Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 32
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
Neuroscience ; 422: 32-43, 2019 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-31678341

RESUMO

A stroke-like event follows seizures which may be responsible for the postictal state and a contributing factor to the development of seizure-induced brain abnormalities and behavioral dysfunction associated with epilepsy. Caffeine is the world's most popular drug with ∼85% of people in the USA consuming it daily. Thus, persons with epilepsy are likely to have caffeine in their body and brain during seizures. This preclinical study investigated the effects of acute caffeine on local hippocampal tissue oxygenation pre and post seizure. We continuously measured local oxygen levels in the CA1 region of the hippocampus and utilized the electrical kindling model in rats. Rats were acutely administered either caffeine, or one of its metabolites, or agonists and antagonists at adenosine sub-receptor types or ryanodine receptors prior to the elicitation of seizures. Acute caffeine administration caused a significant drop in pre-seizure hippocampal pO2. Following a seizure, caffeine, as well as two of its metabolites paraxanthine, and theophylline, increased the time below the severe hypoxic threshold (10 mmHg). Likewise, the specific A2A receptor antagonist, SCH-58261, mimicked caffeine by causing a significant drop in pre-seizure pO2 and the area and time below the severe hypoxic threshold. Moreover, the A2A receptor agonist, CGS-21680 was able to prevent the effect of both caffeine and SCH-58261 adding further evidence that caffeine is likely acting through the A2A receptor. Clinical tracking and investigations are needed to determine the effect of caffeine on postictal symptomology and blood flow in persons with epilepsy.


Assuntos
Cafeína/efeitos adversos , Hipóxia/fisiopatologia , Receptores A2 de Adenosina/fisiologia , Convulsões/fisiopatologia , Adenosina/análogos & derivados , Adenosina/farmacologia , Animais , Região CA1 Hipocampal/metabolismo , Cafeína/análogos & derivados , Cafeína/antagonistas & inibidores , Relação Dose-Resposta a Droga , Hipóxia/complicações , Excitação Neurológica/efeitos dos fármacos , Masculino , Oxigênio/metabolismo , Fenetilaminas/farmacologia , Pirimidinas/antagonistas & inibidores , Pirimidinas/farmacologia , Ratos , Receptores A2 de Adenosina/efeitos dos fármacos , Convulsões/complicações , Triazóis/antagonistas & inibidores , Triazóis/farmacologia
2.
J Neural Transm (Vienna) ; 126(12): 1577-1588, 2019 12.
Artigo em Inglês | MEDLINE | ID: mdl-31522257

RESUMO

Ketamine (KET), an anesthetic, analgesic, and a sedative N-methyl-D-aspartate (NMDA) receptor antagonist agent, exposure during neonatal period may lead to learning impairment, behavioral abnormalities, and cognitive decline in the later years of life. In recent studies, it has been reported that sedative-acting α2 agonist dexmedetomidine (DEX), which is commonly used in clinical practice with KET, has neuroprotective effects and prevents the undesirable effects of anesthesia. To elucidate the underlying mechanisms of these actions, we investigated the interaction between NMDA receptors α2 adrenoceptor and adulthood behaviors in neonatally KET and/or DEX administrated mice. Balb/c male mice were administrated with saline, KET (75 mg/kg), DEX (10 µg/kg), or KET + DEX (75 mg/kg + 10 µg/kg) on postnatal day 7. During adulthood (8-10 weeks old) mice were subjected to elevated plus maze, open field, and Morris water maze tests. After behavioral tests, hippocampus samples were extracted for mRNA expression studies of NMDAR subunits (GluN1, GluN2A, and GluN2B) and α2 adrenoceptor subunits (α2A, α2B, and α2C) by real-time PCR. Ketamine increased horizontal and vertical locomotor activity (p < 0.01) and impaired spatial learning-memory (p < 0.05). DEX increased anxiety-like behavior (p < 0.01), but did not affect spatial learning-memory and locomotor activity. KET + DEX impaired spatial learning-memory (p < 0.01), increased horizontal locomotor activity (p < 0.01), and anxiety-like behavior (p < 0.05). Our study implies that DEX cannot prevent the adverse effects of KET, on spatial learning-memory, and locomotor activity. In addition to this, it can be thought that during brain development, there is an interaction between NMDAR and α2 adrenoceptor systems.


Assuntos
Anestésicos Dissociativos/farmacologia , Comportamento Animal/efeitos dos fármacos , Dexmedetomidina/farmacologia , Hipnóticos e Sedativos/farmacologia , Ketamina/farmacologia , Animais , Animais Recém-Nascidos , Hipocampo/efeitos dos fármacos , Hipocampo/metabolismo , Masculino , Camundongos Endogâmicos BALB C , Receptores A2 de Adenosina/efeitos dos fármacos , Receptores A2 de Adenosina/metabolismo , Receptores de N-Metil-D-Aspartato/efeitos dos fármacos , Receptores de N-Metil-D-Aspartato/metabolismo
3.
Neurochem Int ; 131: 104550, 2019 12.
Artigo em Inglês | MEDLINE | ID: mdl-31563462

RESUMO

Caffeine is the most consumed psychostimulant drug in the world, acting as a non-selective antagonist of adenosine receptors A1R and A2AR, which are widely expressed in retinal layers. We have previously shown that caffeine, when administered acutely, acts on A1R to potentiate the NMDA receptor-induced GABA release. Now we asked if long-term caffeine exposure also modifies GABA uptake in the avian retina and which mechanisms are involved in this process. Chicken embryos aged E11 were injected with a single dose of caffeine (30 mg/kg) in the air chamber. Retinas were dissected on E15 for ex vivo neurochemical assays. Our results showed that [3H]-GABA uptake was dependent on Na+ and blocked at 4 °C or by NO-711 and caffeine. This decrease was observed after 60 min of [3H]-GABA uptake assay at E15, which is accompanied by an increase in [3H]-GABA release. Caffeine increased the protein levels of A1R without altering ADORA1 mRNA and was devoid of effects on A2AR density or ADORA2A mRNA levels. The decrease of GABA uptake promoted by caffeine was reverted by A1R activation with N6-cyclohexyl adenosine (CHA) but not by A2AR activation with CGS 21680. Caffeine exposure increased cAMP levels and GAT-1 protein levels, which was evenly expressed between E11-E15. As expected, we observed an increase of GABA containing amacrine cells and processes in the IPL, also, cAMP pathway blockage by H-89 decreased caffeine mediated [3H]-GABA uptake. Our data support the idea that chronic injection of caffeine alters GABA transport via A1R during retinal development and that the cAMP/PKA pathway plays an important role in the regulation of GAT-1 function.


Assuntos
Bloqueadores do Receptor Tipo 1 de Angiotensina II/farmacologia , Antagonistas de Receptores de Angiotensina/farmacologia , Cafeína/farmacologia , Estimulantes do Sistema Nervoso Central/farmacologia , AMP Cíclico/fisiologia , Ácido gama-Aminobutírico/metabolismo , Adenosina/análogos & derivados , Adenosina/farmacologia , Células Amácrinas/efeitos dos fármacos , Células Amácrinas/metabolismo , Animais , Cafeína/antagonistas & inibidores , Embrião de Galinha , AMP Cíclico/metabolismo , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Proteínas da Membrana Plasmática de Transporte de GABA/metabolismo , Fenetilaminas/farmacologia , Receptor A1 de Adenosina/efeitos dos fármacos , Receptor A1 de Adenosina/metabolismo , Receptor Tipo 1 de Angiotensina/efeitos dos fármacos , Receptores A2 de Adenosina/efeitos dos fármacos , Receptores A2 de Adenosina/metabolismo , Retina/efeitos dos fármacos , Retina/embriologia , Retina/crescimento & desenvolvimento , Transdução de Sinais/efeitos dos fármacos
4.
J Cell Physiol ; 234(2): 1295-1299, 2019 02.
Artigo em Inglês | MEDLINE | ID: mdl-30146778

RESUMO

Adenosine and its analogs are of particular interest as potential therapeutic agents for treatment of cardiovascular diseases (CVDs). A2 adenosine receptor subtypes (A2a and A2b) are extensively expressed in cardiovascular system, and modulation of these receptors using A2 adenosine receptor agonists or antagonists regulates heart rate, blood pressure, heart rate variability, and cardiovascular toxicity during both normoxia and hypoxia conditions. Regulation of A2 adenosine receptor signaling via specific and novel pharmacological regulators is a potentially novel therapeutic approach for a better understanding and hence a better management of CVDs. This review summarizes the role of pharmacological A2 adenosine receptor regulators in the pathogenesis of CVDs.


Assuntos
Agonistas do Receptor A2 de Adenosina/uso terapêutico , Antagonistas do Receptor A2 de Adenosina/uso terapêutico , Doenças Cardiovasculares/tratamento farmacológico , Sistema Cardiovascular/efeitos dos fármacos , Receptores A2 de Adenosina/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos , Agonistas do Receptor A2 de Adenosina/efeitos adversos , Antagonistas do Receptor A2 de Adenosina/efeitos adversos , Animais , Doenças Cardiovasculares/metabolismo , Doenças Cardiovasculares/fisiopatologia , Sistema Cardiovascular/metabolismo , Sistema Cardiovascular/fisiopatologia , Humanos , Terapia de Alvo Molecular , Receptores A2 de Adenosina/metabolismo , Resultado do Tratamento
5.
Brain Res ; 1686: 65-71, 2018 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-29476752

RESUMO

Neural progenitor cells (NPCs) play a key role not only in the maintenance of the adult central nervous system (CNS) but also in the ability to recover from injury and disease. In this study, we established a 96-well-based screening system to screen small molecules modulating the proliferation of NPCs. A compound library composed of 1280 compounds was screened. We found that the A1 adenosine receptor agonist cyclopentyladenosine (CPA) and the A2a adenosine receptor agonist CGS-21680 increased proliferation of NPCs. The A1 adenosine receptor agonist-induced cell proliferation was attenuated by A1 adenosine receptor antagonist 8-cyclopentyl-1,3-dipropylxanthine (DPCPA). Accordingly, the A2a adenosine receptor agonist-induced cell proliferation was attenuated by A2a adenosine receptor antagonist SCH-58261. Further study indicated that CPA and CGS-21680 treatment induced phosphorylation of extracellular signal-regulated kinase (ERK) and Akt, and CPA-induced or CGS-21680-induced cell proliferation was inhibited by ERK and Akt inhibitors. These results suggested that the activation of A1 and A2a adenosine receptor stimulated the proliferation of NPCs via the ERK and Akt signaling pathways.


Assuntos
Proliferação de Células/fisiologia , Receptor A1 de Adenosina/metabolismo , Receptores A2 de Adenosina/metabolismo , Células-Tronco/metabolismo , Agonistas do Receptor A2 de Adenosina/farmacologia , Animais , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Sistema Nervoso/efeitos dos fármacos , Sistema Nervoso/metabolismo , Células-Tronco Neurais/efeitos dos fármacos , Células-Tronco Neurais/metabolismo , Fosforilação , Receptor A1 de Adenosina/efeitos dos fármacos , Receptores A2 de Adenosina/efeitos dos fármacos , Células-Tronco/efeitos dos fármacos
6.
Free Radic Biol Med ; 106: 208-218, 2017 05.
Artigo em Inglês | MEDLINE | ID: mdl-28219781

RESUMO

While it is well known that adenosine receptor activation protects the heart from ischemia/reperfusion injury, the precise mitochondrial mechanism responsible for the action remains unknown. This study probed the mitochondrial events associated with the cardioprotective effect of 5'-(N-ethylcarboxamido) adenosine (NECA), an adenosine A2 receptor agonist. Isolated rat hearts were subjected to 30min ischemia followed by 10min of reperfusion, whereas H9c2 cells experienced 20min ischemia and 10min reperfusion. NECA prevented mitochondrial structural damage, decreases in respiratory control ratio (RCR), and collapse of mitochondrial membrane potential (ΔΨm). Both the adenosine A2A receptor antagonist SCH58261 and A2B receptor antagonist MRS1706 inhibited the action of NECA. NECA reduced mitochondrial proteins carbonylation, H2O2, and superoxide generation at reperfusion, but did not change superoxide dismutase (SOD) activity. In support, the protective effects of NECA and Peg-SOD on ΔΨm upon reperfusion were additive, implying that NECA's protection is attributable to the reduced superoxide generation but not to the enhancement of the superoxide-scavenging capacity. NECA increased the mitochondrial Src tyrosine kinase activity and suppressed complex I activity at reperfusion in a Src-dependent manner. NECA also reduced mitochondrial superoxide through Src tyrosine kinase. Studies with liquid chromatography-mass spectrometer (LC-MS) identified Tyr118 of the NDUFV2 subunit of complex 1 as a likely site of the tyrosine phosphorylation. Furthermore, the complex I activity of cells transfected with the Y118F mutant was increased, suggesting that this site might be a negative regulator of complex I activity. In support, NECA failed to suppress complex I activity at reperfusion in cells transfected with the Y118F mutant of NDUFV2. In conclusion, NECA prevents mitochondrial oxidative stress by decreasing mitochondrial superoxide generation through inhibition of complex I via the mitochondrial Src tyrosine kinase. Phosphorylation of Tyr118 residue in NDUFV2 subunit may account for the inhibitory effect of NECA on complex I.


Assuntos
Adenosina-5'-(N-etilcarboxamida)/administração & dosagem , Mitocôndrias/metabolismo , Infarto do Miocárdio/metabolismo , Traumatismo por Reperfusão Miocárdica/metabolismo , NADH Desidrogenase/genética , Receptores A2 de Adenosina/metabolismo , Animais , Complexo I de Transporte de Elétrons/metabolismo , Humanos , Potencial da Membrana Mitocondrial/efeitos dos fármacos , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/patologia , Mutação , Infarto do Miocárdio/tratamento farmacológico , Infarto do Miocárdio/patologia , Traumatismo por Reperfusão Miocárdica/tratamento farmacológico , Traumatismo por Reperfusão Miocárdica/patologia , Miócitos Cardíacos/efeitos dos fármacos , Miócitos Cardíacos/metabolismo , Miócitos Cardíacos/patologia , NADH Desidrogenase/metabolismo , Estresse Oxidativo/efeitos dos fármacos , Purinas/administração & dosagem , Pirimidinas/administração & dosagem , Ratos , Receptores A2 de Adenosina/efeitos dos fármacos , Triazóis/administração & dosagem
7.
J Invest Dermatol ; 137(1): 123-131, 2017 01.
Artigo em Inglês | MEDLINE | ID: mdl-27498346

RESUMO

Adenosine is a potent regulator of inflammation and immunity, but the role of adenosine receptors in keratinocytes remains controversial. We determined that in addition to A2B receptors, human epidermal keratinocytes also express A2A receptors, although to a lower extent. Through the use of selective adenosine receptor agonists and antagonists, we showed that physiological concentrations of adenosine activate A2B receptors in normal human keratinocytes, inducing cell cycle arrest through the increase of intracellular calcium but not through cAMP signaling. In contrast, the selective activation of A2A receptors by CGS-21680 induces keratinocyte proliferation via p38-mitogen-activated protein kinase activation. Adenosine and selective A2A and A2B agonists presented anti-inflammatory profiles independent of adenosine receptors but mediated by membrane phosphatase activation. Finally, keratinocyte exposure to diverse inflammatory cytokines altered adenosine receptor expression by reducing A2B and increasing A2A, a pattern also observed in psoriatic epidermis. Because increased epidermal turnover and inflammatory response are characteristics of psoriatic disease, further studies are needed to assess the role and consequences of the altered adenosine receptor expression in lesional and nonlesional psoriatic keratinocytes.


Assuntos
Proliferação de Células/efeitos dos fármacos , Queratinócitos/fisiologia , Psoríase/patologia , Agonistas do Receptor Purinérgico P1/farmacologia , Antagonistas de Receptores Purinérgicos P1/farmacologia , Análise de Variância , Biópsia por Agulha , Western Blotting , Citocinas/metabolismo , Células Epidérmicas , Epiderme/metabolismo , Humanos , Imuno-Histoquímica , Queratinócitos/efeitos dos fármacos , Masculino , Psoríase/tratamento farmacológico , Psoríase/metabolismo , Receptor A1 de Adenosina/efeitos dos fármacos , Receptor A1 de Adenosina/metabolismo , Receptores A2 de Adenosina/efeitos dos fármacos , Receptores A2 de Adenosina/metabolismo , Estatísticas não Paramétricas
8.
Cardiovasc Res ; 99(1): 156-63, 2013 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-23539502

RESUMO

AIMS: Adenosine is a potent vasodilator contributing to cerebral blood flow regulation during metabolic stress. However, the distribution of adenosine receptor subtypes and underlying signalling mechanisms for dilation of pial arterioles remain unclear. The present study aimed at addressing these issues. METHODS AND RESULTS: Isolated porcine pial arterioles were subjected to study of vasomotor function, localization of adenosine receptors, and production of nitric oxide (NO). Concentration-dependent vasodilation to adenosine was inhibited by A2A receptor antagonist ZM241385 but not by A1 receptor antagonist 8-cyclopentyl-1,3-dipropylxanthine. A2A receptors were detected in endothelium and smooth muscle of pial arterioles via immunohistochemistry. Adenosine significantly increased arteriolar production of NO, and the induced dilation was insensitive to KATP channel blocker glibenclamide but was attenuated by endothelial denudation, NO synthase inhibitor L-NAME, or guanylyl cyclase inhibitor ODQ in a similar manner. Both inward rectifier potassium (Kir) channel inhibitor barium and cAMP signalling inhibitor Rp-8-Br-cAMPS attenuated adenosine-induced dilation. In the presence of L-NAME or the absence of endothelium, addition of Rp-8-Br-cAMPS but not barium further reduced adenosine-induced responses. Barium diminished endothelium-independent vasodilation to NO donor sodium nitroprusside. Comparable to the adenosine-induced response, vasodilation to A2A receptor agonist CGS21680 was attenuated by endothelial removal, ZM241385, L-NAME, barium, or Rp-8-Br-cAMPS, but not by glibenclamide. CONCLUSION: Adenosine evokes dilation of porcine pial arterioles via parallel activation of endothelial and smooth muscle A2A receptors. Stimulation of endothelial NO production activates smooth muscle guanylyl cyclase for vasodilation by opening Kir channels. Adenosine also activates smooth muscle cAMP signalling leading to vasodilation.


Assuntos
Artérias Cerebrais/metabolismo , Pia-Máter/irrigação sanguínea , Receptores A2 de Adenosina/metabolismo , Transdução de Sinais , Vasodilatação , Agonistas do Receptor A2 de Adenosina/farmacologia , Antagonistas do Receptor A2 de Adenosina/farmacologia , Animais , Artérias Cerebrais/efeitos dos fármacos , AMP Cíclico/metabolismo , Relação Dose-Resposta a Droga , Endotélio Vascular/efeitos dos fármacos , Endotélio Vascular/metabolismo , Inibidores Enzimáticos/farmacologia , Guanilato Ciclase/antagonistas & inibidores , Guanilato Ciclase/metabolismo , Músculo Liso Vascular/efeitos dos fármacos , Músculo Liso Vascular/metabolismo , Óxido Nítrico/metabolismo , Óxido Nítrico Sintase/antagonistas & inibidores , Óxido Nítrico Sintase/metabolismo , Bloqueadores dos Canais de Potássio/farmacologia , Canais de Potássio Corretores do Fluxo de Internalização/antagonistas & inibidores , Canais de Potássio Corretores do Fluxo de Internalização/metabolismo , Receptores A2 de Adenosina/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos , Suínos , Vasodilatação/efeitos dos fármacos , Vasodilatadores/farmacologia
9.
Basic Clin Pharmacol Toxicol ; 112(4): 229-35, 2013 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-23057724

RESUMO

Previous studies from our group have indicated important biological properties of the ethanolic extract and isolated compounds from the bulbs of Cipura paludosa (Iridaceae), a native plant widely distributed in northern Brazil, including antioxidant, neuroprotective and anti-nociceptive activities. In the present study, the effects of the ethanolic extract and its two naphthoquinones (eleutherine and isoeleutherine) on the short- and long-term memory of adult rodents were assessed in social recognition and inhibitory avoidance tasks. Acute pre-training oral administration of the ethanolic extract improved the short-term social memory in rats as well as facilitated the step-down inhibitory avoidance short- and long-term memory in mice. Moreover, the co-administration of 'non-effective' doses of the extract of Cipura paludosa and the adenosine receptor antagonists caffeine (non-selective), DPCPX (adenosine A1 receptor antagonist) and ZM241385 (adenosine A2A receptor antagonist) improved the social recognition memory of rats. In the inhibitory avoidance task, the co-administration of sub-effective doses of the extract with caffeine or ZM241385, but not with DPCPX, improved the short- and long-term memory of mice. Finally, the acute oral administration of eleutherine and isoeleutherine facilitated the inhibitory avoidance short- and long-term memory in mice. These results demonstrate for the first time the cognitive-enhancing properties of the extract and isolated compounds from the bulbs of Cipura paludosa in rodents and suggest a possible involvement of adenosine A1 and A2A receptors in these effects.


Assuntos
Memória de Longo Prazo/efeitos dos fármacos , Memória de Curto Prazo/efeitos dos fármacos , Naftoquinonas/farmacologia , Extratos Vegetais/farmacologia , Animais , Aprendizagem da Esquiva/efeitos dos fármacos , Brasil , Cafeína/farmacologia , Etanol/química , Furanos/isolamento & purificação , Furanos/farmacologia , Iridaceae/química , Masculino , Camundongos , Naftoquinonas/isolamento & purificação , Raízes de Plantas , Antagonistas de Receptores Purinérgicos P1/farmacologia , Ratos , Ratos Wistar , Receptor A1 de Adenosina/efeitos dos fármacos , Receptor A1 de Adenosina/metabolismo , Receptores A2 de Adenosina/efeitos dos fármacos , Receptores A2 de Adenosina/metabolismo , Triazinas/farmacologia , Triazóis/farmacologia , Xantinas/farmacologia
10.
ScientificWorldJournal ; 2012: 143818, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22619589

RESUMO

We examined the contribution of endothelial relaxing factors and potassium channels in actions of CPCA, potent adenosine A(2) receptor agonist, on isolated intact male rat femoral artery (FA). CPCA produced concentration-dependent relaxation of FA, which was notably, but not completely, reduced after endothelial denudation. DPCPX, A(1) receptor antagonist, had no significant effect, while SCH 58261 (A(2A) receptor antagonist) notably reduced CPCA-evoked effect. Pharmacological inhibition of nitric oxide synthase or cyclooxygenase comparably reduced CPCA-evoked action, still in a lesser degree than after denudation. In the presence of buffer with high K(+) (100 mM), CPCA-produced relaxations were almost abolished. TEA (nonselective K(Ca) blocker), glibenclamide (K(ATP) blocker), Ba(++) (K(IR) blocker), or ouabain (Na(+)/K(+)-ATPase inhibitor) did not change CPCA-induced relaxation. Concentration-response curve for CPCA was significantly shifted to the right after the incubation of apamin (SK channel blocker). CPCA produced concentration-dependent relaxation of FA that was partly dependent on endothelial cells. Endothelium-related portion of CPCA-elicited effect was mediated by combined action of endothelial NO, prostacyclin, and EDHF after activation of endothelial A(2A) receptors. Small conductance K(Ca) channels were involved in this action.


Assuntos
Agonistas do Receptor A2 de Adenosina/farmacologia , Adenosina/análogos & derivados , Autacoides/fisiologia , Artéria Femoral/efeitos dos fármacos , Receptores A2 de Adenosina/efeitos dos fármacos , Adenosina/farmacologia , Animais , Artéria Femoral/fisiologia , Relaxamento Muscular/fisiologia , Ratos
11.
Expert Opin Ther Pat ; 22(4): 369-90, 2012 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-22435652

RESUMO

INTRODUCTION: Adenosine exerts its effects by interacting with G-protein coupled receptors (GPCR) namely A(1), A(2A), A(2B) and A(3), respectively. These are involved in several diseases, for example and most importantly, Parkinson's disease, ischemia and inflammation. There is high interest in the development of potent and selective ligands for these adenosine receptor (AR) subtypes, primarily for their therapeutic potential but also as pharmacological tools in receptor studies. AREAS COVERED: This paper concentrates on reviewing the therapeutic potential of A(2) and A(3) ARs, which represent the most interesting subtypes of recent years. A general description of each receptor is reported with novel agonist and antagonist structures, patented in 2008 - 2011. PubMed and Free Patents Online databases were principally used to collect all the material. EXPERT OPINION: In the past years, by modulating A(2) and A(3)ARs, several new possible therapeutic applications were discovered. For this reason, research concerning AR ligands is still of great interest. In particular, few potent and selective A(2B) agonists and antagonists are actually reported and a clear SAR (structure-activity relationship) profile lacks for this AR subtype. At the A(3)AR, allosteric modulation may prevent problems related to the high difference between rat and human orthosteric sites and simplify the preclinical studies on A(3)AR.


Assuntos
Purinérgicos/farmacologia , Receptor A3 de Adenosina/efeitos dos fármacos , Receptores A2 de Adenosina/efeitos dos fármacos , Agonistas do Receptor A2 de Adenosina/farmacologia , Antagonistas do Receptor A2 de Adenosina/farmacologia , Agonistas do Receptor A3 de Adenosina/farmacologia , Antagonistas do Receptor A3 de Adenosina/farmacologia , Desenho de Fármacos , Humanos , Ligantes , Masculino , Estrutura Molecular , Patentes como Assunto , Purinérgicos/química , Receptor A3 de Adenosina/metabolismo , Receptores A2 de Adenosina/metabolismo , Relação Estrutura-Atividade
12.
Acta Physiol (Oxf) ; 205(3): 403-10, 2012 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-22356216

RESUMO

AIM: The precise mechanisms underlying reflex cutaneous vasodilatation during hyperthermia remain unresolved. The purpose of this study was to investigate a potential contribution of adenosine A1/A2 receptor activation to reflex cutaneous vasodilatation. METHODS: Eight subjects were equipped with four microdialysis fibres on the left forearm, and each fibre was randomly assigned one of four treatments: (1) lactated Ringer's (control); (2) 4 mm of the non-selective A1/A2 adenosine receptor antagonist theophylline; (3) 10 mm L-NAME to inhibit nitric oxide (NO) synthase; and (4) combined 4 mm theophylline and 10 mm L-NAME. Laser-Doppler flowmetry (LDF) was used as an index of skin blood flow, and blood pressure was measured beat-by-beat via photoplethysmography and verified via brachial auscultation. Whole-body heat stress to raise oral temperature 0.8 °C above baseline was induced via water-perused suits. Cutaneous vascular conductance (CVC) was calculated as LDF/mean arterial pressure and normalized to maximal (%CVC max) via infusion of 28 mm nitroprusside and local heating to 43 °C. RESULTS: There was no difference between control (65 ± 5%CVC max) and theophylline (63 ± 5%CVC max) sites. L-NAME (44 ± 4%CVC max) and theophylline + L-NAME (32 ± 3%CVC max) sites were significantly attenuated compared to both control and theophylline only sites (P<0.05), and combined theophylline + L-NAME sites were significantly reduced compared to L-NAME only sites (P<0.05). CONCLUSION: These data suggest A1/A2 adenosine receptor activation does not directly contribute to cutaneous active vasodilatation; however, a role for A1/A2 adenosine receptor activation is unmasked when NO synthase is inhibited.


Assuntos
Resposta ao Choque Térmico/fisiologia , Receptor A1 de Adenosina/fisiologia , Receptores A2 de Adenosina/fisiologia , Pele/irrigação sanguínea , Vasodilatação/fisiologia , Adulto , Inibidores Enzimáticos/farmacologia , Feminino , Humanos , Masculino , Microdiálise , NG-Nitroarginina Metil Éster/farmacologia , Óxido Nítrico Sintase/antagonistas & inibidores , Óxido Nítrico Sintase/efeitos dos fármacos , Nitroprussiato/farmacologia , Antagonistas de Receptores Purinérgicos P1/farmacologia , Receptor A1 de Adenosina/efeitos dos fármacos , Receptores A2 de Adenosina/efeitos dos fármacos , Teofilina/farmacologia , Vasodilatação/efeitos dos fármacos , Vasodilatadores/farmacologia
13.
Hippocampus ; 21(3): 265-80, 2011 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-20054814

RESUMO

Stellate neurons in layer II entorhinal cortex (EC) provide the main output from the EC to the hippocampus. It is believed that adenosine plays a crucial role in neuronal excitability and synaptic transmission in the CNS, however, the function of adenosine in the EC is still elusive. Here, the data reported showed that adenosine hyperpolarized stellate neurons in a concentration-dependent manner, accompanied by a decrease in firing frequency. This effect corresponded to the inhibition of the hyperpolarization-activated, cation nonselective (HCN) channels. Surprisingly, the adenosine-induced inhibition was blocked by 3 µM 8-cyclopentyl-1,3-dipropylxanthine (DPCPX), a selective A(1) receptor antagonists, but not by 10 µM 3,7-dimethyl-1-propargylxanthine (DMPX), a selective A(2) receptor antagonists, indicating that activation of adenosine A(1) receptors were responsible for the direct inhibition. In addition, adenosine reduced the frequency but not the amplitude of miniature EPSCs and IPSCs, suggesting that the global depression of glutamatergic and GABAergic transmission is mediated by a decrease in glutamate and GABA release, respectively. Again the presynaptic site of action was mediated by adenosine A(1) receptors. Furthermore, inhibition of spontaneous glutamate and GABA release by adenosine A(1) receptor activation was mediated by voltage-dependent Ca(2+) channels and extracellular Ca(2+) . Therefore, these findings revealed direct and indirect mechanisms by which activation of adenosine A(1) receptors on the cell bodies of stellate neurons and on the presynaptic terminals could regulate the excitability of these neurons.


Assuntos
Adenosina , Córtex Entorrinal/metabolismo , Inibição Neural/fisiologia , Receptor A1 de Adenosina/metabolismo , Transmissão Sináptica/fisiologia , Adenosina/metabolismo , Adenosina/farmacologia , Antagonistas do Receptor A1 de Adenosina/farmacologia , Antagonistas do Receptor A2 de Adenosina/farmacologia , Animais , Cálcio/metabolismo , Canais de Cálcio/metabolismo , Córtex Entorrinal/citologia , Córtex Entorrinal/efeitos dos fármacos , Potenciais Pós-Sinápticos Excitadores/fisiologia , Ácido Glutâmico/metabolismo , Inibição Neural/efeitos dos fármacos , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Técnicas de Cultura de Órgãos , Técnicas de Patch-Clamp , Terminações Pré-Sinápticas/efeitos dos fármacos , Ratos , Ratos Sprague-Dawley , Receptor A1 de Adenosina/efeitos dos fármacos , Receptores A2 de Adenosina/efeitos dos fármacos , Receptores A2 de Adenosina/metabolismo , Transmissão Sináptica/efeitos dos fármacos , Teobromina/análogos & derivados , Teobromina/farmacologia , Xantinas/farmacologia , Ácido gama-Aminobutírico/metabolismo
14.
Am J Physiol Gastrointest Liver Physiol ; 300(3): G485-93, 2011 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-21148396

RESUMO

Clinical studies implicate adenosine acting on esophageal nociceptive pathways in the pathogenesis of noncardiac chest pain originating from the esophagus. However, the effect of adenosine on esophageal afferent nerve subtypes is incompletely understood. We addressed the hypothesis that adenosine selectively activates esophageal nociceptors. Whole cell perforated patch-clamp recordings and single-cell RT-PCR analysis were performed on the primary afferent neurons retrogradely labeled from the esophagus in the guinea pig. Extracellular recordings were made from the isolated innervated esophagus. In patch-clamp studies, adenosine evoked activation (inward current) in a majority of putative nociceptive (capsaicin-sensitive) vagal nodose, vagal jugular, and spinal dorsal root ganglia (DRG) neurons innervating the esophagus. Single-cell RT-PCR analysis indicated that the majority of the putative nociceptive (transient receptor potential V1-positive) neurons innervating the esophagus express the adenosine receptors. The neural crest-derived (spinal DRG and vagal jugular) esophageal nociceptors expressed predominantly the adenosine A(1) receptor while the placodes-derived vagal nodose nociceptors expressed the adenosine A(1) and/or A(2A) receptors. Consistent with the studies in the cell bodies, adenosine evoked activation (overt action potential discharge) in esophageal nociceptive nerve terminals. Furthermore, the neural crest-derived jugular nociceptors were activated by the selective A(1) receptor agonist CCPA, and the placodes-derived nodose nociceptors were activated by CCPA and/or the selective adenosine A(2A) receptor CGS-21680. In contrast to esophageal nociceptors, adenosine failed to stimulate the vagal esophageal low-threshold (tension) mechanosensors. We conclude that adenosine selectively activates esophageal nociceptors. Our data indicate that the esophageal neural crest-derived nociceptors can be activated via the adenosine A(1) receptor while the placodes-derived esophageal nociceptors can be activated via A(1) and/or A(2A) receptors. Direct activation of esophageal nociceptors via adenosine receptors may contribute to the symptoms in esophageal diseases.


Assuntos
Adenosina/metabolismo , Dor no Peito/metabolismo , Esôfago/inervação , Gânglios Espinais/metabolismo , Nociceptores/metabolismo , Gânglio Nodoso/metabolismo , Agonistas do Receptor A1 de Adenosina/farmacologia , Agonistas do Receptor A2 de Adenosina/farmacologia , Animais , Dor no Peito/fisiopatologia , Potenciais Evocados , Gânglios Espinais/efeitos dos fármacos , Cobaias , Mecanorreceptores/metabolismo , Nociceptores/efeitos dos fármacos , Gânglio Nodoso/efeitos dos fármacos , Técnicas de Patch-Clamp , Receptor A1 de Adenosina/efeitos dos fármacos , Receptor A1 de Adenosina/genética , Receptor A1 de Adenosina/metabolismo , Receptores A2 de Adenosina/efeitos dos fármacos , Receptores A2 de Adenosina/genética , Receptores A2 de Adenosina/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa
15.
Am J Physiol Heart Circ Physiol ; 299(6): H2009-17, 2010 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-20889844

RESUMO

Multiple, perhaps interactive, mechanisms participate in the linkage between increased neural activity and cerebral vasodilation. In the present study, we assessed whether neural activation-related pial arteriolar dilation (PAD) involved interactions among adenosine (Ado) A(2) receptors (A(2)Rs), large-conductance Ca(2+)-operated K(+) (BK(Ca)) channels, and inward rectifier K(+) (K(ir)) channels. In rats with closed cranial windows, we monitored sciatic nerve stimulation (SNS)-induced PAD in the absence or presence of pharmacological blockade of A(2)Rs (ZM-241385), ecto-5'-nucleotidase (α,ß-methylene-adenosine diphosphate), BK(Ca) channels (paxilline), and K(ir) channels (BaCl(2)). Individually, these interventions led to 53-66% reductions in SNS-induced PADs. Combined applications of these blockers led to little or no further repression of SNS-induced PADs, suggesting interactions among A(2)Rs and K(+) channels. In the absence of SNS, BaCl(2) blockade of K(ir) channels produced 52-80% reductions in Ado and NS-1619 (BK(Ca) channel activator)-induced PADs. In contrast, paxilline blockade of BK(Ca) channels was without effect on dilations elicited by KCl (K(ir) channel activator) and Ado suffusions, indicating that Ado- and NS-1619-associated PADs involved K(ir) channels. In addition, targeted ablation of the superficial glia limitans was associated with a selective 60-80% loss of NS-1619 responses, suggesting that the BK(Ca) channel participation (and paxilline sensitivity) derived largely from channels within the glia limitans. Additionally, blockade of either PKA or adenylyl cyclase caused markedly attenuated pial arteriolar responses to SNS and, in the absence of SNS, responses to Ado, KCl, and NS-1619. These findings suggested a key, possibly permissive, role for A(2)R-linked cAMP generation and PKA-induced K(+) channel phosphorylation in somatosensory activation-evoked PAD.


Assuntos
Adenosina/metabolismo , Pia-Máter/irrigação sanguínea , Canais de Potássio Corretores do Fluxo de Internalização/metabolismo , Canais de Potássio/metabolismo , Receptores A2 de Adenosina/metabolismo , Transdução de Sinais , Córtex Somatossensorial/fisiologia , Vasodilatação , Inibidores de Adenilil Ciclases , Adenilil Ciclases/metabolismo , Animais , Arteríolas/inervação , Arteríolas/metabolismo , Astrócitos/metabolismo , Sinalização do Cálcio , AMP Cíclico/metabolismo , Proteínas Quinases Dependentes de AMP Cíclico/antagonistas & inibidores , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Relação Dose-Resposta a Droga , Estimulação Elétrica , Inibidores Enzimáticos/farmacologia , Feminino , Ativação do Canal Iônico , Subunidades alfa do Canal de Potássio Ativado por Cálcio de Condutância Alta , Fosforilação , Bloqueadores dos Canais de Potássio/farmacologia , Canais de Potássio/efeitos dos fármacos , Canais de Potássio Corretores do Fluxo de Internalização/antagonistas & inibidores , Antagonistas de Receptores Purinérgicos P1/farmacologia , Ratos , Ratos Sprague-Dawley , Receptores A2 de Adenosina/efeitos dos fármacos , Nervo Isquiático/fisiologia , Sistemas do Segundo Mensageiro , Transdução de Sinais/efeitos dos fármacos , Transmissão Sináptica , Vasodilatação/efeitos dos fármacos , Vasodilatadores/farmacologia
16.
Br J Pharmacol ; 153(6): 1169-76, 2008 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-18264130

RESUMO

BACKGROUND AND PURPOSE: Dipyridamole enhances post-occlusive reactive hyperaemia (PORH) in the human forearm vascular bed. We hypothesize that this effect is completely mediated by increased adenosine receptor stimulation. To test this hypothesis, the effect of caffeine (an adenosine receptor antagonist) on dipyridamole-induced augmentation of PORH was explored. EXPERIMENTAL APPROACH: The forearm vasodilator responses to three increasing periods of forearm ischaemia (2, 5 and 13 min) were determined during placebo infusion. Forty minutes after the last reperfusion period, this procedure was repeated during intra-arterial infusion of dipyridamole (7.4 nmol min(-1) per 100 ml forearm). At least 2 weeks later, this whole procedure was repeated, but now in the presence of caffeine (90 microg min(-1) per 100 ml volume). KEY RESULTS: After 2, 5 and 13 min of ischaemia, the average forearm blood flow increased to 5.6+/-0.7, 9.7+/-1.3 and 34.5+/-2.1 ml min(-1) per 100 ml. After infusion of dipyridamole into the brachial artery, these numbers were significantly increased to 7.7+/-0.8, 12.5+/-1.5 and 41.6+/-3.1 ml min(-1) per 100 ml. This response was abolished by the concomitant infusion of caffeine (6.6+/-0.5, 10.2+/-0.6, 35.1+/-2.2 (caffeine) versus 7.4+/-0.4, 10.5+/-0.6, 33.7+/-2.2 ml min(-1)per 100 ml (caffeine/dipyridamole)). CONCLUSIONS AND IMPLICATIONS: Caffeine prevented the augmenting effect of dipyridamole on PORH. This indicates that dipyridamole-induced augmentation of PORH is mediated via increased adenosine receptor stimulation as a result of elevated extracellular formation of adenosine during ischaemia.


Assuntos
Dipiridamol/farmacologia , Hiperemia/metabolismo , Receptor A1 de Adenosina/efeitos dos fármacos , Receptores A2 de Adenosina/efeitos dos fármacos , Vasodilatadores/farmacologia , Adulto , Cafeína/farmacologia , Estimulantes do Sistema Nervoso Central/farmacologia , Feminino , Antebraço/irrigação sanguínea , Humanos , Hiperemia/etiologia , Isquemia/fisiopatologia , Masculino , Receptor A1 de Adenosina/metabolismo , Receptores A2 de Adenosina/metabolismo , Fluxo Sanguíneo Regional/efeitos dos fármacos , Fatores de Tempo
17.
Handb Exp Pharmacol ; (184): 339-72, 2008.
Artigo em Inglês | MEDLINE | ID: mdl-18064419

RESUMO

Adenine-based purines, such as adenosine and ATP, are ubiquitous molecules that, in addition to their roles in metabolism, act as modulators of neurotransmitter release through activation of presynaptic P1 purinoceptors or adenosine receptors (activated by adenosine) and P2 receptors (activated by nucleotides). Of the latter, the P2Y receptors are G protein-coupled, whereas the P2X receptors are ligand-gated ion channels and not covered in this review.


Assuntos
Adenosina/fisiologia , Receptores Pré-Sinápticos/fisiologia , Receptores Purinérgicos P1/fisiologia , Receptores Purinérgicos P2/fisiologia , Adenosina/farmacologia , Animais , Humanos , Neurotransmissores/metabolismo , Purinas/metabolismo , Receptor A1 de Adenosina/efeitos dos fármacos , Receptor A1 de Adenosina/metabolismo , Receptores A2 de Adenosina/efeitos dos fármacos , Receptores A2 de Adenosina/metabolismo , Receptores Pré-Sinápticos/efeitos dos fármacos , Receptores Purinérgicos P1/efeitos dos fármacos , Receptores Purinérgicos P2/efeitos dos fármacos
18.
J Am Soc Nephrol ; 19(1): 59-68, 2008 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-18045850

RESUMO

Adenosine 2A receptor (A(2A)R) activation was recently shown to be renoprotective in diabetic nephropathy. A(2A)R are found in glomeruli and have been shown to associate with the podocyte cytoskeletal protein alpha-actinin-4, but the effect of their activation on podocyte structure and function is unknown. Podocyte injury was induced in C57BL/6 mice with puromycin aminonucleoside, and the selective A(2A)R agonist ATL313 was found to attenuate the resulting albuminuria and foot process fusion. The selective A(2A)R antagonist ZM241385 reversed the effects of ATL313. In vitro, A(2A)R mRNA and protein were expressed in a conditionally immortalized podocyte cell line, and A(2A)R-like immunoreactivity co-localized with the actin cytoskeleton. Treatment with ATL313 also blocked the increased podocyte permeability to albumin and disruption of the actin cytoskeleton that accompanied puromycin aminonucleoside-induced injury in vitro. ATL313 was ineffective, however, in the presence of the A(2A)R antagonist and in A(2A)R-deficient podocytes. It was concluded that A(2A)R activation reduces glomerular proteinuria, at least in part, by preserving the normal structure of podocyte foot processes, slit diaphragms, and actin cytoskeleton.


Assuntos
Albuminúria/prevenção & controle , Podócitos/fisiologia , Receptores A2 de Adenosina/fisiologia , Agonistas do Receptor A2 de Adenosina , Animais , Nefropatias Diabéticas/fisiopatologia , Nefropatias Diabéticas/prevenção & controle , Camundongos , Camundongos Endogâmicos C57BL , Piperidinas/uso terapêutico , Podócitos/efeitos dos fármacos , Proteinúria/induzido quimicamente , Proteinúria/prevenção & controle , Nucleosídeos de Purina/efeitos adversos , Receptores A2 de Adenosina/efeitos dos fármacos , Triazinas/farmacologia , Triazinas/uso terapêutico , Triazóis/farmacologia , Triazóis/uso terapêutico
19.
Bioorg Med Chem ; 15(22): 6956-74, 2007 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-17827019

RESUMO

The synthesis of N-(un)substituted-phenylalkylpyrimido[2,1-f]purinediones was performed starting with 7-(3-chloropropyl)-8-bromotheophylline and 7-(3-chloropropyl)-8-bromo-1,3-dipropylxanthine. Compounds with unsubstituted or substituted ethylene spacer to an aromatic ring were synthesized. Additionally variations in the spacer-elongation of the linker containing more than two atoms, introduction of a double bond or heteroatoms were performed. Physicochemical properties of the synthesized compounds were described. The obtained compounds envisaged as sterically fixed and configurationally stable analogs of 8-styrylxanthines, were evaluated for their affinity to adenosine A(1) and A(2A) receptors, the receptor subtypes that are predominant in the brain. Selected compounds were also investigated for the affinity to the A(2B) and A(3) receptor subtypes. It was stated that phenylethyl pyrimido[2,1-f]purinediones and their analogs with variations of the ethylene spacer (substituted or extended) exhibit micromolar or submicromolar affinity for A(2A) ARs (adenosine receptors); for example compound 2Ac with p-hydroxy substituent displayed a K(i) value of 0.23 microM at the rat A(2A) receptor. In comparison to the previously obtained phenyl and benzyl pyrimido[2,1-f]purinediones compounds with a shorter spacer, phenethyl derivatives were optimal for A(2A) AR. The kind of substituent at the aromatic ring was important for the affinity. Oxygen and nitrogen atoms in the spacer resulted frequently in a slight decrease of the A(2A) AR affinity, introduction of more heteroatoms into the spacer-in carbamates-caused distinctly negative effect on the activity. In this series of compounds more frequently the adenosine A(1) activity was observed, also in submicromolar range as for dipropyl derivative 2Ba with K(i) value of 0.62 microM at the rat A(2A) AR. 3D-QSAR models were developed for the compounds presented in this paper as well as in the previous publications showing activity at adenosine A(1) and A(2A) ARs. It was concluded that for the activity at adenosine A(1) and A(2A) receptors lipophilicity, steric effects along with the molecule's electrostatic surface properties had greatest value. Chosen compounds were evaluated in vivo as anticonvulsants in MES, scMet tests and examined for neurotoxicity. Contrary to previously obtained phenyl and benzyl pyrimido[2,1-f]purinediones, all tested compounds were inactive as anticonvulsants.


Assuntos
Anticonvulsivantes/farmacologia , Purinas/farmacologia , Receptor A1 de Adenosina/efeitos dos fármacos , Receptores A2 de Adenosina/efeitos dos fármacos , Animais , Anticonvulsivantes/administração & dosagem , Anticonvulsivantes/química , Sítios de Ligação , Simulação por Computador , Cristalografia por Raios X , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Injeções Intraperitoneais , Ligantes , Masculino , Camundongos , Modelos Químicos , Modelos Moleculares , Estrutura Molecular , Purinas/administração & dosagem , Purinas/química , Estereoisomerismo , Relação Estrutura-Atividade
20.
Hypertension ; 50(4): 744-9, 2007 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-17664389

RESUMO

Adenosine is an important paracrine agent regulating renal hemodynamics via adenosine A1 and A2 receptors. To determine the interactions between adenosine A1 and A2 receptors and the possible role of adenosine as a modulator of afferent arteriolar autoregulatory responses, videomicroscopic measurements of afferent arteriolar dimensions were performed at different perfusion pressures (from 100 to 125 and 150 mm Hg) using the isolated-blood-perfused rat juxtamedullary nephron preparation. Single afferent arterioles were visualized and superfused with low or high concentrations of adenosine, either alone or with the adenosine A1 receptor antagonist 8-noradamantan-3-yl-1,3-dipropylxanthine (10 micromol/L) or the adenosine A2 receptor antagonist dimethyl-1-propargylxanthine (10 micromol/L). Adenosine (20 micromol/L) decreased afferent arteriolar diameter by -9.0+/-0.9%, and this effect was enhanced by dimethyl-1-propargylxanthine (10 micromol/L) to -16.1+/-1.2%. However, autoregulatory capability was maintained. Adenosine-induced vasoconstriction was prevented by 8-noradamantan-3-yl-1,3-dipropylxanthine (10 micromol/L) with diameter increasing by 9.6+/-1.2%. Adenosine receptor saturation with a high concentration of adenosine (120 micromol/L) or blocking A1 receptors with 8-noradamantan-3-yl-1,3-dipropylxanthine in the presence of adenosine resulted in marked vasodilation and marked impairment of autoregulatory responses to increases in perfusion pressure (-1.5+/-1.1% and -3.5+/-0.9%). However, afferent arteriolar autoregulatory responses to elevations in perfusion pressure were restored after blockade of A2 receptors alone or in combination with A1 receptor blockade. During treatment with dimethyl-1-propargylxanthine in the presence of adenosine receptor saturation (120 micromol/L), afferent arteriolar autoregulatory responses were intact (-16.5+/-1.6% and -26.4+/-2.1%). These results indicate that the interactions between adenosine A1 and A2 receptors exert important modulatory influences on afferent arteriolar tone and autoregulatory capability. Activation of A2 receptors abrogates the counteracting influences of A1 receptors leading to marked vasodilation and decreased afferent arteriolar autoregulatory efficiency.


Assuntos
Adenosina/farmacologia , Arteríolas/fisiologia , Homeostase/fisiologia , Receptores A2 de Adenosina/fisiologia , Antagonistas do Receptor A1 de Adenosina , Antagonistas do Receptor A2 de Adenosina , Animais , Arteríolas/efeitos dos fármacos , Pressão Sanguínea/efeitos dos fármacos , Relação Dose-Resposta a Droga , Homeostase/efeitos dos fármacos , Rim/irrigação sanguínea , Rim/fisiologia , Masculino , Ratos , Ratos Sprague-Dawley , Receptor A1 de Adenosina/efeitos dos fármacos , Receptor A1 de Adenosina/fisiologia , Receptores A2 de Adenosina/efeitos dos fármacos , Teobromina/análogos & derivados , Teobromina/farmacologia , Vasoconstrição/efeitos dos fármacos , Xantinas/farmacologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA