Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
1.
Arterioscler Thromb Vasc Biol ; 35(6): 1463-71, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25882065

RESUMEN

OBJECTIVE: Although it is accepted that macrophage glycolysis is upregulated under hypoxic conditions, it is not known whether this is linked to a similar increase in macrophage proinflammatory activation and whether specific energy demands regulate cell viability in the atheromatous plaque. APPROACH AND RESULTS: We studied the interplay between macrophage energy metabolism, polarization, and viability in the context of atherosclerosis. Cultured human and murine macrophages and an in vivo murine model of atherosclerosis were used to evaluate the mechanisms underlying metabolic and inflammatory activity of macrophages in the different atherosclerotic conditions analyzed. We observed that macrophage energetics and inflammatory activation are closely and linearly related, resulting in dynamic calibration of glycolysis to keep pace with inflammatory activity. In addition, we show that macrophage glycolysis and proinflammatory activation mainly depend on hypoxia-inducible factor and on its impact on glucose uptake, and on the expression of hexokinase II and ubiquitous 6-phosphofructo-2-kinase. As a consequence, hypoxia potentiates inflammation and glycolysis mainly via these pathways. Moreover, when macrophages' ability to increase glycolysis through 6-phosphofructo-2-kinase is experimentally attenuated, cell viability is reduced if subjected to proinflammatory or hypoxic conditions, but unaffected under control conditions. In addition to this, granulocyte-macrophage colony-stimulating factor enhances anerobic glycolysis while exerting a mild proinflammatory activation. CONCLUSIONS: These findings, in human and murine cells and in an animal model, show that hypoxia potentiates macrophage glycolytic flux in concert with a proportional upregulation of proinflammatory activity, in a manner that is dependent on both hypoxia-inducible factor -1α and 6-phosphofructo-2-kinase.


Asunto(s)
Aterosclerosis/metabolismo , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Macrófagos/metabolismo , Fosfofructoquinasa-2/metabolismo , Animales , Hipoxia de la Célula , Modelos Animales de Enfermedad , Glucólisis , Humanos , Inflamación/metabolismo , Ratones , Factor de Necrosis Tumoral alfa/metabolismo
2.
J Immunol ; 191(12): 6136-46, 2013 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-24249731

RESUMEN

Potassium channels modulate macrophage physiology. Blockade of voltage-dependent potassium channels (Kv) by specific antagonists decreases macrophage cytokine production and inhibits proliferation. In the presence of aspirin, acetylated cyclooxygenase-2 loses the activity required to synthesize PGs but maintains the oxygenase activity to produce 15R-HETE from arachidonate. This intermediate product is transformed via 5-LOX into epimeric lipoxins, termed 15-epi-lipoxins (15-epi-lipoxin A4 [e-LXA4]). Kv have been proposed as anti-inflammatory targets. Therefore, we studied the effects of e-LXA4 on signaling and on Kv and inward rectifier potassium channels (Kir) in mice bone marrow-derived macrophages (BMDM). Electrophysiological recordings were performed in these cells by the whole-cell patch-clamp technique. Treatment of BMDM with e-LXA4 inhibited LPS-dependent activation of NF-κB and IκB kinase ß activity, protected against LPS activation-dependent apoptosis, and enhanced the accumulation of the Nrf-2 transcription factor. Moreover, treatment of LPS-stimulated BMDM with e-LXA4 resulted in a rapid decrease of Kv currents, compatible with attenuation of the inflammatory response. Long-term treatment of LPS-stimulated BMDM with e-LXA4 significantly reverted LPS effects on Kv and Kir currents. Under these conditions, e-LXA4 decreased the calcium influx versus that observed in LPS-stimulated BMDM. These effects were partially mediated via the lipoxin receptor (ALX), because they were significantly reverted by a selective ALX receptor antagonist. We provide evidence for a new mechanism by which e-LXA4 contributes to inflammation resolution, consisting of the reversion of LPS effects on Kv and Kir currents in macrophages.


Asunto(s)
Inmunidad Innata/fisiología , Canal de Potasio Kv1.3/biosíntesis , Canal de Potasio Kv1.5/biosíntesis , Lipoxinas/farmacología , Activación de Macrófagos/fisiología , Canales de Potasio de Rectificación Interna/biosíntesis , Animales , Apoptosis/efectos de los fármacos , Calcio/fisiología , Regulación de la Expresión Génica , Inflamación/genética , Inflamación/metabolismo , Interleucina-13/farmacología , Interleucina-4/farmacología , Transporte Iónico , Canal de Potasio Kv1.3/antagonistas & inhibidores , Canal de Potasio Kv1.3/genética , Canal de Potasio Kv1.5/genética , Lipopolisacáridos/farmacología , Ratones , Ratones Endogámicos BALB C , Proteínas del Tejido Nervioso/biosíntesis , Proteínas del Tejido Nervioso/genética , Técnicas de Placa-Clamp , Potasio/fisiología , Bloqueadores de los Canales de Potasio/farmacología , Canales de Potasio de Rectificación Interna/genética , ARN Mensajero/biosíntesis , ARN Mensajero/genética , Receptores de Formil Péptido/agonistas , Receptores de Formil Péptido/fisiología , Venenos de Escorpión/farmacología , Organismos Libres de Patógenos Específicos , Regulación hacia Arriba
3.
J Immunol ; 190(8): 4226-35, 2013 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-23479225

RESUMEN

Extracellular nucleotides have been recognized as important modulators of inflammation via their action on specific pyrimidine receptors (P2). This regulation coexists with the temporal framework of proinflammatory and proresolution mediators released by the cells involved in the inflammatory response, including macrophages. Under proinflammatory conditions, the expression of cyclooxygenase-2 leads to the release of large amounts of PGs, such as PGE2, that exert their effects through EP receptors and other intracellular targets. The effect of these PGs on P2 receptors expressed in murine and human macrophages was investigated. In thioglycollate-elicited and alternatively activated macrophages, PGE2 selectively impairs P2Y but not P2X7 Ca(2+) mobilization. This effect is absent in LPS-activated cells and is specific for PGE2 because it cannot be reproduced by other PGs with cyclopentenone structure. The inhibition of P2Y responses by PGE2 involves the activation of nPKCs (PKCε) and PKD that can be abrogated by selective inhibitors or by expression of dominant-negative forms of PKD. The inhibition of P2Y signaling by PGE2 has an impact on the cell migration elicited by P2Y agonists in thioglycollate-elicited and alternatively activated macrophages, which provide new clues to understand the resolution phase of inflammation, when accumulation of PGE2, anti-inflammatory and proresolving mediators occurs.


Asunto(s)
Calcio/fisiología , Dinoprostona/fisiología , Macrófagos Peritoneales/inmunología , Macrófagos Peritoneales/metabolismo , Receptores Purinérgicos P2Y/fisiología , Transducción de Señal/inmunología , Animales , Señalización del Calcio/inmunología , Células Cultivadas , Ciclooxigenasa 2/biosíntesis , Ciclooxigenasa 2/genética , Dinoprostona/metabolismo , Inflamación/inmunología , Inflamación/metabolismo , Inflamación/patología , Líquido Intracelular/inmunología , Líquido Intracelular/metabolismo , Macrófagos Peritoneales/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Receptores Purinérgicos P2Y/deficiencia , Receptores Purinérgicos P2Y/metabolismo
4.
J Immunol ; 188(3): 1402-10, 2012 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-22190182

RESUMEN

The activation of immune cells in response to a pathogen involves a succession of signaling events leading to gene and protein expression, which requires metabolic changes to match the energy demands. The metabolic profile associated with the MAPK cascade (ERK1/2, p38, and JNK) in macrophages was studied, and the effect of its inhibition on the specific metabolic pattern of LPS stimulation was characterized. A [1,2-[(13)C](2)]glucose tracer-based metabolomic approach was used to examine the metabolic flux distribution in these cells after MEK/ERK inhibition. Bioinformatic tools were used to analyze changes in mass isotopomer distribution and changes in glucose and glutamine consumption and lactate production in basal and LPS-stimulated conditions in the presence and absence of the selective inhibitor of the MEK/ERK cascade, PD325901. Results showed that PD325901-mediated ERK1/2 inhibition significantly decreased glucose consumption and lactate production but did not affect glutamine consumption. These changes were accompanied by a decrease in the glycolytic flux, consistent with the observed decrease in fructose-2,6-bisphosphate concentration. The oxidative and nonoxidative pentose phosphate pathways and the ratio between them also decreased. However, tricarboxylic acid cycle flux did not change significantly. LPS activation led to the opposite responses, although all of these were suppressed by PD325901. However, LPS also induced a small decrease in pentose phosphate pathway fluxes and an increase in glutamine consumption that were not affected by PD325901. We concluded that inhibition of the MEK/ERK cascade interferes with central metabolism, and this cross-talk between signal transduction and metabolism also occurs in the presence of LPS.


Asunto(s)
Sistema de Señalización de MAP Quinasas/fisiología , Activación de Macrófagos , Macrófagos/metabolismo , Metabolómica/métodos , Metabolismo de los Hidratos de Carbono , Biología Computacional , Glucólisis , Lipopolisacáridos/farmacología , Metabolismo , Vía de Pentosa Fosfato
5.
Mediators Inflamm ; 2014: 832103, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25214717

RESUMEN

The nucleotide uridine trisphosphate (UTP) released to the extracellular milieu acts as a signaling molecule via activation of specific pyrimidine receptors (P2Y). P2Y receptors are G protein-coupled receptors expressed in many cell types. These receptors mediate several cell responses and they are involved in intracellular calcium mobilization. We investigated the role of the prostanoid PGE2 in P2Y signaling in mouse embryonic fibroblasts (MEFs), since these cells are involved in different ontogenic and physiopathological processes, among them is tissue repair following proinflammatory activation. Interestingly, Ca(2+)-mobilization induced by UTP-dependent P2Y activation was reduced by PGE2 when this prostanoid was produced by MEFs transfected with COX-2 or when PGE2 was added exogenously to the culture medium. This Ca(2+)-mobilization was important for the activation of different metabolic pathways in fibroblasts. Moreover, inhibition of COX-2 with selective coxibs prevented UTP-dependent P2Y activation in these cells. The inhibition of P2Y responses by PGE2 involves the activation of PKCs and PKD, a response that can be suppressed after pharmacological inhibition of these protein kinases. In addition to this, PGE2 reduces the fibroblast migration induced by P2Y-agonists such as UTP. Taken together, these data demonstrate that PGE2 is involved in the regulation of P2Y signaling in these cells.


Asunto(s)
Calcio/metabolismo , Ciclooxigenasa 2/metabolismo , Fibroblastos/metabolismo , Receptores Purinérgicos P2Y/metabolismo , Animales , Núcleo Celular/metabolismo , Inmunoensayo , Ratones , Ratones Endogámicos C57BL
6.
J Biol Chem ; 287(53): 44628-44, 2012 Dec 28.
Artículo en Inglés | MEDLINE | ID: mdl-23139414

RESUMEN

P2X7 receptors are involved not only in physiological functions but also in pathological brain processes. Although an increasing number of findings indicate that altered receptor expression has a causative role in neurodegenerative diseases and cancer, little is known about how expression of P2rx7 gene is controlled. Here we reported the first molecular and functional evidence that Specificity protein 1 (Sp1) transcription factor plays a pivotal role in the transcriptional regulation of P2X7 receptor. We delimited a minimal region in the murine P2rx7 promoter containing four SP1 sites, two of them being highly conserved in mammals. The functionality of these SP1 sites was confirmed by site-directed mutagenesis and Sp1 overexpression/down-regulation in neuroblastoma cells. Inhibition of Sp1-mediated transcriptional activation by mithramycin A reduced endogenous P2X7 receptor levels in primary cultures of cortical neurons and astrocytes. Using P2rx7-EGFP transgenic mice that express enhanced green fluorescent protein under the control of P2rx7 promoter, we found a high correlation between reporter expression and Sp1 levels in the brain, demonstrating that Sp1 is a key element in the transcriptional regulation of P2X7 receptor in the nervous system. Finally, we found that Sp1 mediates P2X7 receptor up-regulation in neuroblastoma cells cultured in the absence of serum, a condition that enhances chromatin accessibility and facilitates the exposure of SP1 binding sites.


Asunto(s)
Encéfalo/metabolismo , Regulación de la Expresión Génica , Receptores Purinérgicos P2X7/genética , Factor de Transcripción Sp1/metabolismo , Animales , Encéfalo/citología , Femenino , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Sistema Nervioso/metabolismo , Neuronas/metabolismo , Regiones Promotoras Genéticas , Receptores Purinérgicos P2X7/metabolismo , Factor de Transcripción Sp1/genética , Activación Transcripcional
7.
Redox Biol ; 17: 213-223, 2018 07.
Artículo en Inglés | MEDLINE | ID: mdl-29705509

RESUMEN

Protein tyrosine phosphatase 1B (PTP1B) is widely expressed in mammalian tissues, in particular in immune cells, and plays a pleiotropic role in dephosphorylating many substrates. Moreover, PTP1B expression is enhanced in response to pro-inflammatory stimuli and to different cell stressors. Taking advantage of the use of mice deficient in PTP1B we have investigated the effect of γ-radiation in these animals and found enhanced lethality and decreased respiratory exchange ratio vs. the corresponding wild type animals. Using bone-marrow derived macrophages and mouse embryonic fibroblasts (MEFs) from wild-type and PTP1B-deficient mice, we observed a differential response to various cell stressors. PTP1B-deficient macrophages exhibited an enhanced response to γ-radiation, UV-light, LPS and S-nitroso-glutathione. Macrophages exposed to γ-radiation show DNA damage and fragmentation, increased ROS production, a lack in GSH elevation and enhanced acidic ß-galactosidase activity. Interestingly, these differences were not observed in MEFs. Differential gene expression analysis of WT and KO macrophages revealed that the main pathways affected after irradiation were an up-regulation of protein secretion, TGF-ß signaling and angiogenesis among other, and downregulation of Myc targets and Hedgehog signaling. These results demonstrate a key role for PTP1B in the protection against the cytotoxicity of irradiation in intact animal and in macrophages, which might be therapeutically relevant.


Asunto(s)
Proliferación Celular/efectos de la radiación , Macrófagos/efectos de la radiación , Proteína Tirosina Fosfatasa no Receptora Tipo 1/genética , Traumatismos por Radiación/genética , Animales , Daño del ADN/efectos de la radiación , Fibroblastos/efectos de la radiación , Rayos gamma/efectos adversos , Regulación de la Expresión Génica/efectos de la radiación , Glutatión/genética , Glutatión/metabolismo , Ratones , Ratones Noqueados , Fosforilación/efectos de la radiación , Proteína Tirosina Fosfatasa no Receptora Tipo 1/deficiencia , Interferencia de ARN , Traumatismos por Radiación/patología , Traumatismos por Radiación/prevención & control , Especies Reactivas de Oxígeno/metabolismo , beta-Galactosidasa/genética
8.
Chem Biol ; 21(8): 955-66, 2014 Aug 14.
Artículo en Inglés | MEDLINE | ID: mdl-25065531

RESUMEN

The effect of acanthoic acid analogs on the response to proinflammatory challenge was investigated. Some pimarane diterpenes are known activators of the LXRαß nuclear receptors, but we show here that they also exert a rapid, potent, and selective activation of the p110γ and p110δ subunits of PI3K. Combination of these effects results in an important attenuation of the global transcriptional response to LPS in macrophages. PI3K/Akt activation leads to inhibition of the LPS-dependent stimulation of IKK/NF-κB and p38 and ERK MAPKs. Macrophages from LXRαß-deficient mice exhibited an inhibition of these pathways similar to the corresponding wild-type cells. Silencing or inhibition of p110γ/δ suppressed the effect of these diterpenes (DTPs) on IKK/NF-κB and MAPKs signaling. Taken together, these data show a multitarget anti-inflammatory mechanism by these DTPs including a selective activation of PI3K isoenzymes.


Asunto(s)
Antiinflamatorios no Esteroideos/farmacología , Fosfatidilinositol 3-Quinasa Clase Ia/química , Fosfatidilinositol 3-Quinasa Clase Ia/metabolismo , Diterpenos/farmacología , FN-kappa B/antagonistas & inhibidores , Subunidades de Proteína/metabolismo , Animales , Antiinflamatorios no Esteroideos/química , Diterpenos/química , Relación Dosis-Respuesta a Droga , Lipopolisacáridos/antagonistas & inhibidores , Lipopolisacáridos/farmacología , Receptores X del Hígado , Macrófagos/efectos de los fármacos , Macrófagos/metabolismo , Ratones , Ratones Endogámicos BALB C , Ratones Noqueados , Conformación Molecular , FN-kappa B/metabolismo , Células 3T3 NIH , Receptores Nucleares Huérfanos/deficiencia , Receptores Nucleares Huérfanos/metabolismo , Subunidades de Proteína/química , Relación Estructura-Actividad
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA