Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
1.
J Pain Res ; 5: 401-8, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-23166444

RESUMO

PURPOSE: This study compared the pharmacokinetic profile, and systemic and local absorption of diclofenac, following dermal patch application and oral administration in Yorkshire-Landrace pigs. PATIENTS AND METHODS: Twelve anesthetized, female, Yorkshire-Landrace pigs were randomized to receive either the dermal patch (FLECTOR(®) patch, 10 × 14 cm; Alpharma Pharmaceuticals, a subsidiary of Pfizer Inc, New York, NY) or 50 mg oral diclofenac (Voltaren(®); Novartis, East Hanover, NJ). Tissue (skin area of 2 × 2 cm and underlying muscles approximately 2-3 cm in depth) and blood (10 mL) samples were collected at timed intervals up to 11.5 hours after initial patch application or oral administration. The concentrations of diclofenac in plasma, skin, and muscle samples were analyzed using validated ultra performance liquid chromatography tandem mass spectrometric methods. RESULTS: Peak systemic exposure of diclofenac was very low by dermal application compared with oral administration (maximum concentration [C(max)] values of 3.5 vs 9640 ng/mL, respectively). Absorption of diclofenac into underlying muscles beneath the dermal patch was sustained, and followed apparently zero-order kinetics, with the skin serving as a depot with elevated concentrations of diclofenac. Concentrations of diclofenac in muscles beneath the patch application site were similar to corresponding tissues after oral administration (C(max) values of 879 and 1160 ng/mL, respectively). In contrast to the wide tissue distribution of diclofenac after oral administration, dermal patch application resulted in high concentrations of diclofenac only on the treated skin and immediate tissue underneath the patch. Low concentrations of diclofenac were observed in the skin and muscles collected from untreated areas contralateral to the site of dermal patch application. CONCLUSION: Dermal patch application resulted in low systemic absorption and high tissue penetration of diclofenac compared with oral administration.

2.
Biochem Pharmacol ; 79(10): 1483-95, 2010 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-20096265

RESUMO

In this work, we investigated the biological functions of adenosine (ado) in metalloproteinase-9 (MMP-9) regulation in U87MG human glioblastoma cells. The nucleoside was able to increase both MMP-9 mRNA and protein levels through A3 receptors activation. We revealed that A3 receptor stimulation induced an increase of MMP-9 protein levels in cellular extracts of U87MG cells by phosphorylation of extracellular signal-regulated protein kinases (ERK1/2), c-Jun N-terminal kinase/stress-activated protein kinase (pJNK/SAPK), protein kinase B (Akt/PKB) and finally activator protein 1 (AP-1). A3 receptor activation stimulated also an increase of extracellular MMP-9 in the supernatants from U87MG glioblastoma cells. Finally, the Matrigel invasion assay demonstrated that A3 receptors, by inducing an increase in MMP-9 levels, was responsible for an increase of glioblastoma cells invasion. Collectively, these results suggest that ado, through A3 receptors activation, modulates MMP-9 protein levels and plays a role in increasing invasion of U87MG cells.


Assuntos
Glioblastoma/enzimologia , Metaloproteinase 9 da Matriz/metabolismo , Receptor A3 de Adenosina/metabolismo , Adenosina/metabolismo , Adenosina/fisiologia , Western Blotting , Butadienos/farmacologia , Linhagem Celular Tumoral , Relação Dose-Resposta a Droga , Ensaio de Imunoadsorção Enzimática , MAP Quinases Reguladas por Sinal Extracelular/antagonistas & inibidores , MAP Quinases Reguladas por Sinal Extracelular/efeitos dos fármacos , MAP Quinases Reguladas por Sinal Extracelular/fisiologia , Glioblastoma/metabolismo , Humanos , Imidazóis/farmacologia , Fosfatos de Inositol/farmacologia , Proteínas Quinases JNK Ativadas por Mitógeno/efeitos dos fármacos , Proteínas Quinases JNK Ativadas por Mitógeno/fisiologia , Metaloproteinase 9 da Matriz/fisiologia , Invasividade Neoplásica/fisiopatologia , Nitrilas/farmacologia , Fosforilação , Piridinas/farmacologia , Receptor A3 de Adenosina/fisiologia , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Fator de Transcrição AP-1/efeitos dos fármacos , Fator de Transcrição AP-1/metabolismo , Fator de Transcrição AP-1/fisiologia , Ativação Transcricional/efeitos dos fármacos , Ativação Transcricional/fisiologia
3.
FASEB J ; 24(2): 587-98, 2010 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-19776336

RESUMO

The antagonistic interaction between adenosine and dopamine receptors could have important pathophysiological and therapeutic implications in Parkinson's disease (PD). The primary aim of this study was to investigate the expression, affinity, and density of A(1), A(2A), A(2B), and A(3) adenosine receptors (ARs) and D(2) dopamine receptors (D(2)Rs) in PD. An increase in A(2A)AR density in putamen was found. The presence and functionality of ARs in human lymphocyte and neutrophil membranes from patients with PD revealed a specific A(2A)AR alteration compared with healthy subjects. A statistically significant linear correlation among the A(2A)AR density, functionality, or tumor necrosis factor-alpha (TNF-alpha) levels and Unified Parkinson's Disease Rating Scale (UPDRS) motor score was reported. Adenosine concentration and TNF-alpha levels were increased in plasma of patients with PD. In rat adrenal pheochromocytoma (PC12) cells, a widely useful model, adenosine antagonists decreased dopamine uptake, and an opposite effect was mediated by A(2A) agonists. This is the first report showing the presence of an A(2A)AR alteration in putamen in PD that mirrors a similar up-regulation in human peripheral blood cells. Moreover, the correlation found between A(2A)AR density or A(2A) agonist potency and UPDRS motor score highlights the central role of A(2A)ARs in the pharmacological treatment of PD.


Assuntos
Doença de Parkinson/fisiopatologia , Receptor A2A de Adenosina/genética , Fator de Necrose Tumoral alfa/genética , Adenosina/agonistas , Adenosina/antagonistas & inibidores , Adenosina/sangue , Idoso , Idoso de 80 Anos ou mais , Animais , Autopsia , AMP Cíclico/sangue , Dopamina/metabolismo , Feminino , Humanos , Linfócitos/química , Masculino , Pessoa de Meia-Idade , Neutrófilos/química , Células PC12 , Doença de Parkinson/sangue , Doença de Parkinson/genética , Putamen/metabolismo , RNA Mensageiro/metabolismo , Ratos , Receptor A2B de Adenosina/genética , Receptor A3 de Adenosina/genética , Receptores de Dopamina D2/genética , Fator de Necrose Tumoral alfa/sangue
4.
Arterioscler Thromb Vasc Biol ; 30(1): 90-7, 2010 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-19834107

RESUMO

OBJECTIVE: Foam cell (FC) formation by oxidized low-density lipoprotein (oxLDL) accumulation in macrophages is crucial for development of atherosclerosis. Hypoxia has been demonstrated in atherosclerosis and hypoxia-inducible factor-1 (HIF-1) has been shown to promote intraplaque angiogenesis and FC development. As hypoxia induces HIF-1alpha stabilization and adenosine (ado) accumulation, we investigated whether this nucleoside regulates HIF-1alpha in FCs. METHODS AND RESULTS: Ado, under hypoxia, stimulates HIF-1alpha accumulation by activating all adenosine receptors (ARs). HIF-1alpha modulation involved extracellular signal-regulated kinase 1/2 (ERK 1/2), p38 mitogen-activated protein kinase (p38 MAPK), and protein kinase B (Akt) phosphorylation in the case of A(1), A(2A), A(2B), and ERK 1/2 phosphorylation in the case of A(3) receptors. Ado, through the activation of A(3) and A(2B) receptors, stimulates vascular endothelial growth factor (VEGF) secretion in a HIF-1alpha-dependent way. Furthermore, ado, through the A(2B) subtype, induces an increase of Interleukin-8 (IL-8) secretion in a ERK 1/2, p38, and Akt kinase-dependent but not HIF-1alpha-mediated way. Finally, ado stimulates FC formation, and this effect is strongly reduced by A(3) and A(2B) blockers and by HIF-1alpha silencing. CONCLUSIONS: This study provides the first evidence that A(3,) A(2B), or mixed A(3)/A(2B) antagonists may be useful to block important steps in the atherosclerotic plaque development ado-induced.


Assuntos
Adenosina/metabolismo , Células Espumosas/metabolismo , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Hipóxia/metabolismo , Interleucina-8/metabolismo , Fator A de Crescimento do Endotélio Vascular/metabolismo , Aterosclerose/metabolismo , Aterosclerose/patologia , Compostos Azo , Corantes , Células Espumosas/citologia , Humanos , Hipóxia/patologia , Lipoproteínas LDL/metabolismo , Sistema de Sinalização das MAP Quinases/fisiologia , Macrófagos/citologia , Macrófagos/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , RNA Mensageiro/metabolismo , Receptor A1 de Adenosina/genética , Receptor A1 de Adenosina/metabolismo , Receptor A2A de Adenosina/genética , Receptor A2A de Adenosina/metabolismo , Receptor A2B de Adenosina/genética , Receptor A2B de Adenosina/metabolismo , Células U937
5.
FASEB J ; 24(4): 1192-204, 2010 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-20008542

RESUMO

The primary aim of this study was to investigate adenosine receptors (ARs) in bronchoalveolar lavage (BAL) macrophages from patients with chronic obstructive pulmonary disease (COPD) and age-matched healthy smokers. A(2B)ARs were significantly decreased in BAL macrophages from patients with COPD when compared with healthy smokers. The effect of proinflammatory cytokines and oxidative/nitrosative stress on AR expression and function in U937 cells before and after PMA treatment was evaluated. IL-1beta and TNF-alpha treatment up-regulated A(2A)- and A(3)ARs but not A(1)- or A(2B)ARs, whereas IL-6 did not modify AR expression. In contrast, oxidative/nitrosative stress selectively decreased A(2B)AR expression, which was associated with a reduction in the potency of the adenosine agonist 5'-N-ethylcarboxamideadenosine (NECA) to induce cAMP. Further, the ability of NECA to enhance cell proliferation was increased after oxidative/nitrosative stress. The specific involvement of A(2B)ARs was investigated by using potent and selective A(2B)AR antagonist and by A(2B)AR knockdown using siRNA and demonstrated responses similar to those obtained with oxidative/nitrosative stress. N-acetylcysteine (NAC), an antioxidant agent, counteracted the decrease in A(2B)AR expression, as well as the altered NECA effects on cAMP and cell proliferation. These findings highlight the central role of A(2B)ARs in alveolar macrophages, suggesting that their modulation could represent an innovative pharmacological strategy to manage COPD.-Varani, K., Caramori, G., Vincenzi, F., Tosi, A., Barczyk, A., Contoli, M., Casolari, P., Triggiani, M., Hansel, T., Leung, E., MacLennan, S., Barnes, P. J., Fan Chung, K., Adcock, I., Papi, A., Borea, P. A. Oxidative/nitrosative stress selectively altered A(2B) adenosine receptors in chronic obstructive pulmonary disease.


Assuntos
Regulação para Baixo , Macrófagos Alveolares/metabolismo , Estresse Oxidativo , Doença Pulmonar Obstrutiva Crônica/metabolismo , Receptor A2B de Adenosina/biossíntese , Acetilcisteína/farmacologia , Agonistas do Receptor A2 de Adenosina , Antagonistas do Receptor A2 de Adenosina , Adenosina-5'-(N-etilcarboxamida)/farmacologia , Idoso , Lavagem Broncoalveolar , Proliferação de Células/efeitos dos fármacos , AMP Cíclico/genética , AMP Cíclico/metabolismo , Citocinas/metabolismo , Citocinas/farmacologia , Feminino , Sequestradores de Radicais Livres/farmacologia , Técnicas de Silenciamento de Genes , Humanos , Macrófagos Alveolares/patologia , Masculino , Pessoa de Meia-Idade , Doença Pulmonar Obstrutiva Crônica/genética , Doença Pulmonar Obstrutiva Crônica/patologia , Doença Pulmonar Obstrutiva Crônica/terapia , RNA Interferente Pequeno/farmacologia , Receptor A2B de Adenosina/genética , Fumar/metabolismo , Fumar/patologia , Células U937 , Vasodilatadores/farmacologia
6.
Mol Pharmacol ; 72(2): 395-406, 2007 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-17488804

RESUMO

Frequent coffee consumption has been associated with a reduced risk of colorectal cancer in a number of case-control studies. Coffee is a leading source of methylxanthines, such as caffeine. The induction of vascular endothelial growth factor (VEGF) and interleukin-8 (IL-8) is an essential feature of tumor angiogenesis, and the hypoxia-inducible factor-1 (HIF-1) transcription factor is known to be a key regulator of this process. In this study, we investigated the effects of caffeine on HIF-1 protein accumulation and on VEGF and IL-8 expression in the human colon cancer cell line HT29 under hypoxic conditions. Our results show that caffeine significantly inhibits adenosine-induced HIF-1alpha protein accumulation in cancer cells. We show that HIF-1alpha and VEGF are increased through A3 adenosine receptor stimulation, whereas the effects on IL-8 are mediated via the A2B subtype. Pretreatment of cells with caffeine significantly reduces adenosine-induced VEGF promoter activity and VEGF and IL-8 expression. The mechanism of caffeine seems to involve the inhibition of the extracellular signal-regulated kinase 1/2 (ERK1/2), p38, and Akt, leading to a marked decrease in adenosine-induced HIF-1alpha accumulation, VEGF transcriptional activation, and VEGF and IL-8 protein accumulation. From a functional perspective, we observe that caffeine also significantly inhibits the A3 receptor-stimulated cell migration of colon cancer cells. Conditioned media prepared from colon cells treated with an adenosine analog increased human umbilical vein endothelial cell migration. These data provide evidence that adenosine could modulate the migration of colon cancer cells by an HIF-1alpha/VEGF/IL-8-dependent mechanism and that caffeine has the potential to inhibit colon cancer cell growth.


Assuntos
Adenosina/farmacologia , Cafeína/farmacologia , Hipóxia Celular , Neoplasias do Colo/metabolismo , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Interleucina-8/metabolismo , Fator A de Crescimento do Endotélio Vascular/metabolismo , Linhagem Celular Tumoral , Movimento Celular/efeitos dos fármacos , Neoplasias do Colo/patologia , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Humanos , Fosforilação , Regiões Promotoras Genéticas , Proteínas Proto-Oncogênicas c-akt/metabolismo , Receptor A2B de Adenosina/genética , Receptor A3 de Adenosina/genética , Receptor A3 de Adenosina/fisiologia , Fator A de Crescimento do Endotélio Vascular/genética , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
7.
Mol Pharmacol ; 72(1): 162-72, 2007 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-17400763

RESUMO

Solid tumors contain hypoxic cells that are resistant to radiotherapy and chemotherapy. The resistance in glioblastoma has been linked to the expression of antiapoptotic Bcl-2 family members. In this study, we found that in human glioblastoma cells hypoxia induces the phosphorylation of the Bcl-2 family protein Bad, thus protecting hypoxic cells from paclitaxel-induced apoptosis. Akt activation is required for the hypoxia-induced protection. In contrast, the extracellular signal-regulated kinase 1/2 activities have only a partial effect, being able to modulate Bad phosphorylation but not paclitaxel-induced apoptosis in hypoxia. We also demonstrated that the degradation of adenosine with adenosine deaminase, the knockdown of A(3) adenosine receptor expression by gene silencing, and the blockade of this receptor through A(3) receptor antagonists blocked the hypoxia-induced phosphorylation of Bad and the prolonged cell survival after treatment with paclitaxel in hypoxia. Thus, the adenosinergic signaling may be an essential component in the hypoxia survival pathway. These results suggest that hypoxia-induced chemoresistance of human glioblastoma cells may occur in a novel mechanism involving activation of adenosine-A(3) receptor-Akt pathway, which mediates Bad inactivation and favors cell survival.


Assuntos
Adenosina/fisiologia , Antineoplásicos Fitogênicos/farmacologia , Apoptose/efeitos dos fármacos , Hipóxia Celular , Glioblastoma/tratamento farmacológico , Paclitaxel/farmacologia , Proteína de Morte Celular Associada a bcl/metabolismo , Linhagem Celular Tumoral , Sobrevivência Celular , Glioblastoma/patologia , Humanos , Subunidade alfa do Fator 1 Induzível por Hipóxia/fisiologia , Fosforilação , Proteínas Proto-Oncogênicas c-akt/fisiologia , Proteínas Proto-Oncogênicas c-bcl-2/análise , Receptor A3 de Adenosina/fisiologia
8.
Biochem Pharmacol ; 72(1): 19-31, 2006 Jun 28.
Artigo em Inglês | MEDLINE | ID: mdl-16682012

RESUMO

Hypoxia appears to induce a program which shifts the cellular phenotype toward an increase in extracellular adenosine. Hypoxia-inducible factor-1 (HIF-1) is a key regulator of genes crucial to many aspects of cancer biology. Since in gliomas there is a strong correlation between HIF-1alpha expression, tumor grade and tumor vascularization, the aim of this study was to investigate whether adenosine may regulate HIF-1 in human glioblastoma cell lines. The results indicate that in the human hypoxic A172 and U87MG glioblastoma cell lines adenosine up-regulates HIF-1alpha protein expression via the A(3) receptor subtype. In particular, we investigated the effect of A(3) receptor antagonists on HIF-1 and vascular endothelial growth factor (VEGF) expression. We found that A(3) antagonists inhibit adenosine-induced HIF-1alpha and VEGF protein accumulation in the hypoxic cells. Investigations in the molecular mechanism showed that A(3) receptor stimulation activates p44/p42 and p38 MAPKs that are required for A(3)-induced increase of HIF-1alpha and VEGF. Further studies are required to demonstrate the in vivo relevance of these observations with regard to the proposed role for adenosine as a key element in hypoxia and in tumors.


Assuntos
Adenosina/farmacologia , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Glioblastoma/tratamento farmacológico , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Fator A de Crescimento do Endotélio Vascular/metabolismo , Antagonistas do Receptor A3 de Adenosina , Western Blotting , Hipóxia Celular/fisiologia , Linhagem Celular Tumoral , Relação Dose-Resposta a Droga , Ensaio de Imunoadsorção Enzimática , Inativação Gênica , Glioblastoma/metabolismo , Humanos , Subunidade alfa do Fator 1 Induzível por Hipóxia/genética , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Compostos de Fenilureia/farmacologia , RNA Mensageiro/metabolismo , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/metabolismo , Receptor A3 de Adenosina/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Triazóis/farmacologia , Regulação para Cima , Fator A de Crescimento do Endotélio Vascular/genética , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
9.
Am J Respir Crit Care Med ; 173(4): 398-406, 2006 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-16322645

RESUMO

RATIONALE: Chronic obstructive pulmonary disease (COPD) is the fourth leading cause of mortality worldwide. Adenosine is an inflammatory regulator that acts through four distinct receptors to mediate pro- and antiinflammatory effects. OBJECTIVES: The primary aim of this study was to investigate the expression, affinity, and density of adenosine receptors in peripheral lung parenchyma from age-matched smokers with COPD (n = 14) and smokers with normal lung function (control group; n = 20). METHODS: Adenosine receptors were analyzed by immunohistochemistry and saturation binding assays using typical antagonist radioligands. RESULTS: A(1), A(2A), A(2B), and A(3) receptors were expressed in different cells in peripheral lung parenchyma. The affinity of A(1), A(2A), and A(3) receptors was significantly decreased in patients with COPD compared with the control group (K(D)[A(1)] = 3.15 +/- 0.19 vs. 1.70 +/- 0.14 nM; K(D)[A(2A)] = 7.88 +/- 0.68 vs. 1.87 +/- 0.09 nM; K(D)[A(3)] = 9.34 +/- 0.27 vs. 4.41 +/- 0.25 nM; p < 0.01), whereas their density was increased (Bmax[A(1)] = 53 +/- 4 vs. 32 +/- 3 fmol/mg protein; Bmax[A(2A)] = 852 +/- 50 vs. 302 +/- 12 fmol/mg protein; Bmax[A(3)] = 2,078 +/- 108 vs. 770 +/- 34 fmol/mg protein; p < 0.01). The affinity of A(2B) receptors was not altered, but the density was significantly decreased in patients with COPD compared with the control group (Bmax = 66 +/- 5 vs. 189 +/- 16 fmol/mg protein; p < 0.01). A significant correlation was found between the affinity and density of the adenosine receptors and the FEV(1)/FVC ratio. CONCLUSIONS: This is the first report showing the presence of adenosine receptors in lung parenchyma in subjects with COPD compared with control smokers. These novel findings strengthen the hypothesis of a potential role played by adenosine receptors in the pathogenesis of COPD.


Assuntos
Doença Pulmonar Obstrutiva Crônica/imunologia , Receptores Purinérgicos P1/análise , Idoso , Feminino , Humanos , Imuno-Histoquímica , Pulmão/química , Pulmão/imunologia , Masculino , Receptores Purinérgicos P1/biossíntese
10.
Purinergic Signal ; 2(4): 627-32, 2006 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-18404465

RESUMO

Downstream A3 receptor signalling plays an important role in the regulation of cell death and proliferation. Therefore, it is important to determine the molecular pathways involved through A3 receptor stimulation. The phosphatidylinositide-3-OH kinase (PI3K)/Akt and the Raf/mitogen-activated protein kinase (MAPK/ERK) kinase (MEK)/mitogen-activated protein kinase (MAPK) pathways have central roles in the regulation of cell survival and proliferation. The crosstalk between these two pathways has also been investigated. The focus of this review centres on downstream mediators of A3 adenosine receptor signalling.

11.
Neoplasia ; 7(10): 894-903, 2005 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-16242072

RESUMO

Hypoxia-inducible factor-1 (HIF-1) is a key regulator of genes crucial to many aspects of cancer biology. The purine nucleoside, adenosine, accumulates within many tissues under hypoxic conditions, including that of tumors. Because the levels of both HIF-1 and adenosine are elevated within the hypoxic environment of solid tumors, we investigated whether adenosine may regulate HIF-1. Here we show that, under hypoxic conditions (< 2% O2), adenosine upregulates HIF-1alpha protein expression in a dose-dependent and time-dependent manner, exclusively through the A3 receptor subtype. The response to adenosine was generated at the cell surface because the inhibition of A3 receptor expression, by using small interfering RNA, abolished nucleoside effects. A3 receptor stimulation in hypoxia also increases angiopoietin-2 (Ang-2) protein accumulation through the induction of HIF-1alpha. In particular, we found that A3 receptor stimulation activates p44/p42 and p38 mitogen-activated protein kinases, which are required for A3-induced increase of HIF-1alpha and Ang-2. Collectively, these results suggest a cooperation between hypoxic and adenosine signals that ultimately may lead to the increase in HIF-1-mediated effects in cancer cells.


Assuntos
Regulação Neoplásica da Expressão Gênica , Subunidade alfa do Fator 1 Induzível por Hipóxia/biossíntese , Melanoma/metabolismo , Receptor A3 de Adenosina/fisiologia , Adenosina/química , Adenosina/metabolismo , Angiopoietina-2/metabolismo , Western Blotting , Linhagem Celular Tumoral , Relação Dose-Resposta a Droga , Ensaio de Imunoadsorção Enzimática , Citometria de Fluxo , Humanos , Hipóxia , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Neoplasias/metabolismo , Oxigênio/química , Interferência de RNA , RNA Interferente Pequeno/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Fatores de Tempo , Transfecção , Regulação para Cima , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
12.
J Biol Chem ; 280(20): 19516-26, 2005 May 20.
Artigo em Inglês | MEDLINE | ID: mdl-15774470

RESUMO

Adenosine exerts its effects through four subtypes of G-protein-coupled receptors: A(1), A(2A), A(2B), and A(3). Stimulation of the human A(3) receptor has been suggested to influence cell death and proliferation. The phosphatidylinositide-3-OH kinase (PI3K)/Akt and the Raf/mitogen-activated protein kinase (MAPK/ERK) kinase (MEK)/mitogen-activated protein kinase (MAPK) pathways have central roles in the regulation of cell survival and proliferation. Due to their importance, the cross-talk between these two pathways has been investigated. Here, we show that the A(3) adenosine receptor agonist Cl-IB-MECA stimulates PI3K-dependent phosphorylation of Akt leading to the reduction of basal levels of ERK1/2 phosphorylation, which in turn inhibits cell proliferation. The response to Cl-IB-MECA was not blocked by A(1), A(2A), or A(2B) receptor antagonists, although it was abolished by A(3) receptor antagonists. Furthermore, the response to Cl-IB-MECA was generated at the cell surface, since the inhibition of A(3) receptor expression, by using small interfering RNA, abolished agonist effects. Using A375 cells, we show that A(3) adenosine receptor stimulation results in PI3K-dependent phosphorylation of Akt, leading to the reduction of basal levels of ERK1/2 phosphorylation, which in turn inhibits cell proliferation.


Assuntos
Adenosina/análogos & derivados , Melanoma/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Proto-Oncogênicas/metabolismo , Receptor A3 de Adenosina/metabolismo , Adenosina/farmacologia , Agonistas do Receptor A3 de Adenosina , Antagonistas do Receptor A3 de Adenosina , Sequência de Bases , Divisão Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Humanos , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Melanoma/patologia , Proteína Quinase 1 Ativada por Mitógeno/antagonistas & inibidores , Proteína Quinase 3 Ativada por Mitógeno/antagonistas & inibidores , Fosforilação , Proteínas Proto-Oncogênicas c-akt , RNA Interferente Pequeno/genética , Receptor A3 de Adenosina/genética
13.
Biochem Pharmacol ; 68(2): 305-12, 2004 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-15194002

RESUMO

Until recently, the characterization of adenosine A(2B) receptors has been hampered by the lack of high affinity radioligands. This study describes the synthesis and in vitro characterization of the radiolabeled derivative of OSIP339391, a novel, potent, and selective pyrrolopyrimidine A(2B) antagonist. OSIP339391 had a selectivity of greater than 70-fold for A(2B) receptors over other human adenosine receptor subtypes. The radiolabel was introduced by hydrogenation of the acetylenic precursor with tritium gas resulting in the incorporation (on average) of three tritium atoms in the molecule, yielding a ligand with specific activity of 87Ci/mmol (3.2TBq/mmol). Using membranes from HEK-293 cells expressing the human recombinant A(2B) receptor, [3H]OSIP339391 was characterized in kinetic, saturation, and competition binding experiments. From the association and dissociation rate studies, the affinity was 0.41nM and in close agreement with that found in saturation binding experiments (0.17nM). In competition, binding studies using 0.5nM [3H]OSIP339391, the affinity of a range of agonists and antagonists was consistent with previously reported data. Thus, [3H]OSIP339391 is a novel, selective, and high affinity radioligand that can be a useful tool in the further exploration and characterization of recombinant and endogenous adenosine A(2B) receptors.


Assuntos
Antagonistas do Receptor A2 de Adenosina , Pirimidinas/farmacologia , Pirróis/farmacologia , Células Cultivadas , Humanos , Ensaio Radioligante , Fatores de Tempo , Trítio
14.
J Biomol Screen ; 7(3): 281-9, 2002 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-12097191

RESUMO

The pharmacology of G protein-coupled receptors is widely accepted to depend on the G protein subunit to which the agonist-stimulated receptor couples. In order to investigate whether CB(1) agonist-mediated signal transduction via an engineered G(alpha 16) system is different than that of the G(i/o) coupling normally preferred by the CB(1) receptor, we transfected the human recombinant CB(1) receptor (hCB(1)) or a fusion protein comprising the hCB(1) receptor and G(alpha 16) (hCB(1)-G(alpha 16)) into HEK293 cells. From competition binding studies, the rank order of ligand affinities at the hCB(1)-G(alpha 16) fusion protein was found to be similar to that for hCB(1): HU 210 > CP 55,940 > or = SR 141716A > WIN 55212-2 > anandamide > JWH 015. Agonists increased [(35)S]GTP gamma S binding or inhibited forskolin-stimulated cAMP, presumably by coupling to G(i/o), in cells expressing hCB(1) but not hCB(1)-G(alpha 16). However, an analogous rank order of potencies was observed for these agonists in their ability to evoke increases in intracellular calcium concentration in cells expressing hCB(1)-G(alpha 16) but not hCB(1). These data demonstrate that ligand affinities for the hCB(1) receptor are not affected by fusion to the G(alpha 16) subunit. Furthermore, there is essentially no difference in the function of the hCB(1) receptor when coupled to G(i/o) or G (alpha 16).


Assuntos
Proteínas Heterotriméricas de Ligação ao GTP/farmacologia , Receptores de Droga/metabolismo , Cálcio/metabolismo , AMP Cíclico/análise , Cicloexanóis/metabolismo , Subunidades alfa Gq-G11 de Proteínas de Ligação ao GTP , Guanosina 5'-O-(3-Tiotrifosfato)/metabolismo , Humanos , Receptores de Canabinoides , Proteínas Recombinantes de Fusão/farmacologia , Radioisótopos de Enxofre/metabolismo , Trítio/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...