Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 36
Filtrar
1.
Immunity ; 42(4): 640-53, 2015 Apr 21.
Artículo en Inglés | MEDLINE | ID: mdl-25862090

RESUMEN

Acute lung injury (ALI) remains a serious health issue with little improvement in our understanding of the pathophysiology and therapeutic approaches. We investigated the mechanism that lipopolysaccharide (LPS) induces early neutrophil recruitment to lungs and increases pulmonary vascular permeability during ALI. Intratracheal LPS induced release of pro-interleukin-1α (IL-1α) from necrotic alveolar macrophages (AM), which activated endothelial cells (EC) to induce vascular leakage via loss of vascular endothelial (VE)-cadherin. LPS triggered the AM purinergic receptor P2X7(R) to induce Ca(2+) influx and ATP depletion, which led to necrosis. P2X7R deficiency significantly reduced necrotic death of AM and release of pro-IL-1α into the lung. CD14 was required for LPS binding to P2X7R, as CD14 neutralization significantly diminished LPS induced necrotic death of AM and pro-IL-1α release. These results demonstrate a key role for pro-IL-1α from necrotic alveolar macrophages in LPS-mediated ALI, as a critical initiator of increased vascular permeability and early neutrophil infiltration.


Asunto(s)
Interleucina-1alfa/inmunología , Receptores de Lipopolisacáridos/inmunología , Lipopolisacáridos/farmacología , Macrófagos Alveolares/efectos de los fármacos , Receptores Purinérgicos P2X7/inmunología , Lesión Pulmonar Aguda/inducido químicamente , Lesión Pulmonar Aguda/inmunología , Lesión Pulmonar Aguda/patología , Adenosina Trifosfato/metabolismo , Animales , Cadherinas/genética , Cadherinas/inmunología , Calcio/metabolismo , Permeabilidad Capilar/inmunología , Células Endoteliales/efectos de los fármacos , Células Endoteliales/inmunología , Células Endoteliales/patología , Regulación de la Expresión Génica , Células HEK293 , Humanos , Interleucina-1alfa/genética , Intubación Intratraqueal , Receptores de Lipopolisacáridos/genética , Pulmón/efectos de los fármacos , Pulmón/inmunología , Pulmón/patología , Macrófagos Alveolares/inmunología , Macrófagos Alveolares/patología , Ratones , Ratones Transgénicos , Necrosis/inducido químicamente , Necrosis/inmunología , Necrosis/patología , Infiltración Neutrófila , Neutrófilos/efectos de los fármacos , Neutrófilos/inmunología , Neutrófilos/patología , Precursores de Proteínas/genética , Precursores de Proteínas/inmunología , Receptores Purinérgicos P2X7/genética , Transducción de Señal
2.
Proc Natl Acad Sci U S A ; 118(1)2021 01 05.
Artículo en Inglés | MEDLINE | ID: mdl-33361152

RESUMEN

The balance between NLRP3 inflammasome activation and mitophagy is essential for homeostasis and cellular health, but this relationship remains poorly understood. Here we found that interleukin-1α (IL-1α)-deficient macrophages have reduced caspase-1 activity and diminished IL-1ß release, concurrent with reduced mitochondrial damage, suggesting a role for IL-1α in regulating this balance. LPS priming of macrophages induced pro-IL-1α translocation to mitochondria, where it directly interacted with mitochondrial cardiolipin (CL). Computational modeling revealed a likely CL binding motif in pro-IL-1α, similar to that found in LC3b. Thus, binding of pro-IL-1α to CL in activated macrophages may interrupt CL-LC3b-dependent mitophagy, leading to enhanced Nlrp3 inflammasome activation and more robust IL-1ß production. Mutation of pro-IL-1α residues predicted to be involved in CL binding resulted in reduced pro-IL-1α-CL interaction, a reduction in NLRP3 inflammasome activity, and increased mitophagy. These data identify a function for pro-IL-1α in regulating mitophagy and the potency of NLRP3 inflammasome activation.


Asunto(s)
Cardiolipinas/metabolismo , Interleucina-1alfa/metabolismo , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Animales , Autofagia , Cardiolipinas/fisiología , Caspasa 1/metabolismo , Femenino , Células HEK293 , Humanos , Inflamasomas/metabolismo , Interleucina-1alfa/fisiología , Macrófagos/metabolismo , Masculino , Ratones , Ratones Noqueados , Proteínas Asociadas a Microtúbulos/metabolismo , Mitocondrias/metabolismo , Mitofagia/fisiología , Proteína con Dominio Pirina 3 de la Familia NLR/fisiología , Unión Proteica/fisiología , Dominios Proteicos/fisiología , Especies Reactivas de Oxígeno/metabolismo
3.
Immunity ; 36(3): 401-14, 2012 Mar 23.
Artículo en Inglés | MEDLINE | ID: mdl-22342844

RESUMEN

We report that in the presence of signal 1 (NF-κB), the NLRP3 inflammasome was activated by mitochondrial apoptotic signaling that licensed production of interleukin-1ß (IL-1ß). NLRP3 secondary signal activators such as ATP induced mitochondrial dysfunction and apoptosis, resulting in release of oxidized mitochondrial DNA (mtDNA) into the cytosol, where it bound to and activated the NLRP3 inflammasome. The antiapoptotic protein Bcl-2 inversely regulated mitochondrial dysfunction and NLRP3 inflammasome activation. Mitochondrial DNA directly induced NLRP3 inflammasome activation, because macrophages lacking mtDNA had severely attenuated IL-1ß production, yet still underwent apoptosis. Both binding of oxidized mtDNA to the NLRP3 inflammasome and IL-1ß secretion could be competitively inhibited by the oxidized nucleoside 8-OH-dG. Thus, our data reveal that oxidized mtDNA released during programmed cell death causes activation of the NLRP3 inflammasome. These results provide a missing link between apoptosis and inflammasome activation, via binding of cytosolic oxidized mtDNA to the NLRP3 inflammasome.


Asunto(s)
Apoptosis/inmunología , Proteínas Portadoras/inmunología , Proteínas Portadoras/metabolismo , ADN Mitocondrial/inmunología , ADN Mitocondrial/metabolismo , Inflamasomas/inmunología , Inflamasomas/metabolismo , Animales , Expresión Génica , Interleucina-1beta/biosíntesis , Macrófagos/citología , Macrófagos/inmunología , Macrófagos/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , FN-kappa B/inmunología , Proteína con Dominio Pirina 3 de la Familia NLR , Oxidación-Reducción , Proteínas Proto-Oncogénicas c-bcl-2/genética , Proteínas Proto-Oncogénicas c-bcl-2/inmunología , Salmonella typhimurium/inmunología , Salmonella typhimurium/patogenicidad , Transducción de Señal
4.
Circ Res ; 119(6): e76-90, 2016 Sep 02.
Artículo en Inglés | MEDLINE | ID: mdl-27384322

RESUMEN

RATIONALE: Activation of NLRP3 (nucleotide-binding domain and leucine-rich repeat pyrin domain containing 3) inflammasome-mediating interleukin (IL)-1ß secretion has emerged as an important component of inflammatory processes in atherosclerosis. Mitochondrial DNA (mtDNA) damage is detrimental in atherosclerosis, and mitochondria are central regulators of the nucleotide-binding domain and leucine-rich repeat pyrin domain containing 3 inflammasome. Human atherosclerotic plaques express increased mtDNA damage. The major DNA glycosylase, 8-oxoguanine glycosylase (OGG1), is responsible for removing the most abundant form of oxidative DNA damage. OBJECTIVE: To test the role of OGG1 in the development of atherosclerosis in mouse. METHODS AND RESULTS: We observed that Ogg1 expression decreases over time in atherosclerotic lesion macrophages of low-density lipoprotein receptor (Ldlr) knockout mice fed a Western diet. Ogg1(-/-)Ldlr(-/-) mice fed a Western diet resulted in an increase in plaque size and lipid content. We found increased oxidized mtDNA, inflammasome activation, and apoptosis in atherosclerotic lesions and also higher serum IL-1ß and IL-18 in Ogg1(-/-)Ldlr(-/-) mice than in Ldlr(-/-). Transplantation with Ogg1(-/-) bone marrow into Ldlr(-/-) mice led to larger atherosclerotic lesions and increased IL-1ß production. However, transplantation of Ogg1(-/-)Nlrp3(-/-) bone marrow reversed the Ogg1(-/-) phenotype of increased plaque size. Ogg1(-/-) macrophages showed increased oxidized mtDNA and had greater amounts of cytosolic mtDNA and cytochrome c, increased apoptosis, and more IL-1ß secretion. Finally, we found that proatherogenic miR-33 can directly inhibit human OGG1 expression and indirectly suppress both mouse and human OGG1 via AMP-activated protein kinase. CONCLUSIONS: OGG1 plays a protective role in atherogenesis by preventing excessive inflammasome activation. Our study provides insight into a new target for therapeutic intervention based on a link between oxidative mtDNA damage, OGG1, and atherosclerosis via NLRP3 inflammasome.


Asunto(s)
Aterosclerosis/metabolismo , Aterosclerosis/prevención & control , ADN Glicosilasas/metabolismo , Reparación del ADN/fisiología , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Animales , Aterosclerosis/genética , ADN Glicosilasas/deficiencia , ADN Glicosilasas/genética , ADN Mitocondrial/genética , ADN Mitocondrial/metabolismo , Dieta Occidental/efectos adversos , Humanos , Inflamasomas/genética , Inflamasomas/metabolismo , Ratones , Ratones Noqueados , Proteína con Dominio Pirina 3 de la Familia NLR/genética
5.
J Immunol ; 194(4): 1686-94, 2015 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-25576596

RESUMEN

We previously identified a novel alternatively spliced isoform of human myeloid differentiation protein-2 (MD-2s) that competitively inhibits binding of MD-2 to TLR4 in vitro. In this study, we investigated the protective role of MD-2s in LPS-induced acute lung injury by delivering intratracheally an adenovirus construct that expressed MD-2s (Ad-MD-2s). After adenovirus-mediated gene transfer, MD-2s was strongly expressed in lung epithelial cells and readily detected in bronchoalveolar lavage fluid. Compared to adenovirus serotype 5 containing an empty vector lacking a transgene control mice, Ad-MD-2s delivery resulted in significantly less LPS-induced inflammation in the lungs, including less protein leakage, cell recruitment, and expression of proinflammatory cytokines and chemokines, such as IL-6, keratinocyte chemoattractant, and MIP-2. Bronchoalveolar lavage fluid from Ad-MD-2s mice transferred into lungs of naive mice before intratracheal LPS challenge diminished proinflammatory cytokine levels. As house dust mite (HDM) sensitization is dependent on TLR4 and HDM Der p 2, a structural homolog of MD-2, we also investigated the effect of MD-2s on HDM-induced allergic airway inflammation. Ad-MD-2s given before HDM sensitization significantly inhibited subsequent allergic airway inflammation after HDM challenge, including reductions in eosinophils, goblet cell hyperplasia, and IL-5 levels. Our study indicates that the alternatively spliced short isoform of human MD-2 could be a potential therapeutic candidate to treat human diseases induced or exacerbated by TLR4 signaling, such as Gram-negative bacterial endotoxin-induced lung injury and HDM-triggered allergic lung inflammation.


Asunto(s)
Antígeno 96 de los Linfocitos/inmunología , Neumonía/genética , Neumonía/inmunología , Lesión Pulmonar Aguda/genética , Lesión Pulmonar Aguda/inmunología , Empalme Alternativo , Animales , Western Blotting , Modelos Animales de Enfermedad , Femenino , Citometría de Flujo , Humanos , Hipersensibilidad/genética , Hipersensibilidad/inmunología , Inmunohistoquímica , Antígeno 96 de los Linfocitos/genética , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Isoformas de Proteínas/genética , Isoformas de Proteínas/inmunología , Reacción en Cadena en Tiempo Real de la Polimerasa , Receptor Toll-Like 4/inmunología , Transfección
6.
Arterioscler Thromb Vasc Biol ; 35(12): 2605-16, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26515418

RESUMEN

OBJECTIVE: Kawasaki disease (KD) is the most common cause of acute vasculitis and acquired cardiac disease among US children. We have previously shown that both TLR2/MyD88 and interleukin (IL)-1ß signaling are required for the Lactobacillus casei cell wall extract-induced KD vasculitis mouse model. The objectives of this study were to investigate the cellular origins of IL-1 production, the role of CD11c(+) dendritic cells and macrophages, and the relative contribution of hematopoietic and stromal cells for IL-1 responsive cells, as well the MyD88 signaling, in Lactobacillus casei cell wall extract-induced KD mouse model of vasculitis. APPROACH AND RESULTS: Using mouse knockout models and antibody depletion, we found that both IL-1α and IL-1ß were required for Lactobacillus casei cell wall extract-induced KD. Both dendritic cells and macrophages were necessary, and we found that MyD88 signaling was required in both hematopoietic and stromal cells. However, IL-1 response and signaling were critically required in nonendothelial stromal cells, but not in hematopoietic cells. CONCLUSIONS: Our results suggest that IL-1α and IL-1ß, as well as CD11c(+) dendritic cells and macrophages, are essential for the development of KD vasculitis and coronary arteritis in this mouse model. Bone marrow chimera experiments suggest that MyD88 signaling is important in both hematopoietic and stromal cells, whereas IL-1 signaling and response are required only in stromal cells, but not in endothelial cells. Determining the role of IL-1α and IL-1ß and of specific cell types in the KD vasculitis mouse model may have important implications for the design of more targeted therapies and understanding of the molecular mechanisms of KD immunopathologies.


Asunto(s)
Aortitis/metabolismo , Enfermedad de la Arteria Coronaria/metabolismo , Interleucina-1alfa/metabolismo , Interleucina-1beta/metabolismo , Síndrome Mucocutáneo Linfonodular/metabolismo , Transducción de Señal , Células del Estroma/metabolismo , Animales , Aorta/metabolismo , Aorta/patología , Aortitis/inducido químicamente , Aortitis/genética , Aortitis/patología , Células de la Médula Ósea/metabolismo , Trasplante de Médula Ósea , Proteínas Portadoras/metabolismo , Caspasa 1/metabolismo , Pared Celular , Células Cultivadas , Enfermedad de la Arteria Coronaria/inducido químicamente , Enfermedad de la Arteria Coronaria/genética , Enfermedad de la Arteria Coronaria/patología , Vasos Coronarios/metabolismo , Vasos Coronarios/patología , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Células Dendríticas/metabolismo , Modelos Animales de Enfermedad , Células Endoteliales/metabolismo , Lacticaseibacillus casei , Macrófagos/metabolismo , Ratones Endogámicos C57BL , Ratones Noqueados , Síndrome Mucocutáneo Linfonodular/inducido químicamente , Síndrome Mucocutáneo Linfonodular/genética , Síndrome Mucocutáneo Linfonodular/patología , Factor 88 de Diferenciación Mieloide/genética , Factor 88 de Diferenciación Mieloide/metabolismo , Proteína con Dominio Pirina 3 de la Familia NLR , Receptores Tipo I de Interleucina-1/genética , Receptores Tipo I de Interleucina-1/metabolismo , Células del Estroma/patología , Quimera por Trasplante
7.
Am J Respir Cell Mol Biol ; 50(2): 270-80, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24007300

RESUMEN

IL-1ß is a potent proinflammatory cytokine that is implicated in the pathogenesis of acute respiratory distress syndrome. We hypothesized that LPS and mechanical ventilation (MV) together could lead to IL-1ß secretion and the development of acute lung injury (ALI), and that this process would be dependent on caspase-1 and the nucleotide binding domain and leucine-rich repeat (NLR) pyrin domain containing 3 (NLRP3) inflammasome activation. The objectives of this study were to determine the specific role of IL-1ß, caspase-1, and the NLRP3 inflammasome in a two-hit model of ALI due to LPS plus MV. We used a two-hit murine model of ALI in which both inhaled LPS and MV were required for the development of hypoxemia, pulmonary neutrophil infiltration, and alveolar leakage. Nlrp3-deficent and Casp1-deficient mice had significantly diminished IL-1ß levels in bronchoalveolar lavage fluid, and were specifically protected from hypoxemia, despite similar alveolar neutrophil infiltration and leakage. The IL-1 receptor antagonist, Anakinra, significantly improved the specific development of hypoxemia without significant effects on neutrophil infiltration or alveolar leakage. MV resulted in increased bronchoalveolar lavage extracellular ATP and alveolar macrophage apoptosis as triggers of NLRP3 inflammasome activation. NLRP3 inflammasome activation and IL-1ß production play a key role in ALI caused by the combination of LPS and MV, particularly in the hypoxemia associated with acute respiratory distress syndrome. Blocking IL-1 signaling in this model specifically ameliorates hypoxemia, without affecting neutrophil infiltration and alveolar leakage, disassociating these readouts of ALI. MV causes alveolar macrophage apoptosis, a key step in the activation of NLRP3 inflammasome and production of IL-1ß.


Asunto(s)
Lesión Pulmonar Aguda/metabolismo , Proteínas Portadoras/metabolismo , Hipoxia/inmunología , Infiltración Neutrófila/inmunología , Lesión Pulmonar Aguda/inmunología , Animales , Caspasa 1/inmunología , Caspasa 1/metabolismo , Modelos Animales de Enfermedad , Inflamación/inmunología , Interleucina-1/inmunología , Interleucina-1/metabolismo , Interleucina-1beta/inmunología , Interleucina-1beta/metabolismo , Lipopolisacáridos/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Proteína con Dominio Pirina 3 de la Familia NLR , Receptores de Interleucina-1/inmunología , Receptores de Interleucina-1/metabolismo , Respiración Artificial/efectos adversos , Respiración Artificial/métodos , Transducción de Señal/inmunología
8.
Immunopharmacol Immunotoxicol ; 36(3): 237-41, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24852317

RESUMEN

The effect of spironolactone (SPIR) on lipopolysaccharide (LPS)-induced production of proinflammatory mediators was examined using RAW 264.7 macrophage-like cells and mouse peritoneal macrophages. SPIR significantly inhibited LPS-induced production of nitric oxide (NO), tumor necrosis factor-α and prostaglandin E2. The inhibition was not mediated by cell death. SPIR reduced the expression of an inducible NO synthase mRNA in response to LPS. SPIR significantly inhibited phosphorylation of p65 nuclear factor (NF)-κB in response to LPS. Furthermore, SPIR inhibited phosphorylation of IκB kinase (IKK) as an upstream molecule of NF-κB in response to LPS. LPS did not induce the production of aldosterone in RAW 264.7 cells. Taken together, SPIR is suggested to inhibit LPS-induced proinflammatory mediators via inactivation of IKK/NF-κB in LPS signaling.


Asunto(s)
Mediadores de Inflamación/antagonistas & inhibidores , Lipopolisacáridos/farmacología , FN-kappa B/antagonistas & inhibidores , Espironolactona/farmacología , Aldosterona/biosíntesis , Animales , Células Cultivadas , Dinoprostona/biosíntesis , Quinasa I-kappa B/metabolismo , Mediadores de Inflamación/metabolismo , Ratones , Óxido Nítrico/biosíntesis , Fosforilación , Factor de Necrosis Tumoral alfa/biosíntesis
9.
J Immunol ; 187(8): 4331-7, 2011 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-21918188

RESUMEN

Stimulation with LPS induces tyrosine phosphorylation of numerous proteins involved in the TLR signaling pathway. In this study, we demonstrated that myeloid differentiation factor-2 (MD-2) is also tyrosine phosphorylated following LPS stimulation. LPS-induced tyrosine phosphorylation of MD-2 is specific; it is blocked by the tyrosine kinase inhibitor, herbimycin A, as well as by an inhibitor of endocytosis, cytochalasin D, suggesting that MD-2 phosphorylation occurs during trafficking of MD-2 and not on the cell surface. Furthermore, we identified two possible phospho-accepting tyrosine residues at positions 22 and 131. Mutant proteins in which these tyrosines were changed to phenylalanine had reduced phosphorylation and significantly diminished ability to activate NF-κB in response to LPS. In addition, MD-2 coprecipitated and colocalized with Lyn kinase, most likely in the endoplasmic reticulum. A Lyn-binding peptide inhibitor abolished MD-2 tyrosine phosphorylation, suggesting that Lyn is a likely candidate to be the kinase required for MD-2 tyrosine phosphorylation. Our study demonstrated that tyrosine phosphorylation of MD-2 is important for signaling following exposure to LPS and underscores the importance of this event in mediating an efficient and prompt immune response.


Asunto(s)
Antígeno 96 de los Linfocitos/metabolismo , Fosfotirosina/metabolismo , Transducción de Señal/inmunología , Receptor Toll-Like 4/inmunología , Familia-src Quinasas/metabolismo , Ensayo de Inmunoadsorción Enzimática , Células HEK293 , Humanos , Immunoblotting , Inmunoprecipitación , Lipopolisacáridos/inmunología , Antígeno 96 de los Linfocitos/inmunología , Microscopía Fluorescente , Receptor Toll-Like 4/metabolismo , Familia-src Quinasas/inmunología
10.
J Transl Autoimmun ; 6: 100181, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-36619655

RESUMEN

Rationale: Effective therapies to reduce the severity and high mortality of pulmonary vasculitis and diffuse alveolar hemorrhage (DAH) in patients with systemic lupus erythematosus (SLE) is a serious unmet need. We explored whether biologic neutralization of eNAMPT (extracellular nicotinamide phosphoribosyl-transferase), a novel DAMP and Toll-like receptor 4 ligand, represents a viable therapeutic strategy in lupus vasculitis. Methods: Serum was collected from SLE subjects (n = 37) for eNAMPT protein measurements. In the preclinical pristane-induced murine model of lung vasculitis/hemorrhage, C57BL/6 J mice (n = 5-10/group) were treated with PBS, IgG (1 mg/kg), or the eNAMPT-neutralizing ALT-100 mAb (1 mg/kg, IP or subcutaneously (SQ). Lung injury evaluation (Day 10) included histology/immuno-histochemistry, BAL protein/cellularity, tissue biochemistry, RNA sequencing, and plasma biomarker assessment. Results: SLE subjects showed highly significant increases in blood NAMPT mRNA expression and eNAMPT protein levels compared to healthy controls. Preclinical pristane-exposed mice studies showed significantly increased NAMPT lung tissue expression and increased plasma eNAMPT levels accompanied by marked increases in alveolar hemorrhage and lung inflammation (BAL protein, PMNs, activated monocytes). In contrast, ALT-100 mAb-treated mice showed significant attenuation of inflammatory lung injury, alveolar hemorrhage, BAL protein, tissue leukocytes, and plasma inflammatory cytokines (eNAMPT, IL-6, IL-8). Lung RNA sequencing showed pristane-induced activation of inflammatory genes/pathways including NFkB, cytokine/chemokine, IL-1ß, and MMP signaling pathways, each rectified in ALT-100 mAb-treated mice. Conclusions: These findings highlight the role of eNAMPT/TLR4-mediated inflammatory signaling in the pathobiology of SLE pulmonary vasculitis and alveolar hemorrhage. Biologic neutralization of this novel DAMP appears to serve as a viable strategy to reduce the severity of SLE lung vasculitis.

11.
Front Immunol ; 14: 1155935, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37325640

RESUMEN

Introduction: Osteopontin (OPN; also known as SPP1), an immunomodulatory cytokine highly expressed in bone marrow-derived macrophages (BMMΦ), is known to regulate diverse cellular and molecular immune responses. We previously revealed that glatiramer acetate (GA) stimulation of BMMΦ upregulates OPN expression, promoting an anti-inflammatory, pro-healing phenotype, whereas OPN inhibition triggers a pro-inflammatory phenotype. However, the precise role of OPN in macrophage activation state is unknown. Methods: Here, we applied global proteome profiling via mass spectrometry (MS) analysis to gain a mechanistic understanding of OPN suppression versus induction in primary macrophage cultures. We analyzed protein networks and immune-related functional pathways in BMMΦ either with OPN knockout (OPNKO) or GA-mediated OPN induction compared with wild type (WT) macrophages. The most significant differentially expressed proteins (DEPs) were validated using immunocytochemistry, western blot, and immunoprecipitation assays. Results and discussion: We identified 631 DEPs in OPNKO or GA-stimulated macrophages as compared to WT macrophages. The two topmost downregulated DEPs in OPNKO macrophages were ubiquitin C-terminal hydrolase L1 (UCHL1), a crucial component of the ubiquitin-proteasome system (UPS), and the anti-inflammatory Heme oxygenase 1 (HMOX-1), whereas GA stimulation upregulated their expression. We found that UCHL1, previously described as a neuron-specific protein, is expressed by BMMΦ and its regulation in macrophages was OPN-dependent. Moreover, UCHL1 interacted with OPN in a protein complex. The effects of GA activation on inducing UCHL1 and anti-inflammatory macrophage profiles were mediated by OPN. Functional pathway analyses revealed two inversely regulated pathways in OPN-deficient macrophages: activated oxidative stress and lysosome-mitochondria-mediated apoptosis (e.g., ROS, Lamp1-2, ATP-synthase subunits, cathepsins, and cytochrome C and B subunits) and inhibited translation and proteolytic pathways (e.g., 60S and 40S ribosomal subunits and UPS proteins). In agreement with the proteome-bioinformatics data, western blot and immunocytochemical analyses revealed that OPN deficiency perturbs protein homeostasis in macrophages-inhibiting translation and protein turnover and inducing apoptosis-whereas OPN induction by GA restores cellular proteostasis. Taken together, OPN is essential for macrophage homeostatic balance via the regulation of protein synthesis, UCHL1-UPS axis, and mitochondria-mediated apoptotic processes, indicating its potential application in immune-based therapies.


Asunto(s)
Osteopontina , Complejo de la Endopetidasa Proteasomal , Osteopontina/genética , Osteopontina/metabolismo , Complejo de la Endopetidasa Proteasomal/metabolismo , Proteostasis , Proteoma/metabolismo , Macrófagos , Mitocondrias/metabolismo , Apoptosis
12.
Immunology ; 129(1): 97-104, 2010 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-20050332

RESUMEN

The regulatory role of tumour necrosis factor-a (TNF-a) on the expression of suppressor of cytokine signalling 3 (SOCS-3) in response to lipopolysaccharide (LPS) was examined using peritoneal macrophages from TNF-a-deficient mice. The LPS-induced SOCS-3 expression was markedly augmented in macrophages from wild-type mice whereas such augmentation was not seen in the cells from TNF-a-deficient mice. However, there was no significant difference in the level of SOCS-3 messenger RNA expression between macrophages from wild-type mice and those from TNF-a-deficient mice. The addition of exogenous TNF-a augmented the LPS-induced SOCS-3 expression in macrophages from TNF-a-deficient mice. The pulse chase analysis suggested augmented degradation of LPS-induced SOCS-3 protein in macrophages from TNF-a-deficient mice. Moreover, MG 132, a 26S proteasome inhibitor, sustained the LPS-induced SOCS-3 expression in those cells. The tyrosine phosphorylation of SOCS-3 was definitely induced in LPS-stimulated macrophages from TNF-a-deficient mice but not wild-type mice. A tyrosine phosphatase inhibitor enhanced the tyrosine phosphorylation of SOCS-3 in wild-type mice and accelerated the degradation. Therefore, it was suggested that TNF-a prevented the degradation of SOCS-3 protein via inhibition of the tyrosine phosphorylation in LPS-stimulated macrophages.


Asunto(s)
Macrófagos Peritoneales/metabolismo , Proteínas Tirosina Fosfatasas/metabolismo , Proteínas Recombinantes/farmacología , Proteínas Supresoras de la Señalización de Citocinas/biosíntesis , Factor de Necrosis Tumoral alfa/farmacología , Animales , Células Cultivadas , Inhibidores de Cisteína Proteinasa/farmacología , Leupeptinas/farmacología , Lipopolisacáridos/farmacología , Macrófagos Peritoneales/efectos de los fármacos , Macrófagos Peritoneales/inmunología , Macrófagos Peritoneales/patología , Masculino , Ratones , Ratones Noqueados , Fosforilación , Estabilidad Proteica/efectos de los fármacos , Proteína 3 Supresora de la Señalización de Citocinas , Proteínas Supresoras de la Señalización de Citocinas/genética , Proteínas Supresoras de la Señalización de Citocinas/inmunología , Factor de Necrosis Tumoral alfa/genética
13.
Immunology ; 131(1): 59-66, 2010 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-20406300

RESUMEN

Selective Alzheimer disease indicator-1 (seladin-1) is a broadly expressed oxidoreductase and is related to Alzheimer disease, cholesterol metabolism and carcinogenesis. The effect of lipopolysaccharide (LPS) on the expression of seladin-1 was examined using RAW 264.7 macrophage-like cells and murine peritoneal macrophages. Lipopolysaccharide induced the expression of seladin-1 protein and messenger RNA in those macrophages. The seladin-1 expression was also augmented by a series of Toll-like receptor ligands. The LPS augmented the expression of seladin-1 via reactive oxygen species generation and p38 activation. Seladin-1 inhibited LPS-induced activation of p38 but not nuclear factor-kappaB and inhibited the production of tumour necrosis factor-alpha in response to LPS. Moreover, seladin-1 inhibited LPS-induced osteoclast formation and enhanced LPS-induced alkaline phosphatase activity. Therefore, it was suggested that seladin-1 might be an LPS-responsible gene product and regulate the LPS-induced inflammatory response negatively.


Asunto(s)
Lipopolisacáridos/farmacología , Macrófagos/efectos de los fármacos , Proteínas del Tejido Nervioso/metabolismo , Oxidorreductasas actuantes sobre Donantes de Grupo CH-CH/metabolismo , Factor de Necrosis Tumoral alfa/antagonistas & inhibidores , Animales , Línea Celular , Lipopolisacáridos/inmunología , Macrófagos/inmunología , Macrófagos Peritoneales/efectos de los fármacos , Macrófagos Peritoneales/inmunología , Macrófagos Peritoneales/metabolismo , Ratones , Proteínas del Tejido Nervioso/efectos de los fármacos , Proteínas del Tejido Nervioso/genética , Osteoclastos/citología , Osteoclastos/metabolismo , Oxidorreductasas actuantes sobre Donantes de Grupo CH-CH/efectos de los fármacos , Oxidorreductasas actuantes sobre Donantes de Grupo CH-CH/genética , Reacción en Cadena de la Polimerasa , Factor de Necrosis Tumoral alfa/biosíntesis , Factor de Necrosis Tumoral alfa/efectos de los fármacos
14.
Cell Immunol ; 264(2): 114-8, 2010.
Artículo en Inglés | MEDLINE | ID: mdl-20557878

RESUMEN

The involvement of retinoblastoma protein-interacting zinc finger 1 (RIZ1), a tumor suppressor, in lipopolysaccharide (LPS)-induced inflammatory responses was investigated by using RAW 264.7 macrophage-like cells. LPS significantly augmented the expression of RIZ1 and the augmentation was mediated by the activation of nuclear factor (NF)-kappaB and Akt. The silencing of RIZ1 with the siRNA led to the inactivation of NF-kappaB in response to LPS. Moreover, the RIZ1 silencing caused the down-regulation of p53 activation and a p53 pharmacological inhibitor attenuated the RIZ1 expression. LPS-induced tumor necrosis factor-alpha and interleukin-6 production was prevented by RIZ1 siRNA or a p53 pharmacological inhibitor. Therefore, RIZ1 was suggested to augment LPS-induced NF-kappaB activation in collaboration with p53 and enhance the production of proinflammatory cytokines in response to LPS.


Asunto(s)
N-Metiltransferasa de Histona-Lisina/metabolismo , Interleucina-6/biosíntesis , Macrófagos/metabolismo , FN-kappa B/metabolismo , Factores de Transcripción/metabolismo , Factor de Necrosis Tumoral alfa/biosíntesis , Animales , Línea Celular , Clonación Molecular , N-Metiltransferasa de Histona-Lisina/genética , N-Metiltransferasa de Histona-Lisina/inmunología , Inflamación , Interleucina-6/genética , Interleucina-6/metabolismo , Lipopolisacáridos/inmunología , Lipopolisacáridos/metabolismo , Macrófagos/inmunología , Macrófagos/patología , Ratones , Mutación/genética , FN-kappa B/genética , ARN Interferente Pequeño/genética , Factores de Transcripción/genética , Factores de Transcripción/inmunología , Activación Transcripcional/genética , Factor de Necrosis Tumoral alfa/genética , Factor de Necrosis Tumoral alfa/metabolismo , Proteína p53 Supresora de Tumor/genética , Proteína p53 Supresora de Tumor/metabolismo
15.
Cell Immunol ; 261(2): 122-7, 2010.
Artículo en Inglés | MEDLINE | ID: mdl-20036355

RESUMEN

The effect of a series of toll-like receptor (TLR) ligands on the production of nitric oxide (NO) in mouse B1 cells was examined by using CD5(+) IgM(+) WEHI 231 cells. The stimulation with a series of TLR ligands, which were Pam3Csk4 for TLR1/2, poly I:C for TLR3, lipopolysaccharide (LPS) for TLR4, imiquimod for TLR7 and CpG DNA for TLR9, resulted in enhanced NO production via augmented expression of an inducible type of NO synthase (iNOS). LPS was most potent for the enhancement of NO production, followed by poly I:C and Pam3Csk4. Imiquimod and CpG DNA led to slight NO production. The LPS-induced NO production was dependent on MyD88-dependent pathway consisting of nuclear factor (NF)-kappaB and a series of mitogen-activated protein kinases (MAPKs). Further, it was also dependent on the MyD88-independent pathway consisting of toll-IL-1R domain-containing adaptor-inducing IFN-beta (TRIF) and interferon regulatory factor (IRF)-3. Physiologic peritoneal B1 cells also produced NO via the iNOS expression in response to LPS. The immunological significance of TLR ligands-induced NO production in B1 cells is discussed.


Asunto(s)
Linfocitos B/inmunología , Óxido Nítrico/metabolismo , Receptores Toll-Like/metabolismo , Proteínas Adaptadoras del Transporte Vesicular/genética , Proteínas Adaptadoras del Transporte Vesicular/metabolismo , Aminoquinolinas/metabolismo , Animales , Antígenos CD5/metabolismo , Línea Celular , ADN/metabolismo , Femenino , Imiquimod , Inmunoglobulina M/metabolismo , Inductores de Interferón/metabolismo , Ligandos , Lipopolisacáridos/metabolismo , Ratones , Ratones Endogámicos BALB C , Factor 88 de Diferenciación Mieloide/genética , Factor 88 de Diferenciación Mieloide/metabolismo , Óxido Nítrico Sintasa de Tipo II/genética , Óxido Nítrico Sintasa de Tipo II/metabolismo , Nitritos/metabolismo , Oligodesoxirribonucleótidos/genética , Oligodesoxirribonucleótidos/metabolismo , Poli I-C/metabolismo , Transducción de Señal/fisiología
16.
Cancer Invest ; 28(8): 806-12, 2010 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-20594067

RESUMEN

The role of retinoblastoma protein-interacting zinc finger 1 (RIZ1) on the cell growth of mouse and human monocytic leukemia cells was examined. RIZ1 expression was induced in response to tumor necrosis factor (TNF)-α. The expression was dependent on the nuclear factor-κB and AKT signaling. Further, RIZ1 expression led to the augmentation of p53 expression and the silencing of RIZ1 prevented it. On the other hand, a p53 inhibitor enhanced the TNF-α-induced RIZ1 expression. Silencing of RIZ1 augmented the proliferative activity of TNF-α-treated cells. Therefore, it is suggested that RIZ1 negatively regulated the cell proliferation of monocytic leukemia cells via activation of p53.


Asunto(s)
Proteínas de Unión al ADN/farmacología , N-Metiltransferasa de Histona-Lisina/farmacología , Leucemia Mieloide/patología , Proteínas Nucleares/farmacología , Factores de Transcripción/farmacología , Proteína p53 Supresora de Tumor/metabolismo , Animales , División Celular/efectos de los fármacos , Metilación de ADN , Cartilla de ADN , Proteínas de Unión al ADN/genética , Regulación Neoplásica de la Expresión Génica , Silenciador del Gen , N-Metiltransferasa de Histona-Lisina/genética , Homeostasis , Humanos , Leucemia Mieloide/genética , Ratones , Neoplasias/genética , Neoplasias/patología , Proteínas Nucleares/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Factores de Transcripción/genética , Transfección , Factor de Necrosis Tumoral alfa/genética , Proteína p53 Supresora de Tumor/efectos de los fármacos
17.
Immunology ; 128(1 Suppl): e700-6, 2009 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-19740331

RESUMEN

Astrocyte elevated gene-1 (AEG-1) is induced by human immunodeficiency virus 1 (HIV-1) infection and involved in tumour progression, migration and invasion as a nuclear factor-kappaB (NF-kappaB) -dependent gene. The involvement of AEG-1 on lipopolysaccharide (LPS) -induced proinflammatory cytokine production was examined. AEG-1 was induced via NF-kappaB activation in LPS-stimulated U937 human promonocytic cells. AEG-1 induced by LPS subsequently regulated NF-kappaB activation. The prevention of AEG-1 expression inhibited LPS-induced tumour necrosis factor-alpha and prostaglandin E(2) production. The AEG-1 activation was not induced by toll-like receptor ligands other than LPS. Therefore, AEG-1 was suggested to be a LPS-responsive gene and involved in LPS-induced inflammatory response.


Asunto(s)
Moléculas de Adhesión Celular/biosíntesis , Lipopolisacáridos/inmunología , Monocitos/inmunología , Receptor Toll-Like 4/metabolismo , Moléculas de Adhesión Celular/genética , Línea Celular Tumoral , Dinoprostona/biosíntesis , Dinoprostona/inmunología , Humanos , Proteínas de la Membrana , Monocitos/efectos de los fármacos , FN-kappa B/agonistas , FN-kappa B/metabolismo , Proteínas de Unión al ARN , Transducción de Señal , Factor de Necrosis Tumoral alfa/biosíntesis
18.
Cell Immunol ; 256(1-2): 99-103, 2009.
Artículo en Inglés | MEDLINE | ID: mdl-19251253

RESUMEN

The effect of toll-like receptor (TLR) 7 ligand pretreatment on the production of tumor necrosis factor (TNF)-alpha in response to TLR7 or TLR2 ligand was examined in order to establish a new TLR-mediated tolerance. RAW 264.7 macrophage-like cells were treated with imiquimod R837 as a TLR7 ligand for 18h, washed and incubated in fresh culture medium 6h. The second challenge with imiquimod R837 as a TLR7 ligand or Pam3CysSK4 as a TLR2 ligand resulted in reduced TNF-alpha production in TLR7 ligand-pretreated cells. There was impaired activation of NF-kappaB, p38 and stress-activated protein kinase (SAPK) in the tolerant cells. The expression of IRAK-M as a negative regulator of TLR signaling was markedly augmented in the tolerant cells while the interleukin-1 receptor-associated kinase (IRAK)-1 functioned normally. The involvement of IRAK-M in the TLR7-mediated tolerance is discussed.


Asunto(s)
Tolerancia Inmunológica/fisiología , Quinasas Asociadas a Receptores de Interleucina-1/inmunología , Macrófagos/inmunología , Glicoproteínas de Membrana/inmunología , Receptor Toll-Like 7/inmunología , Aminoquinolinas/farmacología , Animales , Secuencia de Bases , Línea Celular , Cartilla de ADN/genética , Imiquimod , Quinasas Asociadas a Receptores de Interleucina-1/antagonistas & inhibidores , Quinasas Asociadas a Receptores de Interleucina-1/genética , Ligandos , Lipopéptidos/farmacología , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Sistema de Señalización de MAP Quinasas/inmunología , Macrófagos/efectos de los fármacos , Macrófagos/metabolismo , Ratones , FN-kappa B/metabolismo , Óxido Nítrico/biosíntesis , ARN Interferente Pequeño/genética , Transducción de Señal/efectos de los fármacos , Transducción de Señal/inmunología , Receptor Toll-Like 2/metabolismo , Factor de Necrosis Tumoral alfa/biosíntesis , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
19.
Int J Med Microbiol ; 299(8): 554-62, 2009 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-19467928

RESUMEN

The effect of Clostridium perfringens alpha-toxin on production of tumor necrosis factor (TNF)-alpha and nitric oxide (NO) in lipopolysaccharide (LPS)-stimulated RAW 264.7 cells was studied. The pretreatment of wild type alpha-toxin, but not the inactive mutant, significantly decreased LPS-induced TNF-alpha and NO production. alpha-Toxin inhibited the expression of TNF-alpha and an inducible type of NO synthase protein and mRNA. Furthermore, it inhibited the phosphorylation of IkappaB-alpha and p65 NF-kappaB subunit, and the NF-kappaB luciferase reporter gene activity in LPS-stimulated cells. The pretreatment of alpha-toxin increased the level of intracellular ceramide. Taken together, Clostridium perfringens alpha-toxin pretreatment was suggested to inhibit LPS-induced TNF-alpha and NO production through the inhibition of NF-kappaB activation. The relationship between alpha-toxin-induced intracellular ceramide generation and the NF-kappaB inhibition is discussed.


Asunto(s)
Toxinas Bacterianas/inmunología , Toxinas Bacterianas/metabolismo , Proteínas de Unión al Calcio/inmunología , Proteínas de Unión al Calcio/metabolismo , Ceramidas/metabolismo , Lipopolisacáridos/inmunología , Óxido Nítrico/antagonistas & inhibidores , Factor de Necrosis Tumoral alfa/antagonistas & inhibidores , Fosfolipasas de Tipo C/inmunología , Fosfolipasas de Tipo C/metabolismo , Animales , Línea Celular , Regulación de la Expresión Génica/efectos de los fármacos , Genes Reporteros , Quinasa I-kappa B/metabolismo , Luciferasas/biosíntesis , Luciferasas/genética , Macrófagos/química , Macrófagos/efectos de los fármacos , Macrófagos/inmunología , Ratones , Fosforilación , Factor de Transcripción ReIA/metabolismo
20.
J Clin Invest ; 129(7): 2888-2897, 2019 04 30.
Artículo en Inglés | MEDLINE | ID: mdl-31038471

RESUMEN

Influenza A virus (IAV)-specific T cell responses are important correlates of protection during primary and subsequent infections. Generation and maintenance of robust IAV-specific T cell responses relies on T cell interactions with dendritic cells (DCs). In this study, we explore the role of nucleotide-binding domain leucine-rich repeat containing receptor family member NLRC4 in modulating the DC phenotype during IAV infection. Nlrc4-/- mice had worsened survival and increased viral titers during infection, normal innate immune cell recruitment and IAV-specific CD8 T cell responses, but severely blunted IAV-specific CD4 T cell responses compared to wild-type mice. The defect in the pulmonary IAV-specific CD4 T cell response was not a result of defective priming or migration of these cells in Nlrc4-/- mice but was instead due to an increase in FasL+ DCs, resulting in IAV-specific CD4 T cell death. Together, our data support a novel role for NLRC4 in regulating the phenotype of lung DCs during a respiratory viral infection, and thereby influencing the magnitude of protective T cell responses.


Asunto(s)
Proteínas Reguladoras de la Apoptosis/inmunología , Linfocitos T CD4-Positivos/inmunología , Proteínas de Unión al Calcio/inmunología , Células Dendríticas/inmunología , Proteína Ligando Fas/inmunología , Regulación de la Expresión Génica/inmunología , Virus de la Influenza A/inmunología , Pulmón/inmunología , Infecciones por Orthomyxoviridae/inmunología , Animales , Proteínas Reguladoras de la Apoptosis/genética , Linfocitos T CD4-Positivos/patología , Linfocitos T CD8-positivos/inmunología , Proteínas de Unión al Calcio/genética , Células Dendríticas/patología , Proteína Ligando Fas/genética , Pulmón/patología , Ratones , Ratones Noqueados , Infecciones por Orthomyxoviridae/patología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA