Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 197
Filtrar
1.
Turk J Med Sci ; 53(5): 1045-1057, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-38813043

RESUMO

Background: Levetiracetam (LEV) has been found to have an antihyperalgesic effect via acting on the adenosine system. However, the effects of LEV on the modulation of the adenosine system in the brain have not been elucidated in the prevention of seizures and epilepsy. The present study aimed to explore the possible LEV mechanisms of action in the adenosine signaling systems in an animal model of epilepsy. Methodology: A docking study was initially performed to determine the possible interaction of LEV with adenosine A1 receptors (A1Rs) and equilibrative nucleoside transporters-1 (ENT1). The experimental study was divided into an acute seizure test (32 mice distributed into 4 groups) and a chronic kindling model study (40 mice distributed into 5 groups), followed by gene expression analysis and immunohistochemistry. The kindling model lasted 26 days and took 13 subconvulsive doses of pentylenetetrazole (PTZ) to completely kindle the mice in the PTZ control group. Gene expression changes in the A1Rs, potassium inwardly-rectifying channel 3.2 (Kir3.2), and ENT1 in the brain tissue samples of the mice following treatment with LEV were analyzed using reverse transcription-quantitative polymerase chain reaction, and immunohistochemistry was performed for the A1R protein expression. Results: Docking studies predicted a significant interaction of LEV with A1Rs and ENT1 proteins. Results from the acute testing revealed that caffeine (100 mg/kg) and 8-cyclopentyl-1,3-dipropylxanthine (25 mg/kg) significantly reversed the antiseizure effects of LEV by reversing the percent protection and shortening the onset of the first myoclonic jerk (FMJ) and generalized clonic seizures (GCSs). In the PTZ-induced kindling, LEV demonstrated an increased gene expression of A1Rs and Kir3.2 in the brain. LEV also significantly reduced the gene expression of ENT1. Furthermore, the immunohistochemical analysis showed that LEV increased the protein expression of A1Rs in the brain. Conclusion: Based on these results, it can be concluded that LEV modulates epileptogenesis by acting on the adenosine pathway in the central nervous system.


Assuntos
Anticonvulsivantes , Modelos Animais de Doenças , Epilepsia , Excitação Neurológica , Levetiracetam , Animais , Levetiracetam/farmacologia , Camundongos , Epilepsia/tratamento farmacológico , Epilepsia/metabolismo , Anticonvulsivantes/farmacologia , Excitação Neurológica/efeitos dos fármacos , Masculino , Piracetam/farmacologia , Piracetam/análogos & derivados , Receptor A1 de Adenosina/metabolismo , Receptor A1 de Adenosina/efeitos dos fármacos , Receptor A1 de Adenosina/genética , Pentilenotetrazol , Simulação de Acoplamento Molecular , Transdução de Sinais/efeitos dos fármacos , Adenosina/análogos & derivados , Adenosina/farmacologia , Transportador Equilibrativo 1 de Nucleosídeo/metabolismo , Transportador Equilibrativo 1 de Nucleosídeo/genética
2.
Biomed Pharmacother ; 145: 112446, 2022 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-34808556

RESUMO

Cordycepin (known as 3-deoxyadenosine, CRD), a natural product from the valuable traditional Chinese medicine Cordyceps militaris, has been reported to improve cognitive function and modulate neuroprotective effects on the central nervous system (CNS). However, the modulating mechanisms of cordycepin on information processing in hippocampal CA1 pyramidal neurons are not fully understood. To clarify how cordycepin modulates synaptic responses of pyramidal neurons in rat hippocampal CA1 region, we conducted an electrophysiological experiment using whole-cell patch-clamp technique. The spontaneous and miniature excitatory postsynaptic currents (sEPSCs and mEPSCs, respectively) and the spontaneous and miniature inhibitory postsynaptic currents (sIPSCs and mIPSCs, respectively) recorded by this technique evaluated pure single or multi-synapse responses and enabled us to accurately quantify how cordycepin influenced the pre and postsynaptic aspects of synaptic transmission. The present results showed that cordycepin significantly decreased the frequency of both glutamatergic and GABAergic postsynaptic currents without affecting the amplitude, while these inhibitory effects were antagonized by the A1 adenosine receptor antagonist (DPCPX), but not the A2A (ZM 241385), A2B (MRS1754) and A3 (MRS1191) adenosine receptor antagonists. Taken together, our results suggested that cordycepin had a clear presynaptic effect on glutamatergic and GABAergic transmission, and provided novel evidence that cordycepin suppresses the synaptic transmission through the activation of A1AR.


Assuntos
Desoxiadenosinas/farmacologia , Fármacos Neuroprotetores/farmacologia , Células Piramidais/efeitos dos fármacos , Transmissão Sináptica/efeitos dos fármacos , Animais , Feminino , Ácido Glutâmico/metabolismo , Hipocampo/efeitos dos fármacos , Hipocampo/metabolismo , Masculino , Células Piramidais/metabolismo , Ratos , Ratos Sprague-Dawley , Receptor A1 de Adenosina/efeitos dos fármacos , Receptor A1 de Adenosina/metabolismo , Ácido gama-Aminobutírico/metabolismo
3.
J Neurochem ; 160(3): 305-324, 2022 02.
Artigo em Inglês | MEDLINE | ID: mdl-34905223

RESUMO

Extracellular adenosine plays prominent roles in the brain in both physiological and pathological conditions. Adenosine can be generated following the degradation of extracellular nucleotides by various types of ectonucleotidases. Several ectonucleotidases are present in the brain parenchyma: ecto-nucleotide triphosphate diphosphohydrolases 1 and 3 (NTPDase 1 and 3), ecto-nucleotide pyrophosphatase/phosphodiesterase 1 (NPP 1), ecto-5'-nucleotidase (eN), and tissue non-specific alkaline phosphatase (TNAP, whose function in the brain has received little attention). Here we examined, in a living brain preparation, the role of these ectonucleotidases in generating extracellular adenosine. We recorded local field potentials evoked by electrical stimulation of the lateral olfactory tract in the mouse piriform cortex in vitro. Variations in adenosine level were evaluated by measuring changes in presynaptic inhibition generated by adenosine A1 receptors (A1Rs) activation. A1R-mediated presynaptic inhibition was present endogenously and was enhanced by bath-applied AMP and ATP. We hypothesized that inhibiting ectonucleotidases would reduce extracellular adenosine concentration, which would result in a weakening of presynaptic inhibition. However, inhibiting TNAP had no effect in controlling endogenous adenosine action and no effect on presynaptic inhibition induced by bath-applied AMP. Furthermore, contrary to our expectation, inhibiting TNAP reinforced, rather than reduced, presynaptic inhibition induced by bath-applied ATP. Similarly, inhibition of NTPDase 1 and 3, NPP1, and eN induced stronger, rather than weaker, presynaptic inhibition, both in endogenous condition and with bath-applied ATP and AMP. Consequently, attempts to suppress the functions of extracellular adenosine by blocking its extracellular synthesis in living brain tissue could have functional impacts opposite to those anticipated.


Assuntos
Córtex Cerebral/efeitos dos fármacos , Inibidores Enzimáticos/farmacologia , Nucleotidases/antagonistas & inibidores , Transmissão Sináptica/efeitos dos fármacos , 5'-Nucleotidase/antagonistas & inibidores , Adenosina/metabolismo , Agonistas do Receptor A1 de Adenosina/farmacologia , Monofosfato de Adenosina/farmacologia , Trifosfato de Adenosina/farmacologia , Fosfatase Alcalina/antagonistas & inibidores , Animais , Estimulação Elétrica , Potenciais Evocados/efeitos dos fármacos , Feminino , Camundongos , Camundongos Endogâmicos C57BL , Bulbo Olfatório/efeitos dos fármacos , Receptor A1 de Adenosina/efeitos dos fármacos , Receptor A1 de Adenosina/metabolismo
4.
Neuropharmacology ; 205: 108924, 2022 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-34919904

RESUMO

Peripheral A1 adenosine receptor signaling has been shown to have analgesic effects in a variety of pain conditions. However, it is not yet fully elucidated for the precise molecular mechanisms. Acid sensing ion channels (ASICs) are expressed predominantly in nociceptive sensory neurons responding to protons. Given that both A1 adenosine receptors and ASICs are present in dorsal root ganglia (DRG) neurons, we therefore investigated whether there was a cross-talk between the two types of receptors. Herein, electrophysiological recordings showed that the A1 adenosine receptor agonist N6-cyclopentyladenosine (CPA) suppressed acid-induced currents and action potentials, which were mediated by ASICs, in rat DRG neurons. CPA inhibited the maximum response to protons, as shown a downward shift of concentration-response curve for protons. The CPA-induced suppression of ASIC currents was blocked by the A1 adenosine receptor antagonist KW-3902 and also prevented by intracellular application of the Gi/o-protein inhibitor pertussis toxin, the adenylate cyclase activator forskolin, and the cAMP analog 8-Br-cAMP. Finally, intraplantar pretreatment of CPA dose-dependently relieved acid-induced nociceptive responses in rats through peripheral A1 adenosine receptors. These results suggested that CPA suppressed ASICs via A1 adenosine receptors and intracellular Gi/o-proteins and cAMP signaling cascades in rat DRG neurons, which was a novel potential mechanism underlying analgesia of peripheral A1 adenosine receptors.


Assuntos
Canais Iônicos Sensíveis a Ácido/efeitos dos fármacos , Agonistas do Receptor A1 de Adenosina/farmacologia , Antagonistas do Receptor A1 de Adenosina/farmacologia , Analgesia , Fenômenos Eletrofisiológicos/efeitos dos fármacos , Gânglios Espinais/efeitos dos fármacos , Nociceptividade/efeitos dos fármacos , Nociceptores/efeitos dos fármacos , Receptor A1 de Adenosina/efeitos dos fármacos , Animais , Comportamento Animal/efeitos dos fármacos , Ratos
6.
Int J Mol Sci ; 22(22)2021 Nov 19.
Artigo em Inglês | MEDLINE | ID: mdl-34830353

RESUMO

The osmodiuretic agent Mannitol exerts cardioprotection against ischemia and reperfusion (I/R) injury when applied as a pre- and/or postconditioning stimulus. Previously, we demonstrated that these properties are mediated via the activation of mitochondrial ATP-sensitive potassium (mKATP) channels. However, considering Mannitol remains in the extracellular compartment, the question arises as to which receptor and intracellular signaling cascades are involved in myocardial protection by the osmodiuretic substance. Protein kinase B (Akt) and G (PKG), as part of the reperfusion injury salvage kinase (RISK) and/or endothelial nitric oxide (eNOS)/PKG pathway, are two well-investigated intracellular targets conferring myocardial protection upstream of mitochondrial potassium channels. Adenosine receptor subtypes have been shown to trigger different cardioprotective pathways, for example, the reperfusion injury. Further, Mannitol induces an increased activation of the adenosine 1 receptor (A1R) in renal cells conferring its nephroprotective properties. Therefore, we investigated whether (1) Akt and PKG are possible signaling targets involved in Mannitol-induced conditioning upstream of the mKATP channel and/or whether (2) cardioprotection by Mannitol is mediated via activation of the A1R. All experiments were performed on male Wistar rats in vitro employing the Langendorff isolated heart perfusion technique with infarct size determination as the primary endpoint. To unravel possible protein kinase activation, Mannitol was applied in combination with the Akt (MK2206) or PKG (KT5823) inhibitor. In further groups, an A1R blocker (DPCPX) was given with or without Mannitol. Preconditioning with Mannitol (Man) significantly reduced the infarct size compared to the control group. Co-administration of the A1R blocker DPXPC fully abolished myocardial protection of Mannitol. Interestingly and in contrast to the initial hypothesis, neither administration of the Akt nor the PKG blocker had any impact on the cardioprotective properties of Mannitol-induced preconditioning. These results are quite unexpected and show that the protein kinases Akt and PKG-as possible targets of known protective signaling cascades-are not involved in Mannitol-induced preconditioning. However, the cardioprotective effects of Mannitol are mediated via the A1R.


Assuntos
Proteínas Quinases Dependentes de GMP Cíclico/genética , Manitol/farmacologia , Proteínas Proto-Oncogênicas c-akt/genética , Receptor A1 de Adenosina/genética , Traumatismo por Reperfusão/tratamento farmacológico , Animais , Carbazóis/farmacologia , Proteínas Quinases Dependentes de GMP Cíclico/antagonistas & inibidores , Coração/efeitos dos fármacos , Coração/fisiopatologia , Compostos Heterocíclicos com 3 Anéis/farmacologia , Humanos , Precondicionamento Isquêmico Miocárdico , Canais KATP/antagonistas & inibidores , Rim/efeitos dos fármacos , Rim/patologia , Traumatismo por Reperfusão Miocárdica/tratamento farmacológico , Traumatismo por Reperfusão Miocárdica/genética , Traumatismo por Reperfusão Miocárdica/patologia , Miocárdio/metabolismo , Miocárdio/patologia , Óxido Nítrico Sintase Tipo III/genética , Proteínas Proto-Oncogênicas c-akt/antagonistas & inibidores , Ratos , Receptor A1 de Adenosina/efeitos dos fármacos , Traumatismo por Reperfusão/genética , Traumatismo por Reperfusão/patologia , Transdução de Sinais/efeitos dos fármacos , Xantinas/farmacologia
7.
Toxicol Appl Pharmacol ; 416: 115466, 2021 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-33631229

RESUMO

Soman, an organophosphorus (OP) compound, disrupts nervous system function through inactivation of acetylcholinesterase (AChE), the enzyme that breaks down acetylcholine at synapses. Left untreated, a state of prolonged seizure activity (status epilepticus, SE) is induced, causing widespread neuronal damage and associated cognitive and behavioral impairments. Previous research demonstrated that therapeutic stimulation of A1 adenosine receptors (A1ARs) can prevent or terminate soman-induced seizure. This study examined the ability of three potent A1AR agonists to provide neuroprotection and, ultimately, prevent observable cognitive and behavioral deficits following exposure to soman. Sprague Dawley rats were challenged with a seizure-inducing dose of soman (1.2 x LD50) and treated 1 min later with one of the following A1AR agonists: (6)-Cyclopentyladenosine (CPA), 2-Chloro-N6-cyclopentyladenosine (CCPA) or N-bicyclo(2.2.1)hept-2-yl-5'-chloro-5'-deoxyadenosine (cdENBA). An active avoidance shuttle box task was used to evaluate locomotor responses to aversive stimuli at 3, 7 and 14 days post-exposure. Animals treated with CPA, CCPA or cdENBA demonstrated a higher number of avoidance responses and a faster reaction to the aversive stimulus than the soman/saline control group across all three sessions. Findings suggest that A1AR agonism is a promising neuroprotective countermeasure, capable of preventing the long-term deficits in learning and memory that are characteristic of soman intoxication.


Assuntos
Agonistas do Receptor A1 de Adenosina/farmacologia , Anticonvulsivantes/farmacologia , Encéfalo/efeitos dos fármacos , Fármacos Neuroprotetores/farmacologia , Intoxicação por Organofosfatos/prevenção & controle , Receptor A1 de Adenosina/efeitos dos fármacos , Convulsões/prevenção & controle , Soman , Adenosina/análogos & derivados , Adenosina/farmacologia , Animais , Comportamento Animal/efeitos dos fármacos , Encéfalo/metabolismo , Encéfalo/patologia , Desoxiadenosinas/farmacologia , Modelos Animais de Doenças , Masculino , Intoxicação por Organofosfatos/etiologia , Intoxicação por Organofosfatos/metabolismo , Intoxicação por Organofosfatos/fisiopatologia , Ratos Sprague-Dawley , Receptor A1 de Adenosina/metabolismo , Convulsões/induzido quimicamente , Convulsões/metabolismo , Convulsões/patologia
8.
Eur J Pharmacol ; 897: 173946, 2021 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-33607106

RESUMO

Metaplasticity is referred to adjustment in the requirements for induction of synaptic plasticity based on the prior history of activity. Synaptic plasticity, including long-term potentiation (LTP) and long-term depression (LTD), has been considered to be the neural processes underlying learning and memory. Previous observations that cordycepin (an adenosine derivative) improved learning and memory seemed to be contradictory to the findings that cordycepin inhibited LTP. Therefore, we speculated that the conflicting reports of cordycepin might be related to metaplasticity. In the current study, population spike (PS) in hippocampal CA1 area of rats was recorded by using electrophysiological method in vivo. The results showed that cordycepin reduced PS amplitude in hippocampal CA1 with a concentration-dependent relationship, and high frequency stimulation (HFS) failed to induce LTP when cordycepin was intrahippocampally administrated but improved LTP magnitude when cordycepin was pre-treated. Cordycepin increased LTD induced by activating N-Methyl-D-aspartate (NMDA) receptors and subsequently facilitated LTP induced by HFS. Furthermore, we found that 1,3-dipropyl-8-cyclopentylxanthine (DPCPX), an adenosine A1 receptors antagonist, could block the roles of cordycepin on LTD and LTP. Collectively, cordycepin was able to modulate metaplasticity in hippocampal CA1 area of rats through adenosine A1 receptors. These findings would be helpful to reconcile the conflicting reports in the literatures and provided new insights into the mechanisms underlying cognitive function promotions with cordycepin treatment.


Assuntos
Agonistas do Receptor A1 de Adenosina/farmacologia , Região CA1 Hipocampal/efeitos dos fármacos , Desoxiadenosinas/farmacologia , Plasticidade Neuronal/efeitos dos fármacos , Receptor A1 de Adenosina/efeitos dos fármacos , Potenciais de Ação/efeitos dos fármacos , Animais , Região CA1 Hipocampal/metabolismo , Potenciação de Longa Duração/efeitos dos fármacos , Depressão Sináptica de Longo Prazo/efeitos dos fármacos , Masculino , Ratos Sprague-Dawley , Receptor A1 de Adenosina/metabolismo , Receptores de N-Metil-D-Aspartato/metabolismo , Fatores de Tempo
9.
Purinergic Signal ; 17(2): 247-254, 2021 06.
Artigo em Inglês | MEDLINE | ID: mdl-33548045

RESUMO

6-Hydroxydopamine (6-OHDA) is the most used toxin in experimental Parkinson's disease (PD) models. 6-OHDA shows high affinity for the dopamine transporter and once inside the neuron, it accumulates and undergoes non-enzymatic auto-oxidation, promoting reactive oxygen species (ROS) formation and selective damage of catecholaminergic neurons. In this way, our group has established a 6-OHDA in vitro protocol with rat striatal slices as a rapid and effective model for screening of new drugs with protective effects against PD. We have shown that co-incubation with guanosine (GUO, 100 µM) prevented the 6-OHDA-induced damage in striatal slices. As the exact GUO mechanism of action remains unknown, the aim of this study was to investigate if adenosine A1 (A1R) and/or A2A receptors (A2AR) are involved on GUO protective effects on striatal slices. Pre-incubation with DPCPX, an A1R antagonist prevented guanosine effects on 6-OHDA-induced ROS formation and mitochondrial membrane potential depolarization, while CCPA, an A1R agonist, did not alter GUO effects. Regarding A2AR, the antagonist SCH58261 had similar protective effect as GUO in ROS formation and mitochondrial membrane potential. Additionally, SCH58261 did not affect GUO protective effects. The A2AR agonist CGS21680, although, completely blocked GUO effects. Finally, the A1R antagonist DPCPX, and the A2AR agonist CGS21680 also abolished the preventive guanosine effect on 6-OHDA-induced ATP levels decrease. These results reinforce previous evidence for a putative interaction of GUO with A1R-A2AR heteromer as its molecular target and clearly indicate a dependence on adenosine receptors modulation to GUO protective effect.


Assuntos
Guanosina/farmacologia , Doenças Mitocondriais/prevenção & controle , Neostriado/metabolismo , Fármacos Neuroprotetores/farmacologia , Oxidopamina/toxicidade , Receptor A1 de Adenosina/efeitos dos fármacos , Receptor A2A de Adenosina/efeitos dos fármacos , Explosão Respiratória/efeitos dos fármacos , Antagonistas do Receptor A1 de Adenosina/farmacologia , Animais , Avaliação Pré-Clínica de Medicamentos , Técnicas In Vitro , Masculino , Potencial da Membrana Mitocondrial/efeitos dos fármacos , Neostriado/efeitos dos fármacos , Ratos , Ratos Wistar , Espécies Reativas de Oxigênio/metabolismo , Xantinas/uso terapêutico
10.
Sci Rep ; 10(1): 14181, 2020 08 25.
Artigo em Inglês | MEDLINE | ID: mdl-32843670

RESUMO

Glial fibrillary acidic protein expressing (GFAP+) glia modulate nociceptive neuronal activity in both the peripheral nervous system (PNS) and the central nervous system (CNS). Resident GFAP+ glia in dorsal root ganglia (DRG) known as satellite glial cells (SGCs) potentiate neuronal activity by releasing pro-inflammatory cytokines and neuroactive compounds. In this study, we tested the hypothesis that SGC Gq-coupled receptor (Gq-GPCR) signaling modulates pain sensitivity in vivo using Gfap-hM3Dq mice. Complete Freund's adjuvant (CFA) was used to induce inflammatory pain, and mechanical sensitivity and thermal sensitivity were used to assess the neuromodulatory effect of glial Gq-GPCR activation in awake mice. Pharmacogenetic activation of Gq-GPCR signaling in sensory SGCs decreased heat-induced nociceptive responses and reversed inflammation-induced mechanical allodynia via peripheral adenosine A1 receptor activation. These data reveal a previously unexplored role of sensory SGCs in decreasing afferent excitability. The identified molecular mechanism underlying the analgesic role of SGCs offers new approaches for reversing peripheral nociceptive sensitization.


Assuntos
Subunidades alfa Gq-G11 de Proteínas de Ligação ao GTP/fisiologia , Hiperalgesia/prevenção & controle , Inflamação/fisiopatologia , Neuroglia/enzimologia , Nociceptividade/fisiologia , Receptor A1 de Adenosina/fisiologia , Receptor Muscarínico M3/fisiologia , Animais , Benzilatos/farmacologia , Clozapina/análogos & derivados , Clozapina/farmacologia , Adjuvante de Freund/toxicidade , Genes Sintéticos , Temperatura Alta , Hiperalgesia/fisiopatologia , Inflamação/induzido quimicamente , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Agonistas Muscarínicos/farmacologia , Neuroglia/fisiologia , Nortropanos/farmacologia , Regiões Promotoras Genéticas , Agonistas do Receptor Purinérgico P1/farmacologia , Antagonistas de Receptores Purinérgicos P1/farmacologia , Receptor A1 de Adenosina/efeitos dos fármacos , Receptor Muscarínico M3/efeitos dos fármacos , Receptor Muscarínico M3/genética , Receptores Acoplados a Proteínas G , Proteínas Recombinantes de Fusão/efeitos dos fármacos , Proteínas Recombinantes de Fusão/metabolismo , Teofilina/análogos & derivados , Teofilina/farmacologia , Tato , Xantinas/farmacologia
11.
Behav Pharmacol ; 31(6): 544-552, 2020 09.
Artigo em Inglês | MEDLINE | ID: mdl-32701527

RESUMO

This study aimed to investigate the possible gamma-decanolactone mechanisms of action in the GABAergic and adenosine systems using the aminophylline-induced acute crisis model and the pentylenetetrazole-induced kindling model. In the acute model, male mice received administration of bicuculline (GABAA receptor antagonist), 8-cyclopentyl-1,3-dipropylxanthine (A1 receptor antagonist) or ZM241385 (A2A receptor antagonist), 15 min before the treatment with gamma-decanolactone (300 mg/kg). After a single dose of aminophylline was administered, the animals were observed for 60 min. In the chronic model of seizure, 30 min after the treatment with gamma-decanolactone, mice received pentylenetetrazole once every third day. On the last day of kindling, the animals received the same GABA and adenosine antagonists used in the acute model, 15 min before gamma-decanolactone administration. The protein expression of GABAA α1 receptor and adenosine A1 receptor was detected using western blotting technique in hippocampal samples. The results showed that gamma-decanolactone increased the latency to first seizure and decreased seizure occurrence in the acute and chronic models. The adenosine A2A receptor antagonist and GABAA receptor antagonist were not able to change gamma-decanolactone behavioral seizure induced by aminophylline or pentylenetetrazole. The administration of adenosine A1 receptor antagonist reversed the protective effect of gamma-decanolactone in both models. In addition, gamma-decanolactone promoted an increase in the expression GABAA α1 receptor, in the hippocampus. The results suggest that the neuroprotective effect of gamma-decanolactone observed during the investigation could have a straight connection to its action on A1 adenosine receptors.


Assuntos
Lactonas/farmacologia , Fármacos Neuroprotetores/farmacologia , Receptor A1 de Adenosina/fisiologia , Convulsões/tratamento farmacológico , Doença Aguda , Animais , Doença Crônica , Modelos Animais de Doenças , Lactonas/uso terapêutico , Masculino , Camundongos , Receptor A1 de Adenosina/efeitos dos fármacos , Receptores de GABA/fisiologia
12.
Neurosci Lett ; 730: 135016, 2020 06 21.
Artigo em Inglês | MEDLINE | ID: mdl-32371159

RESUMO

Alzheimer's disease (AD) is the most common cause of dementia, characterised by advanced cognitive and memory deterioration with no effective treatments available. Previous in vitro and in vivo studies suggest that paeoniflorin (PF), a major bioactive constituent of Radix Paeoniae, might possess anti-dementia properties; however, the underlying mechanism remains unclear. The aim of the current study was to determine the therapeutic effects of PF in a transgenic mouse model of AD and to identify its mechanism. Transgenic mice with five familial AD mutations (5XFAD) were used in this study. We showed that 28 days of PF (5 mg/kg, ip) treatment significantly decreased the escape latency and path length in the Morris water maze test and increased the alternation rate in the T-maze test, compared to the vehicle treatment group. In addition, PF treatment significantly alleviated amyloid ß plaque burden, inhibited astrocyte activation, and decreased IL-1ß and TNF-α expression in the brain of 5XFAD mice. However, the anti-cognitive deficits, anti-amyloidogenic, and anti-inflammatory effects of PF were abolished by 1,3-dipropyl-8-cyclopentylxanthine (DPCPX, 0.3 mg/kg), an adenosine A1 receptor (A1R) antagonist. In conclusion, our results suggest that PF might act as a potential therapeutic agent for AD via activation of adenosine A1R.


Assuntos
Doença de Alzheimer/tratamento farmacológico , Glucosídeos/farmacologia , Memória/efeitos dos fármacos , Monoterpenos/farmacologia , Fármacos Neuroprotetores/farmacologia , Receptor A1 de Adenosina/efeitos dos fármacos , Adenosina/farmacologia , Doença de Alzheimer/metabolismo , Precursor de Proteína beta-Amiloide/genética , Animais , Modelos Animais de Doenças , Camundongos Transgênicos
13.
Purinergic Signal ; 16(2): 231-240, 2020 06.
Artigo em Inglês | MEDLINE | ID: mdl-32440820

RESUMO

Exosomes, small-sized extracellular vesicles, carry components of the purinergic pathway. The production by cells of exosomes carrying this pathway remains poorly understood. Here, we asked whether type 1, 2A, or 2B adenosine receptors (A1Rs, A2ARs, and A2BRs, respectively) expressed by producer cells are involved in regulating exosome production. Preglomerular vascular smooth muscle cells (PGVSMCs) were isolated from wildtype, A1R-/-, A2AR-/-, and A2BR-/- rats, and exosome production was quantified under normal or metabolic stress conditions. Exosome production was also measured in various cancer cells treated with pharmacologic agonists/antagonists of A1Rs, A2ARs, and A2BRs in the presence or absence of metabolic stress or cisplatin. Functional activity of exosomes was determined in Jurkat cell apoptosis assays. In PGVSMCs, A1R and A2AR constrained exosome production under normal conditions (p = 0.0297; p = 0.0409, respectively), and A1R, A2AR, and A2BR constrained exosome production under metabolic stress conditions. Exosome production from cancer cells was reduced (p = 0.0028) by the selective A2AR agonist CGS 21680. These exosomes induced higher levels of Jurkat apoptosis than exosomes from untreated cells or cells treated with A1R and A2BR agonists (p = 0.0474). The selective A2AR antagonist SCH 442416 stimulated exosome production under metabolic stress or cisplatin treatment, whereas the selective A2BR antagonist MRS 1754 reduced exosome production. Our findings indicate that A2ARs suppress exosome release in all cell types examined, whereas effects of A1Rs and A2BRs are dependent on cell type and conditions. Pharmacologic targeting of cancer with A2AR antagonists may inadvertently increase exosome production from tumor cells.


Assuntos
Agonistas do Receptor A2 de Adenosina/farmacologia , Exossomos/efeitos dos fármacos , Receptor A1 de Adenosina/metabolismo , Receptor A2A de Adenosina/metabolismo , Adenosina/análogos & derivados , Adenosina/farmacologia , Animais , Exossomos/metabolismo , Masculino , Fenetilaminas/farmacologia , Ratos , Receptor A1 de Adenosina/efeitos dos fármacos , Receptor A2A de Adenosina/efeitos dos fármacos , Células Tumorais Cultivadas/metabolismo
14.
Am J Physiol Renal Physiol ; 318(4): F870-F877, 2020 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-31984792

RESUMO

Adenosine plays an important role in various aspects of kidney physiology, but the specific targets and mechanisms of actions are not completely understood. The collecting duct has the highest expression of adenosine receptors, particularly adenosine A1 receptors (A1Rs). Interstitial adenosine levels are greatly increased up to a micromolar range in response to dietary salt loading. We have previously shown that the basolateral membrane of principal cells has primarily K+ conductance mediated by Kir4.1/5.1 channels to mediate K+ recycling and to set up a favorable driving force for Na+/K+ exchange (47). Intercalated cells express the Cl- ClC-K2/b channel mediating transcellular Cl- reabsorption. Using patch-clamp electrophysiology in freshly isolated mouse collecting ducts, we found that acute application of adenosine reversely inhibits ClC-K2/b open probability from 0.31 ± 0.04 to 0.17 ± 0.06 and to 0.10 ± 0.05 for 1 and 10 µM, respectively. In contrast, adenosine (10 µM) had no measureable effect on Kir4.1/5.1 channel activity in principal cells. The inhibitory effect of adenosine on ClC-K2/b was abolished in the presence of the A1R blocker 8-cyclopentyl-1,3-dipropylxanthine (10 µM). Consistently, application of the A1R agonist N6-cyclohexyladenosine (1 µM) recapitulated the inhibitory action of adenosine on ClC-K2/b open probability. The effects of adenosine signaling in the collecting duct were independent from its purinergic counterpartner, ATP, having no measurable actions on ClC-K2/b and Kir4.1/5.1. Overall, we demonstrated that adenosine selectively inhibits ClC-K2/b activity in intercalated cells by targeting A1Rs. We propose that inhibition of transcellular Cl- reabsorption in the collecting duct by adenosine would aid in augmenting NaCl excretion during high salt intake.


Assuntos
Agonistas do Receptor A1 de Adenosina/farmacologia , Adenosina/farmacologia , Proteínas de Transporte de Ânions/antagonistas & inibidores , Canais de Cloreto/antagonistas & inibidores , Cloretos/metabolismo , Túbulos Renais Coletores/efeitos dos fármacos , Receptor A1 de Adenosina/efeitos dos fármacos , Reabsorção Renal/efeitos dos fármacos , Animais , Proteínas de Transporte de Ânions/metabolismo , Sinalização do Cálcio/efeitos dos fármacos , Células Cultivadas , Canais de Cloreto/metabolismo , Túbulos Renais Coletores/citologia , Túbulos Renais Coletores/metabolismo , Masculino , Potenciais da Membrana/efeitos dos fármacos , Camundongos Endogâmicos C57BL , Receptor A1 de Adenosina/metabolismo
15.
SLAS Discov ; 25(2): 186-194, 2020 02.
Artigo em Inglês | MEDLINE | ID: mdl-31583945

RESUMO

Receptor internalization in response to prolonged agonist treatment is an important regulator of G protein-coupled receptor (GPCR) function. The adenosine A1 receptor (A1AR) is one of the adenosine receptor family of GPCRs, and evidence for its agonist-induced internalization is equivocal. The recently developed NanoBiT technology uses split NanoLuc Luciferase to monitor changes in protein interactions. We have modified the human A1AR on the N-terminus with the small high-affinity HiBiT tag. In the presence of the large NanoLuc subunit (LgBiT), complementation occurs, reconstituting a full-length functional NanoLuc Luciferase. Here, we have used complemented luminescence to monitor the internalization of the A1AR in living HEK293 cells. Agonist treatment resulted in a robust decrease in cell-surface luminescence, indicating an increase in A1AR internalization. These responses were inhibited by the A1AR-selective antagonist 1,3-dipropyl-8-cyclopentylxanthine (DPCPX), with an antagonist affinity that closely matched that measured using ligand binding with a fluorescent A1 receptor antagonist (CA200645). The agonist potencies for inducing A1AR internalization were very similar to the affinities previously determined by ligand binding, suggesting little or no amplification of the internalization response. By complementing the HiBiT tag to exogenous purified LgBiT, it was also possible to perform NanoBRET ligand-binding experiments using HiBiT-A1AR. This study demonstrates the use of NanoBiT technology to monitor internalization of the A1AR and offers the potential to combine these experiments with NanoBRET ligand-binding assays.


Assuntos
Adenosina/genética , Receptor A1 de Adenosina/genética , Receptores Acoplados a Proteínas G/genética , Adenosina/química , Agonistas do Receptor A1 de Adenosina/farmacologia , Células HEK293 , Humanos , Cinética , Ligantes , Mapas de Interação de Proteínas/efeitos dos fármacos , Mapas de Interação de Proteínas/genética , Receptor A1 de Adenosina/efeitos dos fármacos , Receptores Acoplados a Proteínas G/agonistas , Xantinas/farmacologia
16.
Life Sci ; 240: 117068, 2020 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-31751583

RESUMO

AIMS: Bradycardia contributes to tachy-brady arrhythmias or sinus arrest during heart failure (HF). Sinoatrial node (SAN) adenosine A1 receptors (ADO A1Rs) are upregulated in HF, and adenosine is known to exert negative chronotropic effects on the SAN. Here, we investigated the role of A1R signaling at physiologically relevant ADO concentrations on HF SAN pacemaker cells. MAIN METHODS: Dogs with tachypacing-induced chronic HF and normal controls (CTL) were studied. SAN tissue was collected for A1R and GIRK mRNA quantification. SAN cells were isolated for perforated patch clamp recordings and firing rate (bpm), slope of slow diastolic depolarization (SDD), and maximum diastolic potential (MDP) were measured. Action potentials (APs) and currents were recorded before and after addition of 1 and 10 µM ADO. To assess contributions of A1R and G protein-coupled Inward Rectifier Potassium Current (GIRK) to ADO effects, APs were measured after the addition of DPCPX (selective A1R antagonist) or TPQ (selective GIRK blocker). KEY FINDINGS: A1R and GIRK mRNA expression were significantly increased in HF. In addition, ADO induced greater rate slowing and membrane hyperpolarization in HF vs CTL (p < 0.05). DPCPX prevented ADO-induced rate slowing in CTL and HF cells. The ADO-induced inward rectifying current, IKado, was observed significantly more frequently in HF than in CTL. TPQ prevented ADO-induced rate slowing in HF. SIGNIFICANCE: An increase in A1R and GIRK expression enhances IKAdo, causing hyperpolarization, and subsequent negative chronotropic effects in canine chronic HF at relevant [ADO]. GIRK blockade may be a useful strategy to mitigate bradycardia in HF.


Assuntos
Agonistas do Receptor A1 de Adenosina/farmacologia , Adenosina/farmacologia , Canais de Potássio Corretores do Fluxo de Internalização Acoplados a Proteínas G/agonistas , Insuficiência Cardíaca/fisiopatologia , Frequência Cardíaca/efeitos dos fármacos , Receptor A1 de Adenosina/metabolismo , Nó Sinoatrial/citologia , Nó Sinoatrial/efeitos dos fármacos , Potenciais de Ação/efeitos dos fármacos , Antagonistas do Receptor A1 de Adenosina/farmacologia , Animais , Venenos de Abelha/farmacologia , Relógios Biológicos , Doença Crônica , Cães , Feminino , Canais de Potássio Corretores do Fluxo de Internalização Acoplados a Proteínas G/antagonistas & inibidores , Canais de Potássio Corretores do Fluxo de Internalização Acoplados a Proteínas G/efeitos dos fármacos , Técnicas In Vitro , Masculino , Técnicas de Patch-Clamp , Bloqueadores dos Canais de Potássio/farmacologia , Receptor A1 de Adenosina/efeitos dos fármacos , Xantinas/farmacologia
17.
Br J Pharmacol ; 177(2): 346-359, 2020 01.
Artigo em Inglês | MEDLINE | ID: mdl-31596949

RESUMO

BACKGROUND AND PURPOSE: Adenosine is a local mediator that regulates physiological and pathological processes via activation of four GPCRs (A1 , A2A , A2B , and A3 ). We have investigated the effect of two A1 -receptor-selective agonists and the novel A1 -receptor bitopic ligand VCP746 on the rat cardiovascular system. EXPERIMENTAL APPROACH: The regional haemodynamic responses of these agonist was investigated in conscious rats. Male Sprague-Dawley rats (350-450 g) were chronically implanted with pulsed Doppler flow probes on the renal, mesenteric arteries and the descending abdominal aorta and the jugular vein and caudal artery catheterized. Cardiovascular responses were measured following intravenous infusion (3 min each dose) of CCPA (120, 400, and 1,200 ng·kg-1 ·min-1 ), capadenoson or adenosine (30, 100, and 300 µg·kg-1 ·min-1 ), or VCP746 (6, 20, and 60 µg·kg-1 ·min-1 ) following pre-dosing with DPCPX (0.1 mg·kg-1 , i.v.) or vehicle. KEY RESULTS: CCPA produced a significant A1 -receptor-mediated decrease in heart rate that was accompanied by vasoconstrictions in the renal and mesenteric vascular beds but an increase in hindquarters vascular conductance. The partial agonist capadenoson also produced an A1 -receptor-mediated bradycardia. In contrast, VCP746 produced increases in heart rate and renal and mesenteric vascular conductance that were not mediated by A1 -receptors. In vitro studies confirmed that VCP746 had potent agonist activity at both A2A - and A2B -receptors. CONCLUSIONS AND IMPLICATIONS: These results suggest VCP746 mediates its cardiovascular effects via activation of A2 rather than A1 adenosine receptors. This has implications for the design of future bitopic ligands that incorporate A1 allosteric ligand moieties.


Assuntos
Agonistas do Receptor A1 de Adenosina/farmacologia , Agonistas do Receptor A2 de Adenosina/farmacologia , Adenosina/análogos & derivados , Sistema Cardiovascular/efeitos dos fármacos , Hemodinâmica/efeitos dos fármacos , Receptor A1 de Adenosina/efeitos dos fármacos , Tiofenos/farmacologia , Adenosina/farmacologia , Aminopiridinas/farmacologia , Animais , Sistema Cardiovascular/metabolismo , Estado de Consciência , Agonismo Parcial de Drogas , Frequência Cardíaca/efeitos dos fármacos , Ligantes , Masculino , Ratos Sprague-Dawley , Receptor A1 de Adenosina/metabolismo , Receptor A2A de Adenosina/efeitos dos fármacos , Receptor A2A de Adenosina/metabolismo , Receptor A2B de Adenosina/efeitos dos fármacos , Receptor A2B de Adenosina/metabolismo , Fluxo Sanguíneo Regional/efeitos dos fármacos , Tiazóis/farmacologia
18.
Artigo em Inglês | MEDLINE | ID: mdl-31707092

RESUMO

Longitudinal and some experimental studies have showed the potential of caffeine to counteract some depressive behaviors and synaptic dysfunctions. In this study, we investigated the potential of caffeine in preventing behavioral outcomes, neurodegeneration and synaptic proteins alterations in a mice model of agitated depression by bilateral olfactory bulbectomy (OB). For this purpose, bulbectomized mice received caffeine (0.3 g/L and 1.0 g/L, drinking water), during the active cycle, for seven weeks (two before the surgery and throughout five weeks after OB). Caffeine prevented OB-induced hyperactivity and recognition memory impairment and rescue self care and motivational behavior. In the frontal cortex, bulbectomized mice presented increase in the adenosine A1 receptors (A1R) and GFAP, while adenosine A2A receptors (A2AR) increased in the hippocampus and striatum and SNAP-25 was decreased in frontal cortex and striatum. Caffeine increased A1R in the striatum of bulbectomized mice and in SHAM-water group caffeine increased A2AR in the striatum and decreased SNAP-25 in the frontal cortex. Astrogliosis observed in the polymorphic layer of the dentate gyrus of OB mice was prevented by caffeine as well as the neurodegeneration in the striatum and piriform cortex. Based on these behavioral and neurochemical evidences, caffeine confirms its efficacy in preventing neurodegeneration associated with memory impairment and may be considered as a promising therapeutic tool in the prophylaxis and/or treatment of depression.


Assuntos
Cafeína/uso terapêutico , Estimulantes do Sistema Nervoso Central/uso terapêutico , Depressão/prevenção & controle , Depressão/psicologia , Doenças Neurodegenerativas/prevenção & controle , Agitação Psicomotora/prevenção & controle , Agitação Psicomotora/psicologia , Animais , Comportamento Animal/efeitos dos fármacos , Encéfalo/patologia , Gliose/patologia , Masculino , Transtornos da Memória/prevenção & controle , Transtornos da Memória/psicologia , Camundongos , Doenças Neurodegenerativas/patologia , Bulbo Olfatório , Receptor A1 de Adenosina/efeitos dos fármacos , Receptor A2A de Adenosina/efeitos dos fármacos , Reconhecimento Psicológico/efeitos dos fármacos , Proteína 25 Associada a Sinaptossoma/metabolismo
19.
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
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA