Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 58
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
Mol Med ; 21(1): 1025-1037, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26623925

RESUMEN

Although the molecular links underlying the causative relationship between chronic low-grade inflammation and insulin resistance are not completely understood, compelling evidence suggests a pivotal role of the nucleotide-binding oligomerization domain (NOD)-like receptor pyrin domain containing 3 (NLRP3) inflammasome. Here we tested the hypothesis that either a selective pharmacological inhibition or a genetic downregulation of the NLRP3 inflammasome results in reduction of the diet-induced metabolic alterations. Male C57/BL6 wild-type mice and NLRP3-/- littermates were fed control diet or high-fat, high-fructose diet (HD). A subgroup of HD-fed wild-type mice was treated with the NLRP3 inflammasome inhibitor BAY 11-7082 (3 mg/kg intraperitoneally [IP]). HD feeding increased plasma and hepatic lipids and impaired glucose homeostasis and renal function. Renal and hepatic injury was associated with robust increases in profibrogenic markers, while only minimal fibrosis was recorded. None of these metabolic abnormalities were detected in HD-fed NLRP3-/- mice, and they were dramatically reduced in HD-mice treated with the NLRP3 inflammasome inhibitor. BAY 11-7082 also attenuated the diet-induced increase in NLRP3 inflammasome expression, resulting in inhibition of caspase-1 activation and interleukin (IL)-1ß and IL-18 production (in liver and kidney). Interestingly, BAY 11-7082, but not gene silencing, inhibited nuclear factor (NF)-κB nuclear translocation. Overall, these results demonstrate that the selective pharmacological modulation of the NLRP3 inflammasome attenuates the metabolic abnormalities and the related organ injury/dysfunction caused by chronic exposure to HD, with effects similar to those obtained by NLRP3 gene silencing.

2.
J Pharmacol Exp Ther ; 359(1): 45-53, 2016 10.
Artículo en Inglés | MEDLINE | ID: mdl-27440421

RESUMEN

The aim of this study was to evaluate the effects of chronic treatment with empagliflozin, a potent and selective sodium glucose cotransporter-2 inhibitor, in a murine model of diet-induced obesity and insulin resistance, focusing on drug effects on body weight reduction and nucleotide-binding domain, leucine-rich repeat containing protein (NLRP)-3 inflammasome activation, which have never been investigated to date. Male C57BL/6 mice were fed control or a high fat-high sugar (HFHS) diet for 4 months. Over the last 2 months, subsets of animals were treated with empagliflozin (1-10 mg/kg) added to the diet. Empagliflozin evoked body weight reduction (P < 0.001 for the highest dose) and positive effects on fasting glycemia and homeostasis model assessment of insulin resistance. In addition, the drug was able to reduce renal tubular damage and liver triglycerides level in a dose-dependent manner. Interestingly, empagliflozin also decreased cardiac lipid accumulation. Moreover, diet-induced activation of NLRP-3 in kidney and liver (not observed in the heart) was dose-dependently attenuated by empagliflozin. Our results clearly demonstrate the ability of empagliflozin to counteract the deleterious effects evoked by chronic exposure to HFHS diet. Most notably, empagliflozin treatment was associated with NLRP-3 inflammasome signaling modulation, suggesting that this inhibition may contribute to the drug therapeutic effects.


Asunto(s)
Compuestos de Bencidrilo/farmacología , Dieta/efectos adversos , Glucósidos/farmacología , Inflamasomas/metabolismo , Metabolismo de los Lípidos/efectos de los fármacos , Animales , Compuestos de Bencidrilo/uso terapéutico , Glucemia/metabolismo , Peso Corporal/efectos de los fármacos , Ayuno/sangre , Prueba de Tolerancia a la Glucosa , Glucósidos/uso terapéutico , Riñón/efectos de los fármacos , Riñón/metabolismo , Hígado/efectos de los fármacos , Hígado/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Miocardio/metabolismo , Obesidad/inducido químicamente , Obesidad/tratamiento farmacológico , Obesidad/metabolismo
3.
Pharmacol Res ; 114: 27-38, 2016 12.
Artículo en Inglés | MEDLINE | ID: mdl-27751877

RESUMEN

Histamine has been reported to decrease the ultrafiltration coefficient, which inversely correlates with glomerular permselectivity, however the mechanism(s) underling this effect have never been investigated. This study aimed to assess whether histamine could exert a direct detrimental effect on podocyte permeability and the possible involvement of two key proteins for the glomerular slit diaphragm (SD) integrity, zonula occludens-1 (ZO-1) and P-cadherin. The effect of histamine (100 pM-1000nM) on coloured podocytes junctional integrity was evaluated functionally by a transwell assay of monolayer permeability and morphologically by electron microscopy. Histamine receptor (H1-4R) presence was evaluated at both mRNA (RT-PCR) and protein (immunofluorescence) levels. The Kd and Bmax values for [3H]mepyramine were determined by saturation binding analysis; IP1 and cAMP production evoked by histamine were measured by TR-FRET. ZO-1, P-cadherin and vimentin expression was assessed by qRT-PCR and quantitative immunoblotting. Histamine elicited a time- and sigmoidal dose-dependent (maximum effect at 8h, 10nM) increase in podocyte paracellular permeability widening the paracellular spaces. Only H1R was predominantly localised to the podocyte membrane. Consistently, histamine elicited a sigmoidal dose-dependent increase in IP1, but not in cAMP. Histamine exposure evoked a concentration-dependent reduction in both ZO-1 and P-cadherin and a parallel induction of vimentin mRNA expression with a maximum effect after 6h, and protein expression with a maximum effect after 8h. These effects were prevented by the selective H1R antagonist chlorpheniramine. In conclusion, our data demonstrate that histamine, via the H1R, modifies SD morphological and functional integrity, in part, by decreasing the expression of ZO-1 and P-cadherin.


Asunto(s)
Agonistas de los Receptores Histamínicos/efectos adversos , Histamina/efectos adversos , Glomérulos Renales/efectos de los fármacos , Podocitos/efectos de los fármacos , Receptores Histamínicos H1/metabolismo , Cadherinas/análisis , Cadherinas/metabolismo , Permeabilidad de la Membrana Celular/efectos de los fármacos , Células Cultivadas , Humanos , Glomérulos Renales/metabolismo , Glomérulos Renales/patología , Glomérulos Renales/ultraestructura , Podocitos/metabolismo , Podocitos/patología , Podocitos/ultraestructura , Proteína de la Zonula Occludens-1/análisis , Proteína de la Zonula Occludens-1/metabolismo
4.
J Immunol ; 192(10): 4921-31, 2014 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-24729612

RESUMEN

Vascular endothelial cells (ECs) and several cancer cells express B7h, which is the ligand of the ICOS T cell costimulatory molecule. We have previously shown that B7h triggering via a soluble form of ICOS (ICOS-Fc) inhibits the adhesion of polymorphonuclear and tumor cell lines to HUVECs; thus, we suggested that ICOS-Fc may act as an anti-inflammatory and antitumor agent. Because cancer cell migration and angiogenesis are crucial for metastasis dissemination, the aim of this work was to evaluate the effect of ICOS-Fc on the migration of cancer cells and ECs. ICOS-Fc specifically inhibited the migration of HUVECs, human dermal lymphatic ECs, and the HT29, HCT116, PC-3, HepG2, JR8, and M14 tumor cell lines expressing high levels of B7h, whereas it was ineffective in the RPMI7932, PCF-2, LM, and BHT-101 cell lines expressing low levels of B7h. Furthermore, ICOS-Fc downmodulated hepatocyte growth factor facilitated the epithelial-to-mesenchymal transition in HepG2 cells. Moreover, ICOS-Fc downmodulated the phosphorylation of focal adhesion kinase and the expression of ß-Pix in both HUVECs and tumor cell lines. Finally, treatment with ICOS-Fc inhibited the development of lung metastases upon injection of NOD-SCID-IL2Rγnull mice with CF-PAC1 cells, as well as C57BL/6 mice with B16-F10 cells. Therefore, the B7h-ICOS interaction may modulate the spread of cancer metastases, which suggests the novel use of ICOS-Fc as an immunomodulatory drug. However, in the B16-F10-metastasized lungs, ICOS-Fc also increased IL-17A/RORc and decreased IL-10/Foxp3 expression, which indicates that it also exerts positive effects on the antitumor immune response.


Asunto(s)
Movimiento Celular/inmunología , Ligando Coestimulador de Linfocitos T Inducibles/inmunología , Neoplasias Pulmonares/inmunología , Animales , Células Hep G2 , Xenoinjertos , Células Endoteliales de la Vena Umbilical Humana , Humanos , Fragmentos Fc de Inmunoglobulinas/genética , Fragmentos Fc de Inmunoglobulinas/inmunología , Ligando Coestimulador de Linfocitos T Inducibles/genética , Proteína Coestimuladora de Linfocitos T Inducibles/genética , Proteína Coestimuladora de Linfocitos T Inducibles/inmunología , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patología , Neoplasias Pulmonares/secundario , Ratones , Ratones Endogámicos NOD , Ratones SCID , Metástasis de la Neoplasia , Trasplante de Neoplasias , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/inmunología , Proteínas Recombinantes de Fusión/farmacología
5.
J Immunol ; 190(3): 1125-34, 2013 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-23275603

RESUMEN

B7h, expressed by several cell types, binds ICOS expressed by activated T cells. We have previously shown that B7h triggering by ICOS-Fc inhibits human endothelial cell adhesiveness. This work investigated the effect of ICOS-Fc on human monocyte-derived dendritic cells (DCs). We found that DCs matured with LPS in the presence of ICOS-Fc (mDCs(ICOS)) produced greater amounts of IL-23 and IL-10, and promoted a higher secretion of IL-17A and IL-17F in MLCs than did those DCs matured with LPS alone (mDCs). Moreover, mDCs(ICOS) pulsed with the keyhole limpet hemocyanin Ag during the maturation phase were better stimulators of Ag-specific MHC class I-, but not class II-restricted T cells than mDCs. This was probably due to promotion of cross-presentation because it was not detected when the Flu-MA(58-66) Ag was directly loaded on already matured DCs and mDCs(ICOS). Finally, ICOS-Fc inhibited the adhesion of both immature DCs and mDCs to vascular and lymphoid endothelial cells, their migratory activity, and the expression of the Rac-1 activator ß-Pix involved in cell motility. These data suggest that B7h stimulation modulates DC function with effects on their maturation and recruitment into tissues. This opens a novel view on the use of interactors of the ICOS:B7h system as immunomodulatory drugs.


Asunto(s)
Células Dendríticas/efectos de los fármacos , Ligando Coestimulador de Linfocitos T Inducibles/inmunología , Proteína Coestimuladora de Linfocitos T Inducibles/inmunología , Presentación de Antígeno/efectos de los fármacos , Adhesión Celular/efectos de los fármacos , Diferenciación Celular/efectos de los fármacos , Movimiento Celular/efectos de los fármacos , Células Cultivadas/citología , Células Cultivadas/inmunología , Células Dendríticas/citología , Células Dendríticas/inmunología , Factores de Intercambio de Guanina Nucleótido/biosíntesis , Factores de Intercambio de Guanina Nucleótido/genética , Antígeno HLA-A2/inmunología , Hemocianinas/farmacología , Células Endoteliales de la Vena Umbilical Humana/citología , Humanos , Fragmentos Fc de Inmunoglobulinas/inmunología , Inmunoglobulina G/inmunología , Proteína Coestimuladora de Linfocitos T Inducibles/genética , Proteína Coestimuladora de Linfocitos T Inducibles/metabolismo , Lipopolisacáridos/farmacología , Activación de Linfocitos/efectos de los fármacos , Activación de Linfocitos/inmunología , Prueba de Cultivo Mixto de Linfocitos , Monocitos/citología , Proteínas Recombinantes de Fusión/farmacología , Factores de Intercambio de Guanina Nucleótido Rho , Transducción de Señal/efectos de los fármacos
6.
J Cell Mol Med ; 17(11): 1494-505, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-24079335

RESUMEN

Although recent preclinical and clinical studies have demonstrated that recombinant human relaxin (rhRLX) may have important therapeutic potential in acute heart failure and chronic kidney diseases, the effects of acute rhRLX administration against renal ischaemia/reperfusion (I/R) injury have never been investigated. Using a rat model of 1-hr bilateral renal artery occlusion followed by 6-hr reperfusion, we investigated the effects of rhRLX (5 µg/Kg i.v.) given both at the beginning and after 3 hrs of reperfusion. Acute rhRLX administration attenuated the functional renal injury (increase in serum urea and creatinine), glomerular dysfunction (decrease in creatinine clearance) and tubular dysfunction (increase in urinary excretion of N-acetyl-ß-glucosaminidase) evoked by renal I/R. These beneficial effects were accompanied by a significant reduction in local lipid peroxidation, free radical-induced DNA damage and increase in the expression/activity of the endogenous antioxidant enzymes Mn- and CuZn-superoxide dismutases (SOD). Furthermore, rhRLX administration attenuated the increase in leucocyte activation, as suggested by inhibition of myeloperoxidase activity, intercellular-adhesion-molecule-1 expression, interleukin (IL)-1ß, IL-18 and tumour necrosis factor-α production as well as increase in IL-10 production. Interestingly, the reduced oxidative stress status and neutrophil activation here reported were associated with rhRLX-induced activation of endothelial nitric oxide synthase and up-regulation of inducible nitric oxide synthase, possibly secondary to activation of Akt and the extracellular signal-regulated protein kinase (ERK) 1/2, respectively. Thus, we report herein that rhRLX protects the kidney against I/R injury by a mechanism that involves changes in nitric oxide signalling pathway.


Asunto(s)
Antiinflamatorios/uso terapéutico , Enfermedades Renales/prevención & control , Riñón/efectos de los fármacos , Relaxina/uso terapéutico , Daño por Reperfusión/prevención & control , Animales , Antiinflamatorios/farmacología , Evaluación Preclínica de Medicamentos , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Humanos , Mediadores de Inflamación/metabolismo , Isquemia/tratamiento farmacológico , Riñón/irrigación sanguínea , Riñón/enzimología , Riñón/fisiopatología , Sistema de Señalización de MAP Quinasas , Masculino , Óxido Nítrico Sintasa de Tipo II/metabolismo , Óxido Nítrico Sintasa de Tipo III/metabolismo , Estrés Oxidativo , Fosforilación , Procesamiento Proteico-Postraduccional , Proteínas Proto-Oncogénicas c-akt/metabolismo , Ratas , Ratas Wistar , Relaxina/farmacología
7.
Mediators Inflamm ; 2013: 509502, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23861559

RESUMEN

Peroxisome Proliferator Activated Receptor (PPAR)- δ agonists may serve for treating metabolic diseases. However, the effects of PPAR- δ agonism within the skeletal muscle, which plays a key role in whole-body glucose metabolism, remain unclear. This study aimed to investigate the signaling pathways activated in the gastrocnemius muscle by chronic administration of the selective PPAR- δ agonist, GW0742 (1 mg/kg/day for 16 weeks), in male C57Bl6/J mice treated for 30 weeks with high-fructose corn syrup (HFCS), the major sweetener in foods and soft-drinks (15% wt/vol in drinking water). Mice fed with the HFCS diet exhibited hyperlipidemia, hyperinsulinemia, hyperleptinemia, and hypoadiponectinemia. In the gastrocnemius muscle, HFCS impaired insulin and AMP-activated protein kinase signaling pathways and reduced GLUT-4 and GLUT-5 expression and membrane translocation. GW0742 administration induced PPAR- δ upregulation and improvement in glucose and lipid metabolism. Diet-induced activation of nuclear factor-κB and expression of inducible-nitric-oxide-synthase and intercellular-adhesion-molecule-1 were attenuated by drug treatment. These effects were accompanied by reduction in the serum concentration of interleukin-6 and increase in muscular expression of fibroblast growth factor-21. Overall, here we show that PPAR- δ activation protects the skeletal muscle against the metabolic abnormalities caused by chronic HFCS exposure by affecting multiple levels of the insulin and inflammatory cascades.


Asunto(s)
Carbohidratos/administración & dosificación , Regulación de la Expresión Génica , Inflamación/metabolismo , Resistencia a la Insulina , Músculo Esquelético/metabolismo , PPAR delta/metabolismo , Animales , Factores de Crecimiento de Fibroblastos/metabolismo , Glucosa/metabolismo , Prueba de Tolerancia a la Glucosa , Inmunohistoquímica , Insulina/metabolismo , Interleucina-6/metabolismo , Metabolismo de los Lípidos , Masculino , Ratones , Ratones Endogámicos C57BL , Edulcorantes Nutritivos/administración & dosificación , PPAR delta/agonistas , Transducción de Señal , Tiazoles/farmacología
8.
J Cell Mol Med ; 16(9): 2196-207, 2012 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-22248092

RESUMEN

Heparin and low molecular weight heparins have been demonstrated to reduce myocardial ischaemia/reperfusion (I/R) injury, although their use is hampered by the risk of haemorrhagic and thrombotic complications. Chemical and enzymatic modifications of K5 polysaccharide have shown the possibility of producing heparin-like compounds with low anticoagulant activity and strong anti-inflammatory effects. Using a rat model of regional myocardial I/R, we investigated the effects of an epimerized N-,O-sulphated K5 polysaccharide derivative, K5-N,OSepi, on infarct size and histological signs of myocardial injury caused by 30 min. ligature of the left anterior descending coronary artery followed by 1 or 24 h reperfusion. K5-N,OSepi (0.1-1 mg/kg given i.v. 15 min. before reperfusion) significantly reduced the extent of myocardial damage in a dose-dependent manner. Furthermore, we investigated the potential mechanism(s) of the cardioprotective effect(s) afforded by K5-N,OSepi. In left ventricular samples, I/R induced mast cell degranulation and a robust increase in lipid peroxidation, free radical-induced DNA damage and calcium overload. Markers of neutrophil infiltration and activation were also induced by I/R in rat hearts, specifically myeloperoxidase activity, intercellular-adhesion-molecule-1 expression, prostaglandin-E(2) and tumour-necrosis-factor-α production. The robust increase in oxidative stress and inflammatory markers was blunted by K5-N,OSepi, in a dose-dependent manner, with maximum at 1 mg/kg. Furthermore, K5-N,OSepi administration attenuated the increase in caspase 3 activity, Bid and Bax activation and ameliorated the decrease in expression of Bcl-2 within the ischaemic myocardium. In conclusion, we demonstrate that the cardioprotective effect of the non-anticoagulant K5 derivative K5-N,OSepi is secondary to a combination of anti-apoptotic and anti-inflammatory effects.


Asunto(s)
Anticoagulantes/farmacología , Cápsulas Bacterianas/metabolismo , Heparina/farmacología , Daño por Reperfusión Miocárdica/tratamiento farmacológico , 8-Hidroxi-2'-Desoxicoguanosina , Animales , Antiinflamatorios/farmacología , Apoptosis/efectos de los fármacos , Calcio/análisis , Caspasa 3/análisis , Caspasa 3/metabolismo , Desoxiguanosina/análogos & derivados , Desoxiguanosina/análisis , Diagnóstico por Computador/métodos , Dinoprostona/análisis , Dinoprostona/metabolismo , Inflamación/tratamiento farmacológico , Inflamación/patología , Masculino , Daño por Reperfusión Miocárdica/patología , Miocardio/metabolismo , Miocardio/patología , Ratas , Ratas Wistar , Sustancias Reactivas al Ácido Tiobarbitúrico , Factor de Necrosis Tumoral alfa/análisis , Factor de Necrosis Tumoral alfa/metabolismo
9.
Mol Med ; 18: 719-27, 2012 May 09.
Artículo en Inglés | MEDLINE | ID: mdl-22415011

RESUMEN

In preclinical studies, erythropoietin (EPO) reduces ischemia-reperfusion-associated tissue injury (for example, stroke, myocardial infarction, acute kidney injury, hemorrhagic shock and liver ischemia). It has been proposed that the erythropoietic effects of EPO are mediated by the classic EPO receptor homodimer, whereas the tissue-protective effects are mediated by a hetero-complex between the EPO receptor monomer and the ß-common receptor (termed "tissue-protective receptor"). Here, we investigate the effects of a novel, selective-ligand of the tissue-protective receptor (pyroglutamate helix B surface peptide [pHBSP]) in a rodent model of acute kidney injury/dysfunction. Administration of pHBSP (10 µg/kg intraperitoneally [i.p.] 6 h into reperfusion) or EPO (1,000 IU/kg i.p. 4 h into reperfusion) to rats subjected to 30 min ischemia and 48 h reperfusion resulted in significant attenuation of renal and tubular dysfunction. Both pHBSP and EPO enhanced the phosphorylation of Akt (activation) and glycogen synthase kinase 3ß (inhibition) in the rat kidney after ischemia-reperfusion, resulting in prevention of the activation of nuclear factor-κB (reduction in nuclear translocation of p65). Interestingly, the phosphorylation of endothelial nitric oxide synthase was enhanced by EPO and, to a much lesser extent, by pHBSP, suggesting that the signaling pathways activated by EPO and pHBSP may not be identical.


Asunto(s)
Lesión Renal Aguda/tratamiento farmacológico , Oligopéptidos/administración & dosificación , Lesión Renal Aguda/metabolismo , Animales , Clusterina/sangre , Glucógeno Sintasa Quinasa 3/metabolismo , Glucógeno Sintasa Quinasa 3 beta , Glomérulos Renales/efectos de los fármacos , Glomérulos Renales/fisiopatología , Túbulos Renales/efectos de los fármacos , Túbulos Renales/fisiopatología , Masculino , FN-kappa B/metabolismo , Óxido Nítrico Sintasa de Tipo III/metabolismo , Osteopontina/sangre , Transporte de Proteínas/efectos de los fármacos , Proteínas Proto-Oncogénicas c-akt/metabolismo , Ratas , Ratas Wistar , Factores de Tiempo
10.
Inflamm Res ; 61(4): 311-7, 2012 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-22223068

RESUMEN

OBJECTIVE: High levels of both angiotensin (Ang) II and tumor necrosis factor (TNF)-α have been implicated in the pathogenesis of glomerular injury by affecting podocytes. The aim of this study was to investigate the Ang II-TNF-α relationship in human podocytes. METHODS: Immortalized podocytes were exposed to Ang II for 6 days in the absence or presence of either losartan or PD123,319 (both at 100 nM), AT(1) and AT(2) receptor antagonists, respectively. RESULTS: Ang II, after at least 72 h of repeated treatment, increased basal TNFA gene expression and cytokine release with a biphasic pattern and maximum response at 10 nM. Losartan dampened the effects of Ang II on TNF-α production throughout the experimental period, demonstrating an AT(1) receptor contribution. PD123,319 affected the second TNF-α production peak, showing also an AT(2) receptor contribution. Moreover, Ang II causes tumor necrosis factor receptor (TNFR) 1 and TNFR2 over-expression in a time-dependent manner. The functional interaction between Ang II and TNF-α was demonstrated when the pro-proliferative effect of Ang II was antagonized by a neutralizing TNF-α antibody. CONCLUSIONS: Our results show a functional interaction between Ang II and TNF-α and implicate this cytokine as a mediator in Ang II long-term pathoadaptive podocytes changes.


Asunto(s)
Angiotensina II/farmacología , Podocitos/efectos de los fármacos , Factor de Necrosis Tumoral alfa/biosíntesis , Bloqueadores del Receptor Tipo 1 de Angiotensina II/farmacología , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Expresión Génica/efectos de los fármacos , Humanos , Imidazoles/farmacología , Losartán/farmacología , Podocitos/metabolismo , Piridinas/farmacología , Receptor de Angiotensina Tipo 1/metabolismo , Receptor de Angiotensina Tipo 2/metabolismo , Receptores Tipo I de Factores de Necrosis Tumoral/genética , Receptores Tipo I de Factores de Necrosis Tumoral/metabolismo , Receptores Tipo II del Factor de Necrosis Tumoral/genética , Receptores Tipo II del Factor de Necrosis Tumoral/metabolismo , Factor de Necrosis Tumoral alfa/genética
11.
J Immunol ; 185(7): 3970-9, 2010 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-20817864

RESUMEN

Vascular endothelial cells (ECs) are key players in leukocyte recruitment into tissues and metastatic dissemination of tumor cells. ECs express B7h, which is the ligand of the ICOS T cell costimulatory molecule. The aim of this work was to assess the effect of B7h triggering by a soluble form of ICOS (ICOS-Fc) on the adhesion of colon carcinoma cell lines to HUVECs. We found that B7h triggering inhibited HUVEC adhesiveness to HT29 and DLD1 cells (by 50 and 35%, respectively) but not to HCT116 cells. The effect was dependent on the ICOS-Fc dose and was detectable as early as 30 min after treatment and was still present after 24 h. It was inhibited by soluble anti-ICOS reagents (mAb and B7h-Fc) and silencing of B7h on HUVECs, and it was not displayed by an F119S mutated form of ICOS-Fc that does not bind B7h. HUVEC treatment with ICOS-Fc did not modulate expression of adhesion molecules and cytokines, but it substantially downmodulated ERK phosphorylation induced by E-selectin triggering or osteopontin, which may influence HUVEC adhesiveness. Moreover, HUVEC treatment with ICOS-Fc also inhibited adhesion of polymorphonuclear cells and several tumor cell lines from different origins. Therefore, the B7h-ICOS interaction may modulate spreading of cancer metastases and recruitment of polymorphonuclear cells in inflammatory sites, which opens a view on the use of ICOS-Fc as an immunomodulatory drug.


Asunto(s)
Antígenos CD/metabolismo , Antígenos de Diferenciación de Linfocitos T/metabolismo , Adhesión Celular/fisiología , Células Endoteliales/metabolismo , Neutrófilos/metabolismo , Antígenos CD/inmunología , Antígenos de Diferenciación de Linfocitos T/inmunología , Antígeno B7-H1 , Western Blotting , Moléculas de Adhesión Celular/inmunología , Moléculas de Adhesión Celular/metabolismo , Línea Celular Tumoral , Células Endoteliales/inmunología , Humanos , Proteína Coestimuladora de Linfocitos T Inducibles , Neutrófilos/inmunología , Transducción de Señal/fisiología , Cordón Umbilical/metabolismo
12.
Mol Med ; 17(9-10): 883-92, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21607291

RESUMEN

Recent studies have shown that erythropoietin, critical for the differentiation and survival of erythrocytes, has cytoprotective effects in a wide variety of tissues, including the kidney and lung. However, erythropoietin has been shown to have a serious side effect-an increase in thrombovascular effects. We investigated whether pyroglutamate helix B-surface peptide (pHBSP), a nonerythropoietic tissue-protective peptide mimicking the 3D structure of erythropoietin, protects against the organ injury/ dysfunction and inflammation in rats subjected to severe hemorrhagic shock (HS). Mean arterial blood pressure was reduced to 35 ± 5 mmHg for 90 min followed by resuscitation with 20 mL/kg Ringer Lactate for 10 min and 50% of the shed blood for 50 min. Rats were euthanized 4 h after the onset of resuscitation. pHBSP was administered 30 min or 60 min into resuscitation. HS resulted in significant organ injury/dysfunction (renal, hepatic, pancreas, neuromuscular, lung) and inflammation (lung). In rats subjected to HS, pHBSP significantly attenuated (i) organ injury/dysfunction (renal, hepatic, pancreas, neuromuscular, lung) and inflammation (lung), (ii) increased the phosphorylation of Akt, glycogen synthase kinase-3ß and endothelial nitric oxide synthase, (iii) attenuated the activation of nuclear factor (NF)-κB and (iv) attenuated the increase in p38 and extracellular signal-regulated kinase (ERK)1/2 phosphorylation. pHBSP protects against multiple organ injury/dysfunction and inflammation caused by severe hemorrhagic shock by a mechanism that may involve activation of Akt and endothelial nitric oxide synthase, and inhibition of glycogen synthase kinase-3ß and NF-κB.


Asunto(s)
Eritropoyetina/farmacología , Insuficiencia Multiorgánica/prevención & control , Péptidos/farmacología , Neumonía/prevención & control , Choque Hemorrágico/complicaciones , Animales , Materiales Biomiméticos/química , Materiales Biomiméticos/farmacología , Presión Sanguínea/efectos de los fármacos , Western Blotting , Eritropoyetina/química , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Glucógeno Sintasa Quinasa 3/metabolismo , Riñón/efectos de los fármacos , Riñón/metabolismo , Riñón/fisiopatología , Hígado/efectos de los fármacos , Hígado/metabolismo , Hígado/fisiopatología , Masculino , Insuficiencia Multiorgánica/etiología , Insuficiencia Multiorgánica/fisiopatología , FN-kappa B/metabolismo , Óxido Nítrico Sintasa de Tipo III/metabolismo , Fosforilación/efectos de los fármacos , Neumonía/etiología , Neumonía/fisiopatología , Proteínas Proto-Oncogénicas c-akt/metabolismo , Ratas , Ratas Wistar , Resucitación
13.
Am J Respir Crit Care Med ; 182(12): 1506-15, 2010 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-20693380

RESUMEN

RATIONALE: Peroxisome proliferator-activated receptor (PPAR)-ß/δ is a transcription factor that belongs to the PPAR nuclear hormone receptor family, but the role of PPAR-ß/δ in sepsis is unknown. OBJECTIVES: We investigated the role of PPAR-ß/δ in murine models of LPS-induced organ injury and dysfunction and cecal ligation and puncture (CLP)-induced polymicrobial sepsis. METHODS: Wild-type (WT) and PPAR-ß/δ knockout (KO) mice and C57BL/6 mice were subjected to LPS for 16 hours. C57BL/6 mice received the PPAR-ß/δ agonist GW0742 (0.03 mg/kg intravenously, 1 h after LPS) or GW0742 plus the PPAR-ß/δ antagonist GSK0660 (0.1 mg/kg intravenously, 30 min before LPS). CD-1 mice subjected to CLP received GW0742 or GW0742 plus GSK0660. MEASUREMENTS AND MAIN RESULTS: In PPAR-ß/δ KO mice, endotoxemia exacerbated organ injury and dysfunction (cardiac, renal, and hepatic) and inflammation (lung) compared with WT mice. In C57BL/6 mice subjected to endotoxemia, GW0742 significantly (1) attenuated organ (cardiac and renal) dysfunction and inflammation (lung); (2) increased the phosphorylation of Akt and glycogen synthase kinase (GSK)-3ß; (3) attenuated the increase in extracellular signal-regulated kinase (ERK)1/2 and signal transducer and activator of transcription (STAT)-3 phosphorylation; and (4) attenuated the activation of nuclear factor (NF)-κB and the expression of inducible nitric oxide synthase (iNOS). In CD-1 mice subjected to CLP, GW0742 improved 10-day survival. All the observed beneficial effects of GW0742 were attenuated by the PPAR-ß/δ antagonist GSK0660. CONCLUSIONS: PPAR-ß/δ protects against multiple organ injury and dysfunction, and inflammation caused by endotoxic shock and improves survival in polymicrobial sepsis by a mechanism that may involve activation of Akt and inhibition of GSK-3ß and NF-κB.


Asunto(s)
PPAR delta/metabolismo , PPAR-beta/metabolismo , Choque Séptico/prevención & control , Animales , Modelos Animales de Enfermedad , Glucógeno Sintasa Quinasa 3/metabolismo , Glucógeno Sintasa Quinasa 3 beta , Masculino , Ratones , Ratones Endogámicos C57BL , FN-kappa B/metabolismo , Óxido Nítrico Sintasa de Tipo II/biosíntesis , Fosforilación/efectos de los fármacos , Proteínas Proto-Oncogénicas c-akt/metabolismo , Choque Séptico/metabolismo , Transducción de Señal , Sulfonas/farmacología , Tiazoles/farmacología , Tiofenos/farmacología
14.
Proc Natl Acad Sci U S A ; 105(52): 20864-9, 2008 Dec 30.
Artículo en Inglés | MEDLINE | ID: mdl-19106301

RESUMEN

Mutations in oncogenes and tumor suppressor genes are responsible for tumorigenesis and represent favored therapeutic targets in oncology. We exploited homologous recombination to knock-in individual cancer mutations in the genome of nontransformed human cells. Sequential introduction of multiple mutations was also achieved, demonstrating the potential of this strategy to construct tumor progression models. Knock-in cells displayed allele-specific activation of signaling pathways and mutation-specific phenotypes different from those obtainable by ectopic oncogene expression. Profiling of a library of pharmacological agents on the mutated cells showed striking sensitivity or resistance phenotypes to pathway-targeted drugs, often matching those of tumor cells carrying equivalent cancer mutations. Thus, knock-in of single or multiple cancer alleles provides a pharmacogenomic platform for the rational design of targeted therapies.


Asunto(s)
Alelos , Antineoplásicos/farmacología , Regulación Neoplásica de la Expresión Génica/genética , Técnicas de Sustitución del Gen , Genes Supresores de Tumor , Mutación , Neoplasias/tratamiento farmacológico , Neoplasias/genética , Antineoplásicos/uso terapéutico , Línea Celular , Sistemas de Liberación de Medicamentos/métodos , Ensayos de Selección de Medicamentos Antitumorales , Humanos , Modelos Biológicos
15.
Br J Pharmacol ; 177(3): 503-515, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-30741418

RESUMEN

Starting with a role for histamine role in renal haemodynamics, evidence has accumulated, over time, suggesting a wider range of actions on renal function and this has renewed interest in the pathophysiological role of histamine in the kidney. Here we provide an up-to-date review of this topic. As the kidney expresses enzymes that synthesize and metabolise histamine, along with its receptors, all the components for histaminergic transmission are present in this tissue. The distribution of histamine receptors matches a wide range of effects. We address the questions of the redundancy of H1 and H2 receptors in renal haemodynamics, the complementary role of H1 and H4 receptors in renal filtration and reabsorption, and the dichotomy between local and neuronal H1 and H3 receptors. Experimental models of renal disease raise the possibility of new therapeutic approaches based on histamine. The effects of histamine on renal function are not yet fully understood and their elucidation is still ongoing. LINKED ARTICLES: This article is part of a themed section on New Uses for 21st Century. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v177.3/issuetoc.


Asunto(s)
Histamina , Receptores Histamínicos , Animales , Hemodinámica , Humanos , Riñón
16.
Neurosci Lett ; 454(2): 134-8, 2009 Apr 24.
Artículo en Inglés | MEDLINE | ID: mdl-19429070

RESUMEN

Several evidences indicate that PPARgamma stimulation promotes neuronal differentiation. However, to date, no data describe the effects of PPARgamma agonists on neurite outgrowth. Here we have evaluated the effects of pioglitazone, a synthetic PPARgamma agonist, on differentiation and neurite outgrowth in SH-SY5Y human neuroblastoma cells. Our results show that pioglitazone promotes cell differentiation and the outgrowth of cell processes in a concentration-dependent manner with the maximal effect at 100 nM-1 microM. It significantly increases both the mean process length and the percentage of neurite-bearing cells. In addition, these effects are accompanied by significant activation of p42 and p44 mitogen-activated protein kinases. In conclusion, albeit preliminary, these findings suggest the possibility that PPARgamma stimulation may contribute to the development and maintenance of a proper neuronal connectivity within neuronal networks.


Asunto(s)
Neuritas/fisiología , Neurogénesis/efectos de los fármacos , PPAR gamma/metabolismo , Tiazolidinedionas/farmacología , Análisis de Varianza , Western Blotting , Línea Celular Tumoral , Relación Dosis-Respuesta a Droga , Activación Enzimática , Humanos , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Neuritas/efectos de los fármacos , PPAR gamma/agonistas , Pioglitazona
18.
Lab Invest ; 88(10): 1038-48, 2008 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-18679378

RESUMEN

The generation of endogenous hydrogen sulfide may either limit or contribute to the degree of tissue injury caused by ischemia/reperfusion. A total of 74 male Wistar rats were used to investigate the effects of endogenous and exogenous hydrogen sulfide in renal ischemia/reperfusion. Administration of the irreversible cystathionine gamma-lyase (CSE) inhibitor, dL-propargylglycine, prevented the recovery of renal function after 45 min ischemia and 72 h reperfusion. The hydrogen sulfide donor sodium hydrosulfide attenuated the (renal, tubular, and glomerular) dysfunction and injury caused by 45 min ischemia and 6 h reperfusion. Western blot analysis of kidneys taken at 30 min reperfusion showed that sodium hydrosulfide significantly attenuated phosphorylation of mitogen-activated protein kinases (p-38, c-JUN N-terminal protein kinase 1/2, and extracellular signal-regulated kinase 1/2) and activation of nuclear factor-kappaB. At 6 h reperfusion, sodium hydrosulfide significantly attenuated the histological score for acute tubular necrosis, the activation of caspase-3 and Bid, the decline in the expression of anti-apoptotic Bcl-2, and the expression of nuclear factor-kappaB-dependent proteins (inducible nitric oxide synthase, cyclo-oxygenase-2, and intercellular adhesion molecule-1). These findings suggest that (1) the synthesis of endogenous hydrogen sulfide by CSE is essential to protect the kidney against ischemia/reperfusion injury and dysfunction and aids in the recovery of renal function following ischemia/reperfusion, (2) hydrogen sulfide generated by sodium hydrosulfide reduces ischemia/reperfusion injury and dysfunction, and morphological changes of the kidney, and (3) the observed protective effects of hydrogen sulfide are due to both anti-apoptotic and anti-inflammatory effects.


Asunto(s)
Cistationina gamma-Liasa/metabolismo , Sulfuro de Hidrógeno/metabolismo , Riñón/fisiopatología , Daño por Reperfusión/fisiopatología , Animales , Antiinflamatorios/metabolismo , Apoptosis/fisiología , Modelos Animales de Enfermedad , Riñón/patología , Masculino , Ratas , Ratas Wistar , Recuperación de la Función/fisiología , Daño por Reperfusión/patología
19.
Crit Care Med ; 36(2): 550-9, 2008 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-18216604

RESUMEN

OBJECTIVE: Sphingosylphosphorylcholine (SPC) has been reported to activate a variety of G-protein coupled receptors, including S1P(1-5), G2A, GPR4, and OGR1 (GPR68). Interestingly, other structurally related lysophospholipid agonists of these receptors have been shown to exhibit immunomodulatory properties both in vitro and in vivo. These include prevention of tumor necrosis factor-alpha-induced monocyte adhesion to aortic endothelium in mice (sphingosine-1-phosphate via S1P(1-5) receptors) and reduction of organ injury and/or mortality in animal models of sepsis and endotoxemia (lysophosphatidylcholine via G2A). Here, we investigate the effects of SPC on the organ injury/dysfunction caused by systemic administration of lipopolysaccharide and the mechanisms underlying the observed effects of SPC. DESIGN: Prospective, randomized study. SETTING: University-based research laboratory. SUBJECTS: Sixty-one anesthetized male Wistar rats. INTERVENTIONS: Rats received either SPC (10 mg/kg intravenously) or vehicle (phosphate-buffered saline 1 mL/kg intravenously) 15 mins before or 15 mins after induction of endotoxemia with lipopolysaccharide (6 mg/kg intravenously). MEASUREMENTS AND MAIN RESULTS: Treatment with SPC significantly reduced the organ/dysfunction injury caused by lipopolysaccharide. SPC pretreatment significantly reduced the circulating levels of interleukin-1beta and interleukin-6, the expression of CD11b (ligand for intercellular adhesion molecule-1) on circulating polymorphonuclear cells, the expression of proteins of intercellular adhesion molecule-1 (Western blot and immunohistochemistry), cyclooxygenase-2 and nuclear translocation of nuclear factor-kappaB (Western blot analysis), and inducible nitric oxide synthase (immunohistochemistry) as well as the lung injury caused by endotoxemia in the rat. CONCLUSIONS: SPC reduced the organ injury/dysfunction caused by endotoxin in the rat. These beneficial effects of SPC are associated with potent anti-inflammatory effects.


Asunto(s)
Endotoxemia/metabolismo , Endotoxemia/patología , Fosforilcolina/análogos & derivados , Esfingosina/análogos & derivados , Animales , Apoptosis/fisiología , Ciclooxigenasa 2/metabolismo , Citocinas/metabolismo , Esquema de Medicación , Endotoxemia/tratamiento farmacológico , Molécula 1 de Adhesión Intercelular/metabolismo , Pulmón/enzimología , Pulmón/patología , Masculino , FN-kappa B/metabolismo , Óxido Nítrico Sintasa de Tipo II/metabolismo , Peroxidasa/metabolismo , Fosforilcolina/uso terapéutico , Ratas , Ratas Wistar , Esfingosina/uso terapéutico
20.
Eur J Hosp Pharm ; 25(6): 301-309, 2018 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-31157047

RESUMEN

OBJECTIVES: (1) To evaluate drug-drug interactions (DDIs) in general practitioners' (GPs) prescriptions; (2) to implement a cooperation project between pharmacists and GPs to improve DDI management and patient care. METHODS: In 2013, pharmacists from the Community Drug Assistance ASL TO1 launched a cooperation project involving 48 GPs. As a first step, GPs were asked to select, from a list, drug associations for which they recommended analysis of occurrence in their prescriptions. The pharmacists (1) analysed GPs' prescriptions dated 2012-2014, according to the list of DDIs selected (n= 9); (2) evaluated solutions for DDI management, using the Micromedex DDI checker database and literature analysis; they then (3) disseminated DDI-related information to GPs through training meetings and (4) assessed the efficacy of these actions through a questionnaire submitted to the GPs in 2013. RESULTS: (1) Prescriptions analysis: a reduction in the number of DDIs was observed (-14% in 2013 vs 2012, -9% in 2014 vs 2012); in some cases these reductions were statistically significant (calcium carbonate + proton pump inhibitors (PPIs) -50%, p<0.0041, amoxicillin+lansoprazole -42%, p<0.0088). (2) Questionnaire: this was completed by 75% of GPs. The literature analysis was considered interesting by 94% of GPs; solutions were adopted by 89% of GPs and 34% of GPs affirmed that clinical improvements after application of the measures were observed in their patients, even if they could not provide quantitative data for this outcome. CONCLUSION: The cooperation project between pharmacists and GPs was effective because it established a professional exchange between the two health professionals. The pharmacist gave support to GPs, which benefited the patients, who gained clinical improvements and improved satisfaction with their medical care, as declared by the GPs in answers to the questionnaire.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA