Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Histopathology ; 70(3): 492-498, 2017 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-27391928

RESUMEN

AIMS: To investigate the status of somatostatin receptors (SSTRs) in resected hepatocellular carcinoma (HCC). METHODS AND RESULTS: Transcript and protein levels of SSTR2, SSTR3 and SSTR5 were investigated, with real-time polymerase chain reaction (PCR) and manual and automated immunohistochemistry (IHC), in 53 resected HCCs and paired non-tumour tissues. SSTR1, SSTR4, SSTR5TMD4 and SSTR5TMD5 were analysed with real-time PCR. SSTR3 and SSTR5 transcripts were expressed in ~25% of HCCs, but not in adjacent non-tumour tissues. SSTR1 and SSTR2 transcripts were overexpressed in 42% and 32% of HCCs, respectively. SSTR4, SSTR5TMD4 and SSTR5TMD5 were not detected. Membrane staining for SSTR2 was detected in 38% of HCCs, whereas SSTR5 protein was detectable in only 11% of HCCs. SSTR3 protein was detected in the majority of HCCs and adjacent non-tumour liver tissues, but membrane staining was <20% of that in HCCs. The results obtained with the two IHC methods were highly correlated (P < 0.0001). Statistical analyses also showed a positive correlation between SSTR2 membrane staining and cytokeratin 19 expression (P = 0.04), serum α-fetoprotein level (P = 0.002), and poor differentiation (P = 0.05). CONCLUSIONS: Membrane SSTR2 is detected reliably in HCCs by IHC, and is a potential therapeutic target, as it is coexpressed with markers of poor prognosis.


Asunto(s)
Carcinoma Hepatocelular/patología , Neoplasias Hepáticas/patología , Receptores de Somatostatina/biosíntesis , Biomarcadores de Tumor/análisis , Carcinoma Hepatocelular/mortalidad , Femenino , Humanos , Inmunohistoquímica , Neoplasias Hepáticas/mortalidad , Masculino , Persona de Mediana Edad , Pronóstico , Reacción en Cadena en Tiempo Real de la Polimerasa , Receptores de Somatostatina/análisis
2.
Bioorg Med Chem Lett ; 22(18): 5909-14, 2012 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-22901389

RESUMEN

Indole-pyrrolidines were identified as inhibitors of 11ß-hydroxysteroid dehydrogenase type 1 (11ß-HSD1) by high-throughput screening. Optimisation of the initial hit through structure-based design led to 7-azaindole-derivatives, with the best analogues displaying single digit nanomolar IC(50) potency. The modeling hypotheses were confirmed by solving the X-ray co-crystal structure of one of the lead compounds. These compounds were selective against 11ß-hydroxysteroid dehydrogenase type 2 (selectivity ratio >200) and exhibited good inhibition of 11ß-HSD1 (IC(50)<1µM) in a cellular model (3T3L1 adipocytes).


Asunto(s)
11-beta-Hidroxiesteroide Deshidrogenasa de Tipo 1/antagonistas & inhibidores , Amidas/farmacología , Diseño de Fármacos , Inhibidores Enzimáticos/farmacología , Indoles/química , Pirrolidinas/química , 11-beta-Hidroxiesteroide Deshidrogenasa de Tipo 1/metabolismo , Amidas/síntesis química , Amidas/química , Animales , Relación Dosis-Respuesta a Droga , Inhibidores Enzimáticos/síntesis química , Inhibidores Enzimáticos/química , Humanos , Ratones , Modelos Moleculares , Estructura Molecular , Relación Estructura-Actividad
3.
Bioorg Med Chem Lett ; 19(10): 2674-8, 2009 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-19395260

RESUMEN

Benzylamides of pentanedioic acid were identified as inhibitors of 11beta-hydroxysteroid dehydrogenase type 1 (11beta-HSD1) by high-throughput screening. Optimisation to 2-adamantyl amides yielded inhibitors with single digit nanomolar IC(50)s on the 11beta-HSD1 human isoform. The hydroxy adamantyl amide lead compound was selective against 11beta-hydroxysteroid dehydrogenase type 2 (selectivity ratio >1000) and displayed good inhibition of 11beta-HSD1 (IC(50)<0.1microM) in a cellular model (3T3L1 adipocytes).


Asunto(s)
11-beta-Hidroxiesteroide Deshidrogenasa de Tipo 1/antagonistas & inhibidores , Amidas/química , Inhibidores Enzimáticos/química , 11-beta-Hidroxiesteroide Deshidrogenasa de Tipo 1/metabolismo , 11-beta-Hidroxiesteroide Deshidrogenasa de Tipo 2/antagonistas & inhibidores , 11-beta-Hidroxiesteroide Deshidrogenasa de Tipo 2/metabolismo , Células 3T3-L1 , Amidas/síntesis química , Amidas/farmacología , Animales , Descubrimiento de Drogas , Inhibidores Enzimáticos/síntesis química , Inhibidores Enzimáticos/farmacología , Humanos , Ratones , Ratas , Relación Estructura-Actividad
4.
Bioorg Med Chem Lett ; 19(13): 3682-5, 2009 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-19450980

RESUMEN

Spiro-carboxamides were identified as inhibitors of 11beta-hydroxysteroid-dehydrogenase type 1 by high-throughput screening. Structure-based drug design was used to optimise the initial hit yielding a sub-nanomolar IC(50) inhibitor (0.5nM) on human 11beta-HSD1 with a high binding efficiency index (BEI of 32.7) which was selective against human 11beta-HSD2 (selectivity ratio>200000).


Asunto(s)
11-beta-Hidroxiesteroide Deshidrogenasa de Tipo 1/antagonistas & inhibidores , Amidas/química , Inhibidores Enzimáticos/química , Compuestos de Espiro/química , 11-beta-Hidroxiesteroide Deshidrogenasa de Tipo 1/metabolismo , Amidas/farmacología , Sitios de Unión , Simulación por Computador , Diseño de Fármacos , Inhibidores Enzimáticos/farmacología , Humanos , Compuestos de Espiro/farmacología , Relación Estructura-Actividad
5.
Clin Exp Pharmacol Physiol ; 35(5-6): 601-5, 2008 May.
Artículo en Inglés | MEDLINE | ID: mdl-18177479

RESUMEN

1. It was proposed previously that oxidative stress is a main component of the inflammatory process in chronic obstructive pulmonary disease (COPD). Thus, in the present study, we investigated the inflammatory response in mice deficient for the p47(phox) subunit of NADPH oxidase (p47 KO) exposed to cigarette smoke (CS). 2. Exposure of mice to CS elicited an increase in the number of macrophages and neutrophils and levels of interleukin (IL)-6, keratinocyte-derived chemokine (KC/CXCL1) and monocyte chemoattractant protein-1 (MCP1/CCL2) in bronchoalveolar lavage fluid (BALF), which were lower in p47 KO mice compared with control mice. In contrast, 24 h after lipopolysaccharide (LPS) exposure, the number of macrophages and neutrophils, as well as KC/CXCL1 levels, in BALF was significantly greater in p47 KO mice compared with control mice. 3. The present study has shown that airway inflammation is decreased in p47 KO mice after exposure to CS, but not LPS, suggesting that oxidative stress is involved in the pathogenesis of airway inflammation associated with COPD.


Asunto(s)
Inflamación/inducido químicamente , Lipopolisacáridos/efectos adversos , Pulmón/efectos de los fármacos , Nicotiana , Estrés Oxidativo/efectos de los fármacos , Humo/efectos adversos , Animales , Líquido del Lavado Bronquioalveolar/química , Líquido del Lavado Bronquioalveolar/citología , Citocinas/análisis , Citocinas/metabolismo , Pulmón/patología , Ratones , Ratones Noqueados , NADPH Oxidasas/genética , NADPH Oxidasas/metabolismo
6.
Oncoimmunology ; 5(6): e1145333, 2016 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-27471612

RESUMEN

The infiltration of myeloid cells helps tumors to overcome immune surveillance and imparts resistance to cancer immunotherapy. Thus, strategies to modulate the effects of these immune cells may offer a potential therapeutic benefit. We report here that tasquinimod, a novel immunotherapy which targets S100A9 signaling, reduces the immunosuppressive properties of myeloid cells in preclinical models of bladder cancer (BCa). As single anticancer agent, tasquinimod treatment was effective in preventing early stage tumor growth, but did not achieve a clear antitumor effect in advanced tumors. Investigations of this response revealed that tasquinimod induces an increase in the expression of a negative regulator of T cell activation, Programmed-death-ligand 1 (PD-L1). This markedly weakens its antitumor immunity, yet provokes an "inflamed" milieu rendering tumors more prone to T cell-mediated immune attack by PD-L1 blockade. Interestingly, the combination of tasquinimod with an Anti-PD-L1 antibody enhanced the antitumor immune response in bladder tumors. This combination synergistically modulated tumor-infiltrating myeloid cells, thereby strongly affecting proliferation and activation of effector T cells. Together, our data provide insight into the rational combination of therapies that activate both innate and adaptive immune system, such as the association of S100A9-targeting agents with immune checkpoints inhibitors, to improve the response to cancer immunotherapeutic agents in BCa.

7.
Pancreas ; 45(10): 1386-1393, 2016 11.
Artículo en Inglés | MEDLINE | ID: mdl-27622342

RESUMEN

OBJECTIVE: Somatostatin receptors (SSTRs), products of gene superfamily SSTR1-5, are commonly expressed in neuroendocrine tumors (NETs). Somatostatin analogs (SSAs) bind to SSTRs and are used as therapeutic agents in patients with advanced NETs. We hypothesized that tumor SSTR expression status would be associated with clinical outcomes in NET. METHODS: Expression of the 5 SSTRs was evaluated by immunohistochemistry, using tissue microarrays comprising 173 primary NETs, 24 matched metastases, and 22 metastatic NETs from 195 patients. Cox proportional hazards regression analysis was used to assess the association of SSTR expression status (high vs low) with clinical outcomes, adjusting for potential confounders. RESULTS: High expression of SSTR2 was associated with longer overall survival (OS) in the cohort overall (multivariate hazard ratio, 0.42; 95% confidence interval, 0.21-0.84; P = 0.013). In a subgroup of patients with metastatic small intestine NET treated with SSAs and evaluable for progression, SSTR2 expression was associated with both longer progression-free survival (PFS) and OS. No associations with PFS or OS were observed with expression of other SSTRs. CONCLUSIONS: Our study demonstrated that expression of SSTR2, but not other SSTRs, is associated with longer OS. In patients treated with SSAs, expression of SSTR2 is associated with longer PFS survival.


Asunto(s)
Tumores Neuroendocrinos , Supervivencia sin Enfermedad , Expresión Génica , Humanos , Receptores de Somatostatina , Somatostatina
8.
J Immunother Cancer ; 3: 53, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26673090

RESUMEN

BACKGROUND: Tasquinimod (a quinoline-3-carboxyamide) is a small molecule immunotherapy with demonstrated effects on the tumor microenvironment (TME) involving immunomodulation, anti-angiogenesis and inhibition of metastasis. A target molecule of tasquinimod is the inflammatory protein S100A9 which has been shown to affect the accumulation and function of suppressive myeloid cell subsets in tumors. Given the major impact of myeloid cells to the tumor microenvironment, manipulation of this cell compartment is a desirable goal in cancer therapeutics. METHODS: To understand the consequences of tasquinimod treatment on the TME, we evaluated early treatment effects in tumor infiltrating myeloid cells. Cellular phenotypes were studied by flow cytometry while gene expression both in tumor tissue and in isolated CD11b(+) cells or tumor cells were measured by real time-PCR. Effects on angiogenesis were monitored by changes in CD31 levels and by gene expression in tumor tissue. Effects on cytokine levels in tumor tissue and serum were determined by multiplex analysis. RESULTS: The MC38-C215 colon carcinoma tumors showed a substantial infiltration of primarily myeloid cells that were dominated by Ly6C(low)F4/80(+)CD206(+) M2-polarized tumor associated macrophages (TAMs), an immuno-suppressive and pro-angiogenic cell population. Here, we show that tasquinimod treatment induces an anti-tumor effect which is subsequent to a reduction in tumor infiltrating CD206(+) M2 macrophages and a simultaneous increase in M1 macrophages expressing MHC class II and CD86. The tasquinimod-induced changes in TAM polarization were evident within 24 h of exposure, emphasizing the ability of tasquinimod to rapidly reprogram the tumor microenvironment. This change in the tumor associated myeloid compartment preceded an increased IL12-production within the tumor and a decrease in tumor neovascularization. The switch in TAM polarization by tasquinimod was confirmed in the 4T1 breast cancer model where tasquinimod also reduce lung metastasis development. CONCLUSION: Our data show that tasquinimod affects tumor infiltrating myeloid cells early after exposure, leading to a change in phenotype from pro-angiogenic and immunosuppressive M2-like TAMs to pro-inflammatory M1-like macrophages. These changes are consistent with the effects of tasquinimod seen on tumor vascularization, immune suppression and metastasis giving further insights to the anti-tumor mechanism of action of tasquinimod.

9.
J Steroid Biochem Mol Biol ; 152: 89-100, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-25958048

RESUMEN

High plasma exposure to estrogens is often associated with prostate cancer. Reducing this phenomenon may present therapeutic benefits. The involvement of estrone sulphate (E1S), the most abundant circulating estrogen in men, has been partially studied in this age-related pathology. To investigate the consequences of plasma E1S overload on blood and prostate sex steroid levels and inflammatory tissue responses, young and middle-aged male rats were treated with E1S with or without steroid sulfatase (STS) inhibitor STX64 for 21 consecutive days. A plasma and prostate tissue steroid profile was determined. STS activity, mRNA expression of E1S organic anion transporting polypeptides (slco1a2, slco2b1, slco4a1) and pro-inflammatory cytokines (Il1-beta, Il6, TNF-alpha) were evaluated in prostate tissue according to age and treatment group. A significant correlation between plasma and prostate steroid levels related to hormone treatment was observed in all rat age groups. However, while the E1S level in prostate tissue increased in middle-aged treated rats (p<0.0001), no significant variation was observed in young treated rats. The protective effect of STX64 during E1S infusion was observed by the maintenance of low free estrogen concentrations in both plasma and tissue. However, this protection was not associated with mRNA expression stability of pro-inflammatory cytokines in older rat prostate. These results suggest that E1S uptake in rat prostate cells increases during aging. Therefore, if a similar phenomenon existed in men, preventively reducing the STS activity could be of interest to limit uptake of estrogens in prostate when high E1S plasma level is assayed.


Asunto(s)
Estrógenos/sangre , Estrona/análogos & derivados , Próstata/metabolismo , Esteril-Sulfatasa/antagonistas & inhibidores , Ácidos Sulfónicos/farmacología , Factores de Edad , Animales , Antiportadores/biosíntesis , Antiportadores/genética , Transporte Biológico , Citocinas/biosíntesis , Citocinas/genética , Estrógenos/farmacología , Estrógenos Conjugados (USP)/farmacología , Estrona/metabolismo , Estrona/farmacología , Proteínas del Ojo/biosíntesis , Proteínas del Ojo/genética , Masculino , Modelos Animales , Transportadores de Anión Orgánico/biosíntesis , Transportadores de Anión Orgánico/genética , ARN Mensajero/biosíntesis , Ratas , Ratas Sprague-Dawley
10.
Endocr Relat Cancer ; 20(1): 53-64, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23132791

RESUMEN

17ß-Hydroxysteroid dehydrogenases (17ß-HSDs) catalyse the 17-position reduction/oxidation of steroids. 17ß-HSD type 3 (17ß-HSD3) catalyses the reduction of the weakly androgenic androstenedione (adione) to testosterone, suggesting that specific inhibitors of 17ß-HSD3 may have a role in the treatment of hormone-dependent prostate cancer and benign prostate hyperplasia. STX2171 is a novel selective non-steroidal 17ß-HSD3 inhibitor with an IC(50) of ∼200 nM in a whole-cell assay. It inhibits adione-stimulated proliferation of 17ß-HSD3-expressing androgen receptor-positive LNCaP(HSD3) prostate cancer cells in vitro. An androgen-stimulated LNCaP(HSD3) xenograft proof-of-concept model was developed to study the efficacies of STX2171 and a more established 17ß-HSD3 inhibitor, STX1383 (SCH-451659, Schering-Plough), in vivo. Castrated male MF-1 mice were inoculated s.c. with 1×10(7) cells 24 h after an initial daily dose of testosterone propionate (TP) or vehicle. After 4 weeks, tumours had not developed in vehicle-dosed mice, but were present in 50% of those mice given TP. One week after switching the stimulus to adione, mice were dosed additionally with the vehicle or inhibitor for a further 4 weeks. Both TP and adione efficiently stimulated tumour growth and increased plasma testosterone levels; however, in the presence of either 17ß-HSD3 inhibitor, adione-dependent tumour growth was significantly inhibited and plasma testosterone levels reduced. Mouse body weights were unaffected. Both inhibitors also significantly lowered plasma testosterone levels in intact mice. In conclusion, STX2171 and STX1383 significantly lower plasma testosterone levels and inhibit androgen-dependent tumour growth in vivo, indicating that 17ß-HSD3 inhibitors may have application in the treatment of hormone-dependent prostate cancer.


Asunto(s)
17-Hidroxiesteroide Deshidrogenasas/antagonistas & inhibidores , Benzazepinas/farmacología , Inhibidores Enzimáticos/farmacología , Neoplasias Hormono-Dependientes/tratamiento farmacológico , Neoplasias de la Próstata/tratamiento farmacológico , Testosterona/sangre , 17-Hidroxiesteroide Deshidrogenasas/genética , Animales , Apoptosis , Benzazepinas/química , Western Blotting , Castración , Proliferación Celular , Inhibidores Enzimáticos/química , Humanos , Masculino , Ratones , Ratones Desnudos , Neoplasias Hormono-Dependientes/enzimología , Neoplasias Hormono-Dependientes/patología , Neoplasias de la Próstata/enzimología , Neoplasias de la Próstata/patología , ARN Mensajero/genética , Radioinmunoensayo , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Carga Tumoral , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto
11.
PLoS One ; 4(9): e6963, 2009 Sep 09.
Artículo en Inglés | MEDLINE | ID: mdl-19742300

RESUMEN

BACKGROUND: Inflammation followed by fibrosis is a component of islet dysfunction in both rodent and human type 2 diabetes. Because islet inflammation may originate from endothelial cells, we assessed the expression of selected genes involved in endothelial cell activation in islets from a spontaneous model of type 2 diabetes, the Goto-Kakizaki (GK) rat. We also examined islet endotheliuml/oxidative stress (OS)/inflammation-related gene expression, islet vascularization and fibrosis after treatment with the interleukin-1 (IL-1) receptor antagonist (IL-1Ra). METHODOLOGY/PRINCIPAL FINDINGS: Gene expression was analyzed by quantitative RT-PCR on islets isolated from 10-week-old diabetic GK and control Wistar rats. Furthermore, GK rats were treated s.c twice daily with IL-1Ra (Kineret, Amgen, 100 mg/kg/day) or saline, from 4 weeks of age onwards (onset of diabetes). Four weeks later, islet gene analysis and pancreas immunochemistry were performed. Thirty-two genes were selected encoding molecules involved in endothelial cell activation, particularly fibrinolysis, vascular tone, OS, angiogenesis and also inflammation. All genes except those encoding angiotensinogen and epoxide hydrolase (that were decreased), and 12-lipoxygenase and vascular endothelial growth factor (that showed no change), were significantly up-regulated in GK islets. After IL-1Ra treatment of GK rats in vivo, most selected genes implied in endothelium/OS/immune cells/fibrosis were significantly down-regulated. IL-1Ra also improved islet vascularization, reduced fibrosis and ameliorated glycemia. CONCLUSIONS/SIGNIFICANCE: GK rat islets have increased mRNA expression of markers of early islet endothelial cell activation, possibly triggered by several metabolic factors, and also some defense mechanisms. The beneficial effect of IL-1Ra on most islet endothelial/OS/immune cells/fibrosis parameters analyzed highlights a major endothelial-related role for IL-1 in GK islet alterations. Thus, metabolically-altered islet endothelium might affect the beta-cell microenvironment and contribute to progressive type 2 diabetic beta-cell dysfunction in GK rats. Counteracting islet endothelial cell inflammation might be one way to ameliorate/prevent beta-cell dysfunction in type 2 diabetes.


Asunto(s)
Diabetes Mellitus Experimental/metabolismo , Diabetes Mellitus Tipo 2/metabolismo , Células Endoteliales/citología , Regulación de la Expresión Génica , Proteína Antagonista del Receptor de Interleucina 1/metabolismo , Islotes Pancreáticos/citología , Animales , Antioxidantes/metabolismo , Inflamación , Interleucina-1/metabolismo , Masculino , Modelos Biológicos , Estrés Oxidativo , Ratas , Ratas Wistar
12.
J Biol Chem ; 280(16): 16076-87, 2005 Apr 22.
Artículo en Inglés | MEDLINE | ID: mdl-15708858

RESUMEN

Mechanisms that arrest G-protein-coupled receptor (GPCR) signaling prevent uncontrolled stimulation that could cause disease. Although uncoupling from heterotrimeric G-proteins, which transiently arrests signaling, is well described, little is known about the mechanisms that permanently arrest signaling. Here we reported on the mechanisms that terminate signaling by protease-activated receptor 2 (PAR(2)), which mediated the proinflammatory and nociceptive actions of proteases. Given its irreversible mechanism of proteolytic activation, PAR(2) is a model to study the permanent arrest of GPCR signaling. By immunoprecipitation and immunoblotting, we observed that activated PAR(2) was mono-ubiquitinated. Immunofluorescence indicated that activated PAR(2) translocated from the plasma membrane to early endosomes and lysosomes where it was degraded, as determined by immunoblotting. Mutant PAR(2) lacking intracellular lysine residues (PAR(2)Delta14K/R) was expressed at the plasma membrane and signaled normally but was not ubiquitinated. Activated PAR(2) Delta14K/R internalized but was retained in early endosomes and avoided lysosomal degradation. Activation of wild type PAR(2) stimulated tyrosine phosphorylation of the ubiquitin-protein isopeptide ligase c-Cbl and promoted its interaction with PAR(2) at the plasma membrane and in endosomes in an Src-dependent manner. Dominant negative c-Cbl lacking the ring finger domain inhibited PAR(2) ubiquitination and induced retention in early endosomes, thereby impeding lysosomal degradation. Although wild type PAR(2) was degraded, and recovery of agonist responses required synthesis of new receptors, lysine mutation and dominant negative c-Cbl impeded receptor ubiquitination and degradation and allowed PAR(2) to recycle and continue to signal. Thus, c-Cbl mediated ubiquitination and lysosomal degradation of PAR(2) to irrevocably terminate signaling by this and perhaps other GPCRs.


Asunto(s)
Proteínas Proto-Oncogénicas/metabolismo , Receptor PAR-2/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Ubiquitina/metabolismo , Regulación hacia Abajo , Técnica del Anticuerpo Fluorescente , Humanos , Lisosomas/metabolismo , Estructura Secundaria de Proteína , Proteínas Proto-Oncogénicas/genética , Proteínas Proto-Oncogénicas c-cbl , Receptor PAR-2/agonistas , Transducción de Señal/genética , Transducción de Señal/fisiología , Ubiquitina-Proteína Ligasas/genética
13.
Am J Physiol Cell Physiol ; 284(5): C1319-29, 2003 May.
Artículo en Inglés | MEDLINE | ID: mdl-12540381

RESUMEN

We evaluated the contribution of rab5a and rab11a to trafficking and signaling of protease-activated receptor 2 (PAR2), a receptor for trypsin and tryptase. Agonists stimulated internalization of PAR2 into early endosomes containing rab5a. Dominant negative rab5aS34N disrupted early endosomes and inhibited agonist-stimulated endocytosis of PAR2. Internalized PAR2 was sorted to lysosomes, and rab5a remained in early endosomes. Rab5a promoted and rab5aS34N impeded resensitization of trypsin-induced calcium mobilization. Rab11a was detected in the Golgi apparatus with PAR2, and PAR2 agonists stimulated redistribution of rab11a into vesicles containing PAR2 that migrated to the cell surface. Dominant negative rab11aS25N was mostly confined to the Golgi apparatus. Although expression of rab11aS25N caused retention of PAR2 in the Golgi apparatus, it did not abolish trafficking of PAR2 to the cell surface. However, expression of wild-type rab11a accelerated both recovery of PAR2 at the cell surface and resensitization of PAR2 signaling. Thus rab5a is required for PAR2 endocytosis and resensitization, whereas rab11a contributes to trafficking of PAR2 from the Golgi apparatus to the plasma membrane.


Asunto(s)
Receptores de Trombina/agonistas , Receptores de Trombina/metabolismo , Proteínas de Unión al GTP rab/fisiología , Proteínas de Unión al GTP rab5/fisiología , Animales , Línea Celular , Endocitosis/fisiología , Humanos , Transporte de Proteínas/fisiología , Ratas , Receptor PAR-2 , Transducción de Señal/fisiología , Distribución Tisular
14.
Am J Physiol Cell Physiol ; 285(4): C945-58, 2003 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-12958028

RESUMEN

Tachykinins interact with three neurokinin receptors (NKRs) that are often coexpressed by the same cell. Cellular responses to tachykinins depend on the NKR subtype that is activated. We compared the colocalization of NK1R and NK3R with beta-arrestins 1 and 2, which play major roles in receptor desensitization, endocytosis, and signaling. In cells expressing NK1R, the selective agonist Sar-Met-substance P induced rapid translocation of beta-arrestins 1 and 2 from the cytosol to the plasma membrane and then endosomes, indicative of interaction with both isoforms. In contrast, the NK3R interacted transiently with only beta-arrestin 2 at the plasma membrane. Despite these differences, both NK1R and NK3R similarly desensitized, internalized, and activated MAP kinases. Because interactions with beta-arrestins can explain differences in the rate of receptor resensitization, we compared resensitization of agonist-induced Ca2+ mobilization. The NK1R resensitized greater than twofold more slowly than the NK3R. Replacement of intracellular loop 3 and the COOH tail of the NK1R with comparable domains of the NK3R diminished colocalization of the NK1R with beta-arrestin 1 and accelerated resensitization to that of the NK3R. Thus loop 3 and the COOH tail specify colocalization of the NK1R with beta-arrestin 1 and determine the rate of resensitization.


Asunto(s)
Arrestinas/metabolismo , Neuroquinina B/análogos & derivados , Receptores de Neuroquinina-1/química , Receptores de Neuroquinina-1/metabolismo , Receptores de Neuroquinina-3/química , Receptores de Neuroquinina-3/metabolismo , Sustancia P/análogos & derivados , Animales , Transporte Biológico/efectos de los fármacos , Calcio/metabolismo , Señalización del Calcio , Línea Celular , Activación Enzimática/fisiología , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Neuroquinina B/farmacología , Estructura Terciaria de Proteína/fisiología , Ratas , Receptores de Neuroquinina-1/agonistas , Receptores de Neuroquinina-3/agonistas , Receptores de Neuroquinina-3/antagonistas & inhibidores , Sustancia P/farmacología , Distribución Tisular , beta-Arrestina 1 , Arrestina beta 2 , beta-Arrestinas
15.
Proc Natl Acad Sci U S A ; 99(5): 3324-9, 2002 Mar 05.
Artículo en Inglés | MEDLINE | ID: mdl-11880656

RESUMEN

Cells express multiple G protein-coupled receptors that are simultaneously or sequentially activated by agonists. The consequences of activating one receptor on signaling and trafficking of another receptor are unknown. We examined the effects of selective activation of the neurokinin 1 receptor (NK1R) on signaling and trafficking of the NK3R and vice versa. Selective agonists of NK1R and NK3R induced membrane translocation of beta-arrestins (beta-ARRs). Dominant negative beta-ARR(319-418) inhibited endocytosis of NK1R and NK3R. Whereas an NK1R agonist caused sequestration of NK1R with beta-ARR in the same endosomes, thereby depleting them from the cytosol, beta-ARRs did not prominently sequester with the activated NK3R and rapidly returned to the cytosol. In cells coexpressing both receptors, prior activation of the NK1R inhibited endocytosis and homologous desensitization of the NK3R, which was dose-dependently reversed by overexpression of beta-ARR1. Similar results were obtained in enteric neurons that naturally coexpress the NK1R and NK3R. In contrast, activation of the NK3R did not affect NK1R endocytosis or desensitization. Thus, the high-affinity and prolonged interaction of the NK1R with beta-ARRs depletes beta-ARRs from the cytosol and limits their role in desensitization and endocytosis of the NK3R. Because beta-ARRs are critical for desensitization, endocytosis, and mitogenic signaling of many receptors, this sequestration is likely to have important and widespread implications.


Asunto(s)
Arrestinas/metabolismo , Proteínas de Unión al GTP/metabolismo , Receptores de Neuroquinina-1/metabolismo , Receptores de Neuroquinina-3/metabolismo , Transducción de Señal , Animales , Arrestinas/genética , Transporte Biológico , Línea Celular Transformada , Endocitosis , Endosomas/metabolismo , Ratones , Ratas , Receptores de Neuroquinina-1/agonistas , Receptores de Neuroquinina-1/genética , Receptores de Neuroquinina-3/agonistas , Receptores de Neuroquinina-3/genética , beta-Arrestina 1 , beta-Arrestinas
16.
J Biol Chem ; 279(29): 30670-9, 2004 Jul 16.
Artículo en Inglés | MEDLINE | ID: mdl-15128739

RESUMEN

Substance P (SP) induces endocytosis and recycling of the neurokinin 1 receptor (NK1R) in endothelial cells and spinal neurons at sites of inflammation and pain, and it is thus important to understand the mechanism and function of receptor trafficking. We investigated how the SP concentration affects NK1R trafficking and determined the role of Rab GTPases in trafficking. NK1R trafficking was markedly influenced by the SP concentration. High SP (10 nM) induced translocation of the NK1R and beta-arrestin 1 to perinuclear sorting endosomes containing Rab5a, where NK1R remained for >60 min. Low SP (1 nM) induced translocation of the NK1R to early endosomes located immediately beneath the plasma membrane that also contained Rab5a and beta-arrestin 1, followed by rapid recycling of the NK1R. Overexpression of Rab5a promoted NK1R translocation to perinuclear sorting endosomes, whereas the GTP binding-deficient mutant Rab5aS34N caused retention of the NK1R in superficial early endosomes. NK1R translocated from superficial early endosomes to recycling endosomes containing Rab4a and Rab11a, and Rab11aS25N inhibited NK1R recycling. Rapid NK1R recycling coincided with resensitization of SP-induced Ca2+ mobilization and with the return of surface SP binding sites. Resensitization was minimally affected by inhibition of vacuolar H(+)-ATPase and phosphatases but was markedly suppressed by disruption of Rab4a and Rab11a. Thus, whereas beta-arrestins mediate NK1R endocytosis, Rab5a regulates translocation between early and sorting endosomes, and Rab4a and Rab11a regulate trafficking through recycling endosomes. We have thus identified a new function of Rab5a as a control protein for directing concentration-dependent trafficking of the NK1R into different intracellular compartments and obtained evidence that Rab4a and Rab11a contribute to G-protein-coupled receptor recycling from early endosomes.


Asunto(s)
Receptores de Neuroquinina-1/química , Proteínas de Unión al GTP rab/metabolismo , Proteínas de Unión al GTP rab4/metabolismo , Animales , Arrestinas/química , Arrestinas/metabolismo , Western Blotting , Calcio/metabolismo , Línea Celular , ADN Complementario/metabolismo , Relación Dosis-Respuesta a Droga , Endocitosis , Endosomas/metabolismo , Citometría de Flujo , Glutatión Transferasa/metabolismo , Proteínas Fluorescentes Verdes , Cinética , Proteínas Luminiscentes/metabolismo , Microscopía Fluorescente , Estructura Terciaria de Proteína , Transporte de Proteínas , Ratas , Receptores Acoplados a Proteínas G/metabolismo , Receptores de Neuroquinina-1/metabolismo , Temperatura , Factores de Tiempo , beta-Arrestina 1 , beta-Arrestinas , Proteínas de Unión al GTP rab5/metabolismo
17.
J Immunol ; 169(9): 5315-21, 2002 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-12391252

RESUMEN

Trypsin and mast cell tryptase can signal to epithelial cells, myocytes, and nerve fibers of the respiratory tract by cleaving proteinase-activated receptor 2 (PAR2). Since tryptase inhibitors are under development to treat asthma, a precise understanding of the contribution of PAR2 to airway inflammation is required. We examined the role of PAR2 in allergic inflammation of the airway by comparing OVA-sensitized and -challenged mice lacking or overexpressing PAR2. In wild-type mice, immunoreactive PAR2 was detected in airway epithelial cells and myocytes, and intranasal administration of a PAR2 agonist stimulated macrophage infiltration into bronchoalveolar lavage fluid. OVA challenge of immunized wild-type mice stimulated infiltration of leukocytes into bronchoalveolar lavage and induced airway hyperreactivity to inhaled methacholine. Compared with wild-type animals, eosinophil infiltration was inhibited by 73% in mice lacking PAR2 and increased by 88% in mice overexpressing PAR2. Similarly, compared with wild-type animals, airway hyperreactivity to inhaled methacholine (40 micro g/ml) was diminished 38% in mice lacking PAR2 and increased by 52% in mice overexpressing PAR2. PAR2 deletion also reduced IgE levels to OVA sensitization by 4-fold compared with those of wild-type animals. Thus, PAR2 contributes to the development of immunity and to allergic inflammation of the airway. Our results support the proposal that tryptase inhibitors and PAR2 antagonists may be useful therapies for inflammatory airway disease.


Asunto(s)
Alérgenos/administración & dosificación , Hiperreactividad Bronquial/inmunología , Movimiento Celular/inmunología , Pulmón/patología , Eosinofilia Pulmonar/inmunología , Receptores de Trombina/fisiología , Alérgenos/inmunología , Animales , Hiperreactividad Bronquial/genética , Hiperreactividad Bronquial/metabolismo , Hiperreactividad Bronquial/patología , Línea Celular , Movimiento Celular/genética , Cruzamientos Genéticos , Femenino , Humanos , Inmunoglobulina E/biosíntesis , Inflamación/genética , Inflamación/inmunología , Inflamación/metabolismo , Inyecciones Intraperitoneales , Pulmón/inmunología , Pulmón/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Ovalbúmina/administración & dosificación , Ovalbúmina/inmunología , Eosinofilia Pulmonar/genética , Eosinofilia Pulmonar/metabolismo , Eosinofilia Pulmonar/patología , Receptor PAR-2 , Receptores de Trombina/deficiencia , Receptores de Trombina/genética , Receptores de Trombina/metabolismo
18.
Gastroenterology ; 125(1): 126-35, 2003 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-12851878

RESUMEN

BACKGROUND & AIMS: The components of the kinin system, including kinongens, kininogenases, and B(2) and B(1) receptors, are expressed and activated during inflammation. Here, we investigated the expression of the kinin B(2) receptor messenger RNA, kininogen and kallikrein immunoreactivity, and the ability of kinins to contract control and inflamed gallbladders in vitro. METHODS: Human gallbladders, obtained from patients undergoing cholecystectomy either for acute cholecystitis secondary to gallstone disease or during elective gastro-entero-pancreatic surgery (controls), were processed for reverse-transcription polymerase chain reaction analysis, kallikrein and kininogen immunohistochemistry, binding studies, and in vitro contractility studies. RESULTS: Tissue expression of B(2) receptor messenger RNA and specific binding of [(3)H]-bradykinin increased significantly in acute cholecystitis compared to controls. Kallikrein immunoreactivity was detected in the epithelium and infiltrating leukocytes, whereas kininogen immunoreactivity in the lumen of blood vessels and interstitial space. Bradykinin contracted isolated strips of control and acute cholecystitis gallbladders. In acute cholecystitis tissue, efficacy of bradykinin was higher than that of control gallbladders and similar to that of cholecystokinin. The contraction induced by bradykinin was significantly attenuated by B(2) receptor antagonism but not by cyclooxygenase inhibition and B(1), muscarinic, or tachykinin receptor antagonism. CONCLUSIONS: All the components of the kinin system are expressed in the human gallbladder. Bradykinin is a powerful spasmogen via B(2) receptor activation in the normal and, especially, in the inflamed human gallbladder.


Asunto(s)
Bradiquinina/análogos & derivados , Bradiquinina/metabolismo , Vaciamiento Vesicular/fisiología , Vesícula Biliar/fisiología , Receptores de Bradiquinina/genética , Receptores de Bradiquinina/metabolismo , Antagonistas Adrenérgicos beta/farmacología , Adulto , Anciano , Antipsicóticos/farmacología , Atropina/farmacología , Benzamidas/farmacología , Bradiquinina/farmacología , Antagonistas de los Receptores de Bradiquinina , Inhibidores de la Ciclooxigenasa/farmacología , Femenino , Vaciamiento Vesicular/efectos de los fármacos , Expresión Génica , Humanos , Inmunohistoquímica , Técnicas In Vitro , Indometacina/farmacología , Masculino , Persona de Mediana Edad , Antagonistas Muscarínicos/farmacología , Piperidinas/farmacología , Quinuclidinas/farmacología , Receptor de Bradiquinina B1 , Receptor de Bradiquinina B2 , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Tritio
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA