Therefore, the aim of the present study was to explore the molecular mechanism of Pg induced CD36 expression in macrophages. Our results showed that Pg promoted ox-LDL uptake by macrophages and the formation of foam cells. Pg infection increased CD36 mRNA and protein levels in ox-LDL-untreated macrophages. Moreover, small interferon RNA (siRNA) targeting CD36 significantly reduced foam cell formation induced by Pg. Additionally, Pg stimulated nuclear translocation of p65, which directly bound to the promoters of CD36 to facilitate its transcription. Inhibition of p65, NF-κB or ERK1/2 blocked Pg-induced CD36 production; whereas, overexpression of NF-κB subunits p65 and p50 upregulated CD36. Furthermore, Ras inhibitors significantly attenuated ERK1/2 activation and CD36 expression. Taken together, the data indicated that stimulation of the ERK/NF-κB pathway by Pg led to transactivation of the CD36 promoters, thereby upregulating CD36 expression in the infected macrophages. These findings may help design new treatment strategies in atherosclerosis.


Asunto(s)
Infecciones por Bacteroidaceae/genética , Infecciones por Bacteroidaceae/microbiología , Antígenos CD36/genética , Sistema de Señalización de MAP Quinasas , Macrófagos Peritoneales/microbiología , FN-kappa B/metabolismo , Porphyromonas gingivalis/fisiología , Regulación hacia Arriba/genética , Animales , Infecciones por Bacteroidaceae/patología , Antígenos CD36/metabolismo , Femenino , Células Espumosas/metabolismo , Células Espumosas/microbiología , Células Espumosas/patología , Células HEK293 , Humanos , Macrófagos Peritoneales/metabolismo , Macrófagos Peritoneales/patología , Ratones , Modelos Biológicos , Regiones Promotoras Genéticas/genética , Unión Proteica , Subunidades de Proteína/metabolismo , Células RAW 264.7
20.
Cell Signal ; 24(10): 1889-98, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-22683306

RESUMEN

The uptake of oxidized low density lipoprotein (ox-LDL) by macrophages usually leads to the formation of lipid-laden macrophages known as "foam cells," and this process plays an important role in the development of atherosclerosis. Ox-LDL activates mitogen-activated protein kinase (MAP) kinases and nuclear factor (NF)-κB, and activations of p38 and NF-κB are important for the formation of foam cells. MAP kinase phosphatase (MKP) 5 is a member of the dual specificity phosphatases (DUSPs) family that can selectively dephosphorylate activated MAPKs to regulate innate and adaptive immune responses. However, the role of MKP5 in the formation of foam cells remains unknown. Here, we found that stimulation of ox-LDL induces the expression of MKP5 in macrophages. MKP5 deficiency blocked the uptake of ox-LDL and the formation of foam cells. Further analysis revealed that deletion of MKP5 reduced the ox-LDL-induced activation of NF-κB. Also, MKP5 deficiency markedly inhibited the production of TNF-α, but enhanced the levels of TGF-ß1 in ox-LDL-stimulated macrophages. Moreover, inhibition of NF-κB by p65 RNAi significantly reduced foam cell formation in macrophages from WT mice relative to MKP5-deficient mice. Thus, MKP5 has an essential role in the formation of foam cells through activation of NF-κB, and MKP5 represents a novel target for the therapeutic intervention of atherosclerosis.


Asunto(s)
Fosfatasas de Especificidad Dual/inmunología , Células Espumosas/inmunología , Lipoproteínas LDL/inmunología , FN-kappa B/inmunología , Animales , Células Cultivadas , Fosfatasas de Especificidad Dual/genética , Quinasas MAP Reguladas por Señal Extracelular/genética , Quinasas MAP Reguladas por Señal Extracelular/inmunología , Femenino , Células Espumosas/citología , Células Espumosas/metabolismo , Regulación de la Expresión Génica , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Macrófagos/citología , Macrófagos/efectos de los fármacos , Macrófagos/inmunología , Macrófagos/metabolismo , Ratones , Ratones Endogámicos C57BL , Interferencia de ARN , Receptores Depuradores/genética , Factor de Crecimiento Transformador beta1/inmunología , Factor de Necrosis Tumoral alfa/inmunología , Regulación hacia Arriba , eIF-2 Quinasa/genética , eIF-2 Quinasa/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA