Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
Mais filtros

Medicinas Complementares
Métodos Terapêuticos e Terapias MTCI
Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Sci Rep ; 10(1): 20781, 2020 11 27.
Artigo em Inglês | MEDLINE | ID: mdl-33247159

RESUMO

The adenosine A3 receptor (A3R) belongs to a family of four adenosine receptor (AR) subtypes which all play distinct roles throughout the body. A3R antagonists have been described as potential treatments for numerous diseases including asthma. Given the similarity between (adenosine receptors) orthosteric binding sites, obtaining highly selective antagonists is a challenging but critical task. Here we screen 39 potential A3R, antagonists using agonist-induced inhibition of cAMP. Positive hits were assessed for AR subtype selectivity through cAMP accumulation assays. The antagonist affinity was determined using Schild analysis (pA2 values) and fluorescent ligand binding. Structure-activity relationship investigations revealed that loss of the 3-(dichlorophenyl)-isoxazolyl moiety or the aromatic nitrogen heterocycle with nitrogen at α-position to the carbon of carboximidamide group significantly attenuated K18 antagonistic potency. Mutagenic studies supported by molecular dynamic simulations combined with Molecular Mechanics-Poisson Boltzmann Surface Area calculations identified the residues important for binding in the A3R orthosteric site. We demonstrate that K18, which contains a 3-(dichlorophenyl)-isoxazole group connected through carbonyloxycarboximidamide fragment with a 1,3-thiazole ring, is a specific A3R (< 1 µM) competitive antagonist. Finally, we introduce a model that enables estimates of the equilibrium binding affinity for rapidly disassociating compounds from real-time fluorescent ligand-binding studies. These results demonstrate the pharmacological characterisation of a selective competitive A3R antagonist and the description of its orthosteric binding mode. Our findings may provide new insights for drug discovery.


Assuntos
Antagonistas do Receptor A3 de Adenosina/química , Antagonistas do Receptor A3 de Adenosina/farmacologia , Antagonistas do Receptor A3 de Adenosina/farmacocinética , Animais , Sítios de Ligação/genética , Ligação Competitiva , Células CHO , Cricetulus , AMP Cíclico/metabolismo , Avaliação Pré-Clínica de Medicamentos , Humanos , Cinética , Simulação de Dinâmica Molecular , Mutagênese Sítio-Dirigida , Ensaio Radioligante , Ratos , Receptor A3 de Adenosina/química , Receptor A3 de Adenosina/genética , Receptor A3 de Adenosina/metabolismo , Receptores Purinérgicos P1/química , Receptores Purinérgicos P1/genética , Receptores Purinérgicos P1/metabolismo , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Especificidade da Espécie , Relação Estrutura-Atividade
2.
Nutrients ; 12(2)2020 Feb 14.
Artigo em Inglês | MEDLINE | ID: mdl-32075205

RESUMO

Current evidence on the effects of coffee intake on cardiovascular diseases is not consistent, in part contributed by the genetic variability of the study subjects. While adenosine receptors (ADORAs) are involved in caffeine signaling, it remains unknown how genetic variations at the ADORA loci correlate the coffee intake with cardiovascular diseases. The present study examined the associations of coffee intake with dyslipidemia risk depending on genetic variants in the ADORA gene family. The study involved a population-based cohort of 4898 Korean subjects. Consumption of more than or equal to a cup of coffee per day was associated with lower dyslipidemia risk in females carrying the ADORA2B minor allele rs2779212 (OR: 0.645, 95% CI: 0.506-0.823), but not in those with the major allele. At the ADORA2A locus, male subjects with the minor allele of rs5760423 showed instead an increased risk of dyslipidemia when consuming more than or equal to a cup of coffee per day (OR: 1.352, 95% CI: 1.014-1.802). The effect of coffee intake on dyslipidemia risk differs depending on genetic variants at the ADORA loci in a sex-specific manner. Our study suggests that a dietary guideline for coffee intake in the prevention and management of dyslipidemia ought to consider ADORA-related biomarkers carefully.


Assuntos
Café , Dislipidemias/etiologia , Dislipidemias/prevenção & controle , Ingestão de Alimentos/genética , Variação Genética/genética , Família Multigênica/genética , Receptores Purinérgicos P1/genética , Adulto , Idoso , Alelos , Povo Asiático/genética , Café/efeitos adversos , Estudos de Coortes , Dislipidemias/genética , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Receptor A2A de Adenosina , Receptor A2B de Adenosina , Risco , Caracteres Sexuais
3.
Int J Mol Sci ; 20(6)2019 Mar 22.
Artigo em Inglês | MEDLINE | ID: mdl-30909368

RESUMO

BACKGROUND: Vascular endothelial injury during ischemia generates apoptotic cell death and precedes apoptosis of underlying tissues. We aimed at studying the role of extracellular adenosine triphosphate (ATP) on endothelial cells protection against hypoxia injury. METHODS: In a hypoxic model on endothelial cells, we quantified the extracellular concentration of ATP and adenosine. The expression of mRNA (ectonucleotidases, adenosine, and P2 receptors) was measured. Apoptosis was evaluated by the expression of cleaved caspase 3. The involvement of P2 and adenosine receptors and signaling pathways was investigated using selective inhibitors. RESULTS: Hypoxic stress induced a significant increase in extracellular ATP and adenosine. After a 2-h hypoxic injury, an increase of cleaved caspase 3 was observed. ATP anti-apoptotic effect was prevented by suramin, pyridoxalphosphate-6-azophenyl-2',4'-disulfonic acid (PPADS), and CGS15943, as well as by selective A2A, A2B, and A3 receptor antagonists. P2 receptor-mediated anti-apoptotic effect of ATP involved phosphoinositide 3-kinase (PI3K), extracellular signal-regulated kinases (ERK1/2), mitoKATP, and nitric oxide synthase (NOS) pathways whereas adenosine receptor-mediated anti-apoptotic effect involved ERK1/2, protein kinase A (PKA), and NOS. CONCLUSIONS: These results suggest a complementary role of P2 and adenosine receptors in ATP-induced protective effects against hypoxia injury of endothelial. This could be considered therapeutic targets to limit the development of ischemic injury of organs such as heart, brain, and kidney.


Assuntos
Trifosfato de Adenosina/metabolismo , Apoptose , Células Endoteliais da Veia Umbilical Humana/metabolismo , Hipóxia/metabolismo , Receptores Purinérgicos P1/metabolismo , Receptores Purinérgicos P2/metabolismo , Adenosina/metabolismo , Apoptose/genética , Biomarcadores , Espaço Extracelular/metabolismo , Expressão Gênica , Humanos , Hipóxia/genética , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Óxido Nítrico Sintase/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , RNA Mensageiro/genética , Receptores Purinérgicos P1/genética , Receptores Purinérgicos P2/genética , Transdução de Sinais , Estresse Fisiológico/genética
4.
J Cell Physiol ; 234(5): 5863-5879, 2019 05.
Artigo em Inglês | MEDLINE | ID: mdl-29271489

RESUMO

Maintenance of the endothelial cell (EC) barrier is critical to vascular homeostasis and a loss of barrier integrity results in increased vascular permeability. While the mechanisms that govern increased EC permeability have been under intense investigation over the past several decades, the processes regulating the preservation/restoration of the EC barrier remain poorly understood. Herein we show that the extracellular purines, adenosine (Ado) and adenosine 5'-[γ-thio]-triphosphate (ATPγS) can strengthen the barrier function of human lung microvascular EC (HLMVEC). This ability involves protein kinase A (PKA) activation and decreases in myosin light chain 20 (MLC20) phosphorylation secondary to the involvement of MLC phosphatase (MLCP). In contrast to Ado, ATPγS-induced PKA activation is accompanied by a modest, but significant decrease in cyclic adenosine monophosphate (cAMP) levels supporting the existence of an unconventional cAMP-independent pathway of PKA activation. Furthermore, ATPγS-induced EC barrier strengthening does not involve the Rap guanine nucleotide exchange factor 3 (EPAC1) which is directly activated by cAMP but is instead dependent upon PKA-anchor protein 2 (AKAP2) expression. We also found that AKAP2 can directly interact with the myosin phosphatase-targeting protein MYPT1 and that depletion of AKAP2 abolished ATPγS-induced increases in transendothelial electrical resistance. Ado-induced strengthening of the HLMVEC barrier required the coordinated activation of PKA and EPAC1 in a cAMP-dependent manner. In summary, ATPγS-induced enhancement of the EC barrier is EPAC1-independent and is instead mediated by activation of PKA which is then guided by AKAP2, in a cAMP-independent mechanism, to activate MLCP which dephosphorylates MLC20 resulting in reduced EC contraction and preservation.


Assuntos
Trifosfato de Adenosina/análogos & derivados , Permeabilidade Capilar/efeitos dos fármacos , Microvasos/efeitos dos fármacos , Agonistas do Receptor Purinérgico P1/farmacologia , Receptores Purinérgicos P1/efeitos dos fármacos , Proteínas de Ancoragem à Quinase A/genética , Proteínas de Ancoragem à Quinase A/metabolismo , Trifosfato de Adenosina/farmacologia , AMP Cíclico/metabolismo , Proteínas Quinases Dependentes de AMP Cíclico/genética , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Impedância Elétrica , Fatores de Troca do Nucleotídeo Guanina/genética , Fatores de Troca do Nucleotídeo Guanina/metabolismo , Células HEK293 , Humanos , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Microvasos/metabolismo , Cadeias Leves de Miosina/metabolismo , Fosfatase de Miosina-de-Cadeia-Leve/genética , Fosfatase de Miosina-de-Cadeia-Leve/metabolismo , Fosforilação , Receptores Purinérgicos P1/genética , Receptores Purinérgicos P1/metabolismo , Transdução de Sinais
5.
Food Chem Toxicol ; 123: 298-313, 2019 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-30291944

RESUMO

Coffee is a drink prepared from roasted coffee beans and is lauded for its aroma and flavour. It is the third most popular beverage in the world. This beverage is known by its stimulant effect associated with the presence of methylxanthines. Caffeine, a purine-like molecule (1,3,7 trymetylxantine), is the most important bioactive compound in coffee, among others such as chlorogenic acid (CGA), diterpenes, and trigonelline. CGA is a phenolic acid with biological properties as antioxidant, anti-inflammatory, neuroprotector, hypolipidemic, and hypoglicemic. Purinergic system plays a key role inneuromodulation and homeostasis. Extracellular ATP, other nucleotides and adenosine are signalling molecules that act through their specific receptors, namely purinoceptors, P1 for nucleosides and P2 for nucleotides. They regulate many pathological processes, since adenosine, for instance, can limit the damage caused by ATP in the excitotoxicity from the neuronal cells. The primary purpose of this review is to discuss the effects of coffee, caffeine, and CGA on the purinergic system. This review focuses on the relationship/interplay between coffee, caffeine, CGA, and adenosine, and their effects on ectonucleotidases activities as well as on the modulation of P1 and P2 receptors from central nervous system and also in peripheral tissue.


Assuntos
Cafeína/metabolismo , Ácido Clorogênico/metabolismo , Extratos Vegetais/metabolismo , Purinas/metabolismo , Animais , Cafeína/química , Ácido Clorogênico/química , Coffea/química , Café/química , Café/metabolismo , Humanos , Extratos Vegetais/química , Receptores Purinérgicos P1/genética , Receptores Purinérgicos P1/metabolismo , Receptores Purinérgicos P2/genética , Receptores Purinérgicos P2/metabolismo , Transdução de Sinais
6.
Zhen Ci Yan Jiu ; 42(1): 39-44, 2017 Feb 25.
Artigo em Chinês | MEDLINE | ID: mdl-29071996

RESUMO

OBJECTIVE: To investigate the effects of electroacupuncture (EA) on the expression of adenosine receptor (AR) in the white adipose tissue (WAT) of diet-induced obese (DIO) mice, so as to reveal a peripheral mechanism of EA underlying improvement of body weight. METHODS: Forty three-week-old C 57 BL/6 male mice were divided into normal diet group (n=12) and high fat diet group (n=28) randomly, and fed by normal diet and high fat diet for 12 weeks, respectively. In the high fat diet group, mice with body weight over 20% heavier than that of the normal diet group were considered as obese mice. The normal diet mice and the obese mice were divided into normal group (CD, n=5) and normal plus EA group (CD+EA, n=7), or obese group (HFD, n=6) and obese plus EA group (HFD+EA, n=12). The CD+EA group and the HFD+EA group were treated with EA at "Zusanli"(ST 36) and "Neiting"(ST 44, 2 Hz/15 Hz, 0.6-1.0 mA) for 20 min, 6 times a week for 4 weeks. Body weight, ratio of WAT/body weight were calculated, qPCR and Western blot were applied to detect mRNA and protein levels of adenosine receptors in the epididymal adipose tissue (Epi-WAT), respectively. RESULTS: Compared with the normal diet group, high fat diet significantly increased body weight in C 57 BL/6 mice after feeding for 12 weeks (P<0.01); 18 out of 28 mice in the high fat diet group were classified as obesity. After treatment, the body weight and the ratio of Epi-WAT/body weight of the HFD group were increased than those in the CD group (P<0.05), the change of body weight in the HFD group was bigger than that in the CD group (P<0.01). Compared with the HFD group, the body weight and the ratio of Epi-WAT/body weight of the HFD+EA group were decreased after EA (P<0.05), the change of body weight was also significantly increased (P<0.01). No significant differences were found among the four groups in the expression level of A1R mRNA (P>0.05). The expression of A3R mRNA in the HFD group was lower than that in the CD group (P<0.01), while the expressions of A2A R and A2BR proteins were decreased in the HFD group than in the CD group (P<0.01). In comparison with the HFD group, the expression levels of A2AR and A2BR mRNAs and proteins were significantly up-regulated in the HFD+EA group, respectively (P<0.05, P<0.01). CONCLUSIONS: EA intervention is able to reduce the body weight of DIO mice, which Feb be associated with its effects in regulating the expression of A2AR and A2BR in WAT, suggesting a new mechanism of EA in accelerating peripheral WAT metabolism.


Assuntos
Tecido Adiposo Branco/metabolismo , Eletroacupuntura , Obesidade/terapia , Receptores Purinérgicos P1/genética , Receptores Purinérgicos P1/metabolismo , Animais , Dieta Hiperlipídica/efeitos adversos , Expressão Gênica , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Obesidade/genética , Obesidade/metabolismo
7.
Nat Commun ; 8: 15904, 2017 06 27.
Artigo em Inglês | MEDLINE | ID: mdl-28654087

RESUMO

Caffeine, an antagonist of the adenosine receptor A1R, is used as a dietary supplement to reduce body weight, although the underlying mechanism is unclear. Here, we report that adenosine level in the cerebrospinal fluid, and hypothalamic expression of A1R, are increased in the diet-induced obesity (DIO) mouse. We find that mice with overexpression of A1R in the neurons of paraventricular nucleus (PVN) of the hypothalamus are hyperphagic, have glucose intolerance and high body weight. Central or peripheral administration of caffeine reduces the body weight of DIO mice by the suppression of appetite and increasing of energy expenditure. We also show that caffeine excites oxytocin expressing neurons, and blockade of the action of oxytocin significantly attenuates the effect of caffeine on energy balance. These data suggest that caffeine inhibits A1Rs expressed on PVN oxytocin neurons to negatively regulate energy balance in DIO mice.


Assuntos
Cafeína/metabolismo , Hipotálamo/metabolismo , Obesidade/metabolismo , Ocitocina/metabolismo , Receptores Purinérgicos P1/metabolismo , Animais , Cafeína/administração & dosagem , Dieta Hiperlipídica/efeitos adversos , Metabolismo Energético , Glucose/metabolismo , Humanos , Leptina/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Neurônios/metabolismo , Obesidade/dietoterapia , Obesidade/etiologia , Obesidade/genética , Núcleo Hipotalâmico Paraventricular/metabolismo , Antagonistas de Receptores Purinérgicos P1/metabolismo , Receptores Purinérgicos P1/genética
8.
Sci Rep ; 7: 44816, 2017 03 20.
Artigo em Inglês | MEDLINE | ID: mdl-28317879

RESUMO

Agonists that target the A1, A2A, A2B and A3 adenosine receptors have potential to be potent treatment options for a number of diseases, including autoimmune diseases, cardiovascular disease and cancer. Because each of these adenosine receptors plays a distinct role throughout the body, obtaining highly specific receptor agonists is essential. Of these receptors, the adenosine A2AR and A2BR share many sequence and structural similarities but highly differ in their responses to inflammatory stimuli. Our laboratory, using a combination of specially developed cell lines and calcium release analysis hardware, has created a new and faster method for determining specificity of synthetic adenosine agonist compounds for the A2A and A2B receptors in human cells. A2A receptor expression was effectively removed from K562 cells, resulting in the development of a distinct null line. Using HIV-lentivector and plasmid DNA transfection, we also developed A2A and A2B receptor over-expressing lines. As adenosine is known to cause changes in intracellular calcium levels upon addition to cell culture, calcium release can be determined in these cell lines upon compound addition, providing a functional readout of receptor activation and allowing us to isolate the most specific adenosine agonist compounds.


Assuntos
Descoberta de Drogas/métodos , Agonistas do Receptor Purinérgico P1/química , Agonistas do Receptor Purinérgico P1/farmacologia , Receptores Purinérgicos P1/genética , Receptores Purinérgicos P1/metabolismo , Adenosina/metabolismo , Sistemas CRISPR-Cas , Cálcio/metabolismo , Células Cultivadas , Avaliação Pré-Clínica de Medicamentos/métodos , Citometria de Fluxo , Expressão Gênica , Técnicas de Inativação de Genes , Marcação de Genes , Humanos , Células K562 , Receptores Purinérgicos P1/química , Receptores Purinérgicos P1/classificação
9.
Mol Aspects Med ; 55: 9-19, 2017 06.
Artigo em Inglês | MEDLINE | ID: mdl-28093236

RESUMO

Extracellular adenosine 5'-triphosphate (ATP) and adenosine molecules are intimately involved in immune responses. ATP is mostly a pro-inflammatory molecule and is released during hypoxic condition and by necrotic cells, as well as by activated immune cells and endothelial cells. However, under certain conditions, for instance at low concentrations or at prolonged exposure, ATP may also have anti-inflammatory properties. Extracellular ATP can activate both P2X and P2Y purinergic receptors. Extracellular ATP can be hydrolyzed into adenosine in a two-step enzymatic process involving the ectonucleotidases CD39 (ecto-apyrase) and CD73. These enzymes are expressed by many cell types, including endothelial cells and immune cells. The counterpart of ATP is adenosine, which is produced by breakdown of intra- or extracellular ATP. Adenosine has mainly anti-inflammatory effects by binding to the adenosine, or P1, receptors (A1, A2A, A2B, and A3). These receptors are also expressed in many cells, including immune cells. The final effect of ATP and adenosine in immune responses depends on the fine regulatory balance between the 2 molecules. In the present review, we will discuss the current knowledge on the role of these 2 molecules in the immune responses.


Assuntos
Trifosfato de Adenosina/metabolismo , Imunidade Celular/genética , Inflamação/genética , Receptores Purinérgicos P1/genética , 5'-Nucleotidase/genética , Adenosina/genética , Adenosina/imunologia , Adenosina/metabolismo , Trifosfato de Adenosina/imunologia , Antígenos CD/genética , Apirase/genética , Células Endoteliais/imunologia , Células Endoteliais/metabolismo , Humanos , Inflamação/imunologia , Inflamação/patologia , Receptores Purinérgicos P1/metabolismo , Receptores Purinérgicos P2X/genética , Receptores Purinérgicos P2Y/genética
10.
J Clin Endocrinol Metab ; 98(8): E1345-51, 2013 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-23788688

RESUMO

CONTEXT AND OBJECTIVE: Caffeine is a highly consumed psychoactive substance present in our daily drinks. Independent studies have reported associations between caffeine consumption, low bone mineral density, and urinary calcium loss, as well as impaired bone development in vitro and in vivo. Calcium (Ca(2+)), vitamin D, and PTH are critical regulators of bone remodeling. A possible association between caffeine and parathyroid gland function has been suggested in the literature. DESIGN, SETTING, AND PATIENTS: Effects of caffeine on PTH secretion and Ca(2+) levels were determined by batch incubation and Fura-2, respectively, in pathological parathyroid cells. Protein expressions were studied by Western blot and immunohistochemistry in normal and parathyroid adenoma tissues. Alterations in gene expressions of adenosine receptor A1 (ADORA1) and A2 (ADORA2A) and PTH were quantified by PCR; intracellular cAMP levels and protein kinase A activity were analyzed by an antibody-based assay. RESULTS: We studied physiological concentrations of caffeine ranging from 1 to 50 µm and found that 50 µm caffeine caused a significant decrease of PTH secretion and PTH gene expression. This decrease occurred in parallel with a decrease of the intracellular cAMP level, protein kinase A activity, and ADORA1 gene expression, indicating a possible causal relationship. The intracellular level of Ca(2+) was unaffected even by high concentrations of caffeine. Protein expressions demonstrated two main targets for caffeine-ADORA1 and ADORA2A. CONCLUSION: A physiological high dose of caffeine inhibits PTH secretion in human parathyroid cells, possibly due to a decrease of the intracellular level of cAMP. The observation demonstrates a functional link between caffeine and parathyroid cell function.


Assuntos
Cafeína/farmacologia , Glândulas Paratireoides/efeitos dos fármacos , Hormônio Paratireóideo/metabolismo , Adulto , Idoso , Idoso de 80 Anos ou mais , Cálcio/metabolismo , AMP Cíclico/análise , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Glândulas Paratireoides/citologia , Glândulas Paratireoides/metabolismo , Hormônio Paratireóideo/genética , Receptores Purinérgicos P1/análise , Receptores Purinérgicos P1/genética
11.
Am J Physiol Endocrinol Metab ; 290(5): E940-51, 2006 May.
Artigo em Inglês | MEDLINE | ID: mdl-16352677

RESUMO

Inosine, an endogenous nucleoside, has recently been shown to exert potent effects on the immune, neural, and cardiovascular systems. This work addresses modulation of intermediary metabolism by inosine through adenosine receptors (ARs) in isolated rat hepatocytes. We conducted an in silico search in the GenBank and complete genomic sequence databases for additional adenosine/inosine receptors and for a feasible physiological role of inosine in homeostasis. Inosine stimulated glycogenolysis (approximately 40%, EC50 4.2 x 10(-9) M), gluconeogenesis (approximately 40%, EC50 7.8 x 10(-9) M), and ureagenesis (approximately 130%, EC50 7.0 x 10(-8) M) compared with basal values; these effects were blunted by the selective A3 AR antagonist 9-chloro-2-(2-furanyl)-5-[(phenylacetyl)amino][1,2,4]-triazolo[1,5-c]quinazoline (MRS 1220) but not by selective A1, A2A, and A2B AR antagonists. In addition, MRS 1220 antagonized inosine-induced transient increase (40%) in cytosolic Ca2+ and enhanced (90%) glycogen phosphorylase activity. Inosine-induced Ca2+ mobilization was desensitized by adenosine; in a reciprocal manner, inosine desensitized adenosine action. Inosine decreased the cAMP pool in hepatocytes when A1, A2A, and A2B AR were blocked by a mixture of selective antagonists. Inosine-promoted metabolic changes were unrelated to cAMP decrease but were Ca2+ dependent because they were absent in hepatocytes incubated in EGTA- or BAPTA-AM-supplemented Ca2+-free medium. After in silico analysis, no additional cognate adenosine/inosine receptors were found in human, mouse, and rat. In both perfused rat liver and isolated hepatocytes, hypoxia/reoxygenation produced an increase in inosine, adenosine, and glucose release; these actions were quantitatively greater in perfused rat liver than in isolated cells. Moreover, all of these effects were impaired by the antagonist MRS 1220. On the basis of results obtained, known higher extracellular inosine levels under ischemic conditions, and inosine's higher sensitivity for stimulating hepatic gluconeogenesis, it is suggested that, after tissular ischemia, inosine contributes to the maintenance of homeostasis by releasing glucose from the liver through stimulation of A3 ARs.


Assuntos
Glucose/metabolismo , Hepatócitos/metabolismo , Inosina/metabolismo , Receptor A3 de Adenosina/fisiologia , Adenosina/metabolismo , Adenosina/farmacologia , Antagonistas do Receptor A3 de Adenosina , Animais , Cálcio/metabolismo , Hipóxia Celular , AMP Cíclico/metabolismo , Gluconeogênese/efeitos dos fármacos , Glicogênio Fosforilase/metabolismo , Glicogenólise/efeitos dos fármacos , Hepatócitos/efeitos dos fármacos , Inosina/farmacologia , Fígado/metabolismo , Masculino , Filogenia , Agonistas do Receptor Purinérgico P1 , Antagonistas de Receptores Purinérgicos P1 , Quinazolinas/farmacologia , Ratos , Ratos Wistar , Receptor A3 de Adenosina/genética , Receptores Acoplados a Proteínas G/genética , Receptores Purinérgicos/genética , Receptores Purinérgicos P1/genética , Triazóis/farmacologia , Ureia/metabolismo
12.
Nucleosides Nucleotides Nucleic Acids ; 24(10-12): 1507-17, 2005.
Artigo em Inglês | MEDLINE | ID: mdl-16438031

RESUMO

Residues of the second extracellular loop are believed to be important for ligand recognition in adenosine receptors. Molecular modeling studies have suggested that one such residue, Gln167 of the human A3 receptor, is in proximity to the C2 moiety of some adenosine analogs when bound. Here this putative interaction was systematically explored using a neoceptor strategy, i.e., by site-directed mutagenesis and examination of the affinities of nucleosides modified to have complementary functionality. Gln167 was mutated to Ala, Glu, and Arg, while the 2-position of several adenosine analogs was substituted with amine or carboxylic acid groups. All compounds tested lost affinity to the mutant receptors in comparison to the wild type. However, comparing affinities among the mutant receptors, several compounds bearing charge at the 2-position demonstrated preferential affinity for the mutant receptor bearing a residue of complementary charge. 13, with a positively-charged C2 moiety, displayed an 8.5-fold increase in affinity at the Q167E mutant receptor versus the Q167R mutant receptor Preferential affinity for specific mutant receptors was also observed for 8 and 12. The data suggests that a direct contact is made between the C2 substituent of some charged ligands and the mutant receptor bearing the opposite charge at position 167.


Assuntos
Adenosina/farmacologia , Substituição de Aminoácidos , Mutagênese Sítio-Dirigida , Mutação Puntual , Receptores Purinérgicos P1/genética , Adenosina/análogos & derivados , Adenosina/metabolismo , Linhagem Celular , Glutamina/genética , Humanos , Mutagênese Sítio-Dirigida/métodos , Ligação Proteica/genética , Receptores Purinérgicos P1/metabolismo
13.
Infect Immun ; 72(3): 1349-57, 2004 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-14977938

RESUMO

Adenosine is a potent anti-inflammatory agent that modulates the function of cells involved in the inflammatory response. Here we show that it inhibits lipopolysaccharide (LPS)-induced formation of reactive oxygen intermediates (ROI) in both freshly isolated and cultured human monocytes. Blocking of adenosine uptake and inactivation of the adenosine-degrading enzyme adenosine deaminase enhanced the inhibitory action of adenosine, indicating that both pathways regulate the extracellular adenosine concentration. Adenosine-mediated inhibition could be reversed by XAC (xanthine amine congener), an antagonist of the adenosine receptor A(2A), and MRS 1220 [N-9-chloro-2-(2-furanyl)[1, 2, 4]-triazolo[1,5-c]quinazolin-5-benzeneacetamide], an A(3) receptor antagonist, in both cell populations, while DPCPX (1,3-dipropyl-8-cyclopentylxanthine), an A(1) receptor antagonist, had no effect. Similar to what was seen with adenosine, CGS 21680, an A(2A) and A(3) receptor agonist, and IB-MECA, a nonselective A(1) and A(3) receptor agonist, dose dependently prevented ROI formation, indicating the involvement of A(3) and probably also A(2A) in the suppressive effect of adenosine. Pretreatment of monocytes with adenosine did not lead to changes in the LPS-induced increase in intracellular calcium levels ([Ca(2+)](i)). Thus, participation of [Ca(2+)](i) in the action of adenosine seems unlikely. The adenosine-mediated suppression of ROI production was found to be more pronounced when monocytes were cultured for 18 h, a time point at which changes in the mRNA expression of adenosine receptors were observed. Most prominent was the increase in the A(2A) receptor mRNA. These data demonstrate that cultivation of monocytes is accompanied by changes in the inhibitory action of adenosine mediated by A(3) and probably also the A(2A) receptor and that regulation of adenosine receptors is an integral part of the monocyte differentiation program.


Assuntos
Adenina/análogos & derivados , Monócitos/metabolismo , Receptores Purinérgicos P1/classificação , Receptores Purinérgicos P1/metabolismo , Adenina/farmacologia , Adenosina/farmacologia , Adenosina Desaminase/metabolismo , Sequência de Bases , Sinalização do Cálcio/efeitos dos fármacos , Células Cultivadas , DNA Complementar/genética , Dipiridamol/farmacologia , Humanos , Técnicas In Vitro , Lipopolissacarídeos/farmacologia , Monócitos/efeitos dos fármacos , Antagonistas de Receptores Purinérgicos P1 , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Receptores Purinérgicos P1/genética , Explosão Respiratória/efeitos dos fármacos
14.
Behav Brain Res ; 143(2): 201-7, 2003 Aug 14.
Artigo em Inglês | MEDLINE | ID: mdl-12900046

RESUMO

Adenosine and dopamine receptors interact in the CNS to modulate behaviour, including sensorimotor gating. Prepulse inhibition (PPI) has been suggested to be an operational measure of sensorimotor gating. PPI and startle habituation are disrupted in patients with schizophrenia. In experimental animals, both parameters are modulated by dopaminergic and adenosine receptor agonists and antagonists. In the present study, we measured PPI and startle habituation in mice that lack functional adenosine A(2A) receptors. Startle amplitudes, startle habituation and PPI were significantly reduced in mice homozygous null for the adenosine A(2A) receptor (A(2A)(-/-)). In addition, differential effects of amphetamine and MK-801 on startle amplitude, startle habituation and PPI were observed between A(2A)(-/-) and wildtype controls. These data support the involvement of adenosine A(2A) receptors in regulation of PPI and startle habituation.


Assuntos
Habituação Psicofisiológica/fisiologia , Inibição Neural/fisiologia , Receptores Purinérgicos P1/fisiologia , Reflexo de Sobressalto/fisiologia , Limiar Sensorial/fisiologia , Estimulação Acústica , Anfetamina/farmacologia , Animais , Maleato de Dizocilpina/farmacologia , Dopaminérgicos/farmacologia , Antagonistas de Aminoácidos Excitatórios/farmacologia , Habituação Psicofisiológica/efeitos dos fármacos , Habituação Psicofisiológica/genética , Masculino , Camundongos , Camundongos Knockout , Inibição Neural/efeitos dos fármacos , Inibição Neural/genética , Receptor A2A de Adenosina , Receptores Purinérgicos P1/deficiência , Receptores Purinérgicos P1/genética , Reflexo de Sobressalto/efeitos dos fármacos , Reflexo de Sobressalto/genética , Limiar Sensorial/efeitos dos fármacos
15.
J Biol Chem ; 276(32): 30199-207, 2001 Aug 10.
Artigo em Inglês | MEDLINE | ID: mdl-11333255

RESUMO

The G(s)-coupled rat A(2B) adenosine receptor (A(2B)-AR) was epitope-tagged at the NH(2) terminus with hemagglutinin (HA) and subjected to progressive deletions or point mutations of the COOH terminus in order to determine regions of the receptor that contribute to agonist-induced desensitization and internalization. When expressed stably in Chinese hamster ovary cells, a mutant receptor in which the final 2 amino acids were deleted, the Leu(330)-stop mutant, underwent rapid agonist-induced desensitization and internalization as did the wild type (WT) receptor. However, the Phe(328) and the Gln(325)-stop mutants were resistant to rapid agonist-induced desensitization and internalization. Co-expression of arrestin-2-green fluorescent protein (arrestin-2-GFP) with WT receptor or Leu(330)-stop mutant resulted in rapid translocation of arrestin-2-GFP from cytosol to membrane upon agonist addition. On the other hand, agonist activation of the Phe(328)-stop or Gln(325)-stop mutant did not result in translocation of arrestin-2-GFP from cytosol. A COOH terminus point mutant, S329G, was also unable to undergo rapid agonist-induced desensitization and internalization, indicating that Ser(329) is a critical residue for these processes. A further deletion mutant (Ser(326)-stop) unexpectedly underwent rapid agonist-induced desensitization and internalization. However, activation of this mutant did not promote translocation of arrestin-2-GFP from cytosol to membrane. In addition, whereas WT receptor internalization was markedly inhibited by co-expression of dominant negative mutants of arrestin-2 (arrestin-2-(319-418)), dynamin (dynamin K44A), or Eps-15 (EDelta95-295), Ser(326)-stop receptor internalization was only inhibited by dominant negative mutant dynamin. Taken together these results indicate that Ser(329), close to the COOH terminus of the rat A(2B)-AR, is critical for the rapid agonist-induced desensitization and internalization of the receptor. However, deletion of the COOH terminus also uncovers a motif that is able to redirect internalization of the receptor to an arrestin- and clathrin-independent pathway.


Assuntos
Receptores Purinérgicos P1/química , Receptores Purinérgicos P1/metabolismo , Serina/química , Sequência de Aminoácidos , Animais , Arrestinas/genética , Arrestinas/metabolismo , Sítios de Ligação , Células CHO , Membrana Celular/metabolismo , Clatrina/metabolismo , Clonagem Molecular , Cricetinae , Citosol/metabolismo , DNA Complementar/metabolismo , Relação Dose-Resposta a Droga , Dinaminas , Ensaio de Imunoadsorção Enzimática , Epitopos , GTP Fosfo-Hidrolases/genética , Deleção de Genes , Genes Dominantes , Glutamina/química , Proteínas de Fluorescência Verde , Leucina/química , Proteínas Luminescentes/metabolismo , Microscopia de Fluorescência , Dados de Sequência Molecular , Mutagênese Sítio-Dirigida , Mutação , Fenilalanina/química , Fosfoproteínas/genética , Fosfoproteínas/metabolismo , Mutação Puntual , Ratos , Receptor A2B de Adenosina , Receptores Purinérgicos P1/genética , Proteínas Recombinantes de Fusão/metabolismo , Homologia de Sequência de Aminoácidos , Fatores de Tempo , Transfecção
16.
Naunyn Schmiedebergs Arch Pharmacol ; 363(1): 81-6, 2001 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-11191840

RESUMO

We have reported previously the cloning and partial characterization of a chick A1 adenosine receptor expressed in the heart. We report herein the cloning of a chick A3 adenosine receptor and a comprehensive characterization of both the A1 and A3 receptors expressed in human embryonic kidney 293 cells. [125I]N6-(p-aminobenzyl)adenosine bound to both receptors with similar affinities and was used in competition studies. Although the selectivities of both agonists and antagonists were less than in other species, two antagonists, 3-ethyl-5-benzyl-2-methyl-6-phenyl-4-phenylethynal-(+/-)-dihydropyridine-3,5-dicarboxylate and 3,6-dichloro-2'-(isopropyloxy)-4'-methylflavone), were at least partially selective for A3 receptors while one antagonist [C8-(N-methylisopropyl)amine-N6-(5'endohydroxy)endonorboman-2-yl-9-methyladenine] was selective for A1 receptors. While both receptors coupled to the inhibition of adenylyl cyclase, we were unable to detect coupling of either receptor to phospholipase C or D.


Assuntos
Adenosina/análogos & derivados , Galinhas/genética , Receptores Purinérgicos P1/genética , Adenosina/metabolismo , Adenosina/farmacologia , Adenosina-5'-(N-etilcarboxamida)/farmacologia , Sequência de Aminoácidos , Animais , Sequência de Bases , Ligação Competitiva/efeitos dos fármacos , Encéfalo/metabolismo , Linhagem Celular , Clonagem Molecular , AMP Cíclico/metabolismo , DNA Complementar/química , DNA Complementar/genética , DNA Complementar/isolamento & purificação , DNA Recombinante , Di-Hidropiridinas/farmacologia , Expressão Gênica , Humanos , Radioisótopos do Iodo , Iodobenzenos/metabolismo , Iodobenzenos/farmacologia , Ligantes , Dados de Sequência Molecular , Fosfolipase D/metabolismo , Ensaio Radioligante , Receptor A3 de Adenosina , Receptores Purinérgicos P1/efeitos dos fármacos , Receptores Purinérgicos P1/metabolismo , Análise de Sequência de DNA , Transfecção , Fosfolipases Tipo C/metabolismo , Xantinas/farmacologia
17.
J Biol Chem ; 274(6): 3617-21, 1999 Feb 05.
Artigo em Inglês | MEDLINE | ID: mdl-9920910

RESUMO

To provide new insights into ligand-A1 adenosine receptor (A1AR) interactions, site-directed mutagenesis was used to test the role of several residues in the first four transmembrane domains of the human A1AR. First, we replaced eight unique A1AR residues with amino acids present at corresponding transmembrane (TM) positions of A2AARs. We also tested the role of carboxamide amino acids in TMs 1-4, and the roles of Val-87, Leu-88, and Thr-91 in TM3. Following conversion of Gly-14 in TM1 to Thr-14, the affinity for adenosine agonists increased 100-fold, and after Pro-25 in TM1 was converted to Leu-25, the affinity for agonists fell. After conversion of TM3 sites Thr-91 to Ala-91, and Gln-92 to Ala-92, the affinity for N6-substituted agonists was reduced, and binding of ligands without N6 substituents was eliminated. When Leu-88 was converted to Ala-88, the binding of ligands with N6 substituents was reduced to a greater extent than ligands without N6 substituents. Following conversion of Pro-86 to Phe-86, the affinity for N6-substituted agonists was lost, and the affinity for ligands without N6 substitution was reduced. These observations strongly suggest that Thr-91 and Gln-92 in TM3 interact with the adenosine adenine moiety, and Leu-88 and Pro-86 play roles in conferring specificity for A1AR selective compounds. Using computer modeling based on the structure of rhodopsin, a revised model of adenosine-A1AR interactions is proposed with the N6-adenine position oriented toward the top of TM3 and the ribose group interacting with the bottom half of TMs 3 and 7.


Assuntos
Adenina/metabolismo , Receptores Purinérgicos P1/metabolismo , Amidas/metabolismo , Sequência de Aminoácidos , Aminoácidos/metabolismo , Sítios de Ligação , DNA Complementar , Humanos , Modelos Moleculares , Dados de Sequência Molecular , Mutagênese Sítio-Dirigida , Receptores Purinérgicos P1/química , Receptores Purinérgicos P1/genética
18.
Am J Physiol ; 274(2): H441-7, 1998 02.
Artigo em Inglês | MEDLINE | ID: mdl-9486246

RESUMO

There is evidence to suggest that during ischemia adenosine acts on cardiac vagal afferent neurons to activate systemic reflexes and to modulate cardiac nociception. The purpose of this study was to determine whether adenosine receptors are present and have direct cellular electrophysiological actions on cardiac vagal afferent neurons. In radioreceptor assays of nodose ganglion tissue from rats, binding was detectable for A1 (39.6 +/- 1.2 fmol/mg protein) but not A2a adenosine receptors. These findings were confirmed using the complementary approach of receptor-labeling autoradiography. Using in situ hybridization, we saw specific labeling over approximately 50% of neurons in the nodose ganglia, but not over nonneuronal cells. In colabeling studies, cardiac vagal afferent neurons were identified by retroneuronal labeling with fluororuby. Of cardiac vagal afferents approximately one-half were strongly positive for A1 adenosine receptors (immunocytochemistry). In patch-clamping experiments, adenosine inhibited peak inward calcium current in 7 of 11 cells by 48 +/- 13%. In conclusion, adenosine A1 receptors reside on a subset of vagal afferent neurons, including cardiac vagal afferents, and have electrophysiological effects that modulate neuroexcitability in cultured nodose ganglion neurons.


Assuntos
Vias Aferentes/fisiologia , Coração/inervação , Receptores Purinérgicos P1/análise , Receptores Purinérgicos P1/fisiologia , Nervo Vago/fisiologia , Animais , Autorradiografia , Condutividade Elétrica , Eletrofisiologia , Expressão Gênica , Hibridização In Situ , Neurônios/química , Neurônios/fisiologia , Gânglio Nodoso/química , Gânglio Nodoso/metabolismo , Técnicas de Patch-Clamp , RNA/análise , Ensaio Radioligante , Ratos , Ratos Sprague-Dawley , Receptores Purinérgicos P1/genética , Trítio , Xantinas/metabolismo
19.
Mol Pharmacol ; 52(5): 846-60, 1997 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-9351976

RESUMO

We cloned and characterized the canine A3 adenosine receptor (AR) and examined AR-induced degranulation of the BR line of canine mastocytoma cells. Canine A3AR transcript is found predominantly in spleen, lung, liver, and testes and encodes a 314-amino acid heptahelical receptor. 125I-N6-Aminobenzyladenosine binds to two affinity states of canine A3AR with KD values of 0.7 +/- 0.1 and 16 +/- 0.8 nM, reflecting G protein-coupled and -uncoupled receptors, respectively. Xanthine antagonists bind with similar affinities to human, canine, and rabbit receptors but with 80-400-fold lower affinities to rat A3AR. Although canine BR mastocytoma cells contain A1AR, A2BAR, and A3AR, degranulation seems to be mediated primarily by A2BARs stimulated by the nonselective agonist 5'-N-ethylcarboxamidoadenosine (NECA) but not by the A3-selective agonist N6-(3-iodobenzyl)adenosine-5'-N-methylcarboxamide. NECA-stimulated degranulation is not prevented by pertussis toxin and is blocked by enprofylline (Ki = 7 microM), an antiasthmatic xanthine with low affinity (Ki > 100 microM) for A1AR, A2AAR, and A3AR. NECA increases canine mastocytoma cell cAMP, Ca2+, and inositol trisphosphate levels; these responses are antagonized half-maximally by 7-15 microM enprofylline. The results suggest that (i) the cloned canine A3AR is structurally and pharmacologically more similar to human than to rat A3AR; (ii) the A2BAR, and not the A1AR or A3AR, is principally responsible for adenosine-mediated degranulation of canine BR mastocytoma cells; and (iii) the BR cell A2BAR couples to both Ca2+ mobilization and cAMP accumulation. Although A2B receptors play a major role in the regulation of BR mast cell degranulation, multiple AR subtypes and G proteins may influence mast cell functions.


Assuntos
DNA Complementar/genética , Mastócitos/química , Sarcoma de Mastócitos/química , Proteínas de Neoplasias/genética , Receptores Purinérgicos P1/genética , Adenina/análogos & derivados , Adenina/farmacologia , Adenosina/análogos & derivados , Adenosina/farmacologia , Sequência de Aminoácidos , Animais , Sequência de Bases , Células COS , Cálcio/metabolismo , Chlorocebus aethiops , DNA Complementar/metabolismo , Fosfatos de Dinucleosídeos/farmacologia , Cães , Mastócitos/fisiologia , Sarcoma de Mastócitos/metabolismo , Dados de Sequência Molecular , Proteínas de Neoplasias/metabolismo , Norbornanos/farmacologia , RNA Mensageiro/metabolismo , Receptor A3 de Adenosina , Receptores Purinérgicos P1/metabolismo , Alinhamento de Sequência , Homologia de Sequência de Aminoácidos , Xantinas/farmacologia , beta-N-Acetil-Hexosaminidases/metabolismo
20.
Blood ; 88(9): 3569-74, 1996 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-8896425

RESUMO

Adenosine (ADO) is a potent bronchoconstrictor in allergic patients and has been shown to increase the release of histamine from human lung tissues. Antagonists of ADO A1 and A2A receptors are not effective in attenuating these effects. Therefore, involvement of ADO A3 receptors in the bronchoconstrictor and/or inflammatory effects have to be considered. Eosinophils also play a pivotal role in allergic diseases such as asthma, thus it is natural to consider a link between the A3 receptor and eosinophils. Human peripheral blood eosinophils express the ADO A3 receptor as indicated by detection of the transcript for A3 receptors in polymerase chain reaction-amplified cDNA derived from the cells. A3 receptors on eosinophil membranes were characterized using the A3 receptor agonist radioligand 125I-labeled AB-MECA, which yielded Bmax and Kd values of 1.31 pmol/mg protein and 3.19 nmol/L, respectively. Treatment of eosinophils with the highly potent and selective A3 receptor agonist CI-IB-MECA clearly induced Ca2+ release from intracellular Ca2+ pools followed by Ca2+ influx, suggesting the presence of phospholipase C-coupled A3 receptors. In contrast, the ADO receptor agonists CPA and CGS 21680, selective for A1 and A2A receptors, respectively, at concentrations of < or = 30 mumol/ L did not elevate the intracellular Ca2+ level. These results attest to the existence of ADO A3 receptors on eosinophils and suggest that ADO stimulates these cells to release Ca2+ from intracellular stores via the activation of A3 receptors.


Assuntos
Cálcio/metabolismo , Eosinófilos/metabolismo , Receptores Purinérgicos P1/metabolismo , Transdução de Sinais , Adenosina/análogos & derivados , Adenosina/farmacologia , Células Cultivadas , DNA Complementar/genética , DNA Complementar/isolamento & purificação , Humanos , Reação em Cadeia da Polimerase , Agonistas do Receptor Purinérgico P1 , Receptores Purinérgicos P1/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA