Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 40
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Purinergic Signal ; 17(3): 503-514, 2021 09.
Artigo em Inglês | MEDLINE | ID: mdl-34313915

RESUMO

Previous studies suggest that adenosine A1 receptors (A1R) modulate the processing of pain. The aim of this study was to characterize the distribution of A1R in nociceptive tissues and to evaluate whether targeting A1R with the partial agonist capadenoson may reduce neuropathic pain in mice. The cellular distribution of A1R in dorsal root ganglia (DRG) and the spinal cord was analyzed using fluorescent in situ hybridization. In behavioral experiments, neuropathic pain was induced by spared nerve injury or intraperitoneal injection of paclitaxel, and tactile hypersensitivities were determined using a dynamic plantar aesthesiometer. Whole-cell patch-clamp recordings were performed to assess electrophysiological properties of dissociated DRG neurons. We found A1R to be expressed in populations of DRG neurons and dorsal horn neurons involved in the processing of pain. However, administration of capadenoson at established in vivo doses (0.03-1.0 mg/kg) did not alter mechanical hypersensitivity in the spared nerve injury and paclitaxel models of neuropathic pain, whereas the standard analgesic pregabalin significantly inhibited the pain behavior. Moreover, capadenoson failed to affect potassium currents in DRG neurons, in contrast to a full A1R agonist. Despite expression of A1R in nociceptive neurons, our data do not support the hypothesis that pharmacological intervention with partial A1R agonists might be a valuable approach for the treatment of neuropathic pain.


Assuntos
Agonistas do Receptor A1 de Adenosina/uso terapêutico , Neuralgia/tratamento farmacológico , Neuralgia/metabolismo , Receptor A1 de Adenosina/biossíntese , Agonistas do Receptor A1 de Adenosina/farmacologia , Animais , Células Cultivadas , Feminino , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Medição da Dor/efeitos dos fármacos , Medição da Dor/métodos , Receptor A1 de Adenosina/genética , Resultado do Tratamento
2.
Circulation ; 134(6): 486-98, 2016 Aug 09.
Artigo em Inglês | MEDLINE | ID: mdl-27462069

RESUMO

BACKGROUND: Adenosine provokes atrial fibrillation (AF) with a higher activation frequency in right atria (RA) versus left atria (LA) in patients, but the underlying molecular and functional substrates are unclear. We tested the hypothesis that adenosine-induced AF is driven by localized reentry in RA areas with highest expression of adenosine A1 receptor and its downstream GIRK (G protein-coupled inwardly rectifying potassium channels) channels (IK,Ado). METHODS: We applied biatrial optical mapping and immunoblot mapping of various atrial regions to reveal the mechanism of adenosine-induced AF in explanted failing and nonfailing human hearts (n=37). RESULTS: Optical mapping of coronary-perfused atria (n=24) revealed that adenosine perfusion (10-100 µmol/L) produced more significant shortening of action potential durations in RA (from 290±45 to 239±41 ms, 17.3±10.4%; P<0.01) than LA (from 307±24 to 286±23 ms, 6.7±6.6%; P<0.01). In 10 hearts, adenosine induced AF (317±116 s) that, when sustained (≥2 minutes), was primarily maintained by 1 to 2 localized reentrant drivers in lateral RA. Tertiapin (10-100 nmol/L), a selective GIRK channel blocker, counteracted adenosine-induced action potential duration shortening and prevented AF induction. Immunoblotting showed that the superior/middle lateral RA had significantly higher adenosine A1 receptor (2.7±1.7-fold; P<0.01) and GIRK4 (1.7±0.8-fold; P<0.05) protein expression than lateral/posterior LA. CONCLUSIONS: This study revealed a 3-fold RA-to-LA adenosine A1 receptor protein expression gradient in the human heart, leading to significantly greater RA versus LA repolarization sensitivity in response to adenosine. Sustained adenosine-induced AF is maintained by reentrant drivers localized in lateral RA regions with the highest adenosine A1 receptor/GIRK4 expression. Selective atrial GIRK channel blockade may effectively treat AF during conditions with increased endogenous adenosine.


Assuntos
Adenosina/toxicidade , Fibrilação Atrial/induzido quimicamente , Fibrilação Atrial/metabolismo , Canais de Potássio Corretores do Fluxo de Internalização Acoplados a Proteínas G/biossíntese , Átrios do Coração/metabolismo , Receptor A1 de Adenosina/biossíntese , Adulto , Idoso , Feminino , Regulação da Expressão Gênica , Coração/diagnóstico por imagem , Coração/efeitos dos fármacos , Átrios do Coração/diagnóstico por imagem , Átrios do Coração/efeitos dos fármacos , Sistema de Condução Cardíaco/diagnóstico por imagem , Sistema de Condução Cardíaco/efeitos dos fármacos , Sistema de Condução Cardíaco/metabolismo , Humanos , Masculino , Pessoa de Meia-Idade , Técnicas de Cultura de Órgãos , Tomografia por Emissão de Pósitrons combinada à Tomografia Computadorizada
3.
Neurochem Res ; 41(7): 1774-83, 2016 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-27038930

RESUMO

Level of adenosine, an endogenous astrocyte-based neuromodulator, is primarily regulated by adenosine P1 receptors. This study assessed expression of adenosine P1 receptors, ADORA1 (adenosine A1 receptor) and ADORA2A (adenosine A2a receptor) and their association with glioma development and epilepsy in glioma patients. Expression of ADORA1/ADORA2A was assessed immunohistochemically in 65 surgically removed glioma tissue and 21 peri-tumor tissues and 8 cases of normal brain tissues obtained from hematoma patients with cerebral trauma. Immunofluorescence, Western blot, and qRT-PCR were also used to verify immunohistochemical data. Adenosine P1 receptor ADORA1 and ADORA2A proteins were localized in the cell membrane and cytoplasm and ADORA1/ADORA2A immunoreactivity was significantly stronger in glioma and peri-tumor tissues that contained infiltrating tumor cells than in normal brain tissues (p < 0.05). The World Health Organization (WHO) grade III gliomas expressed even higher level of ADORA1 and ADORA2A. Western blot and qRT-PCR confirmed immunohistochemical data. Moreover, higher levels of ADORA1 and ADORA2A expression occurred in high-grade gliomas, in which incidence of epilepsy were lower (p < 0.05). In contrast, a lower level of ADORA1/ADORA2A expression was found in peri-tumor tissues with tumor cell presence from patients with epilepsy compared to patients without epilepsy (p < 0.05). The data from the current study indicates that dysregulation in ADORA1/ADORA2A expression was associated with glioma development, whereas low level of ADORA1/ADORA2A expression could increase susceptibility of tumor-associated epilepsy.


Assuntos
Neoplasias Encefálicas/metabolismo , Epilepsia/metabolismo , Regulação Neoplásica da Expressão Gênica , Glioma/metabolismo , Receptor A1 de Adenosina/biossíntese , Receptor A2A de Adenosina/biossíntese , Adolescente , Adulto , Idoso , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/patologia , Criança , Epilepsia/genética , Epilepsia/patologia , Feminino , Glioma/genética , Glioma/patologia , Humanos , Masculino , Pessoa de Meia-Idade , Receptor A1 de Adenosina/genética , Receptor A2A de Adenosina/genética , Adulto Jovem
4.
Circulation ; 130(4): 315-24, 2014 Jul 22.
Artigo em Inglês | MEDLINE | ID: mdl-24838362

RESUMO

BACKGROUND: Although sinoatrial node (SAN) dysfunction is a hallmark of human heart failure (HF), the underlying mechanisms remain poorly understood. We aimed to examine the role of adenosine in SAN dysfunction and tachy-brady arrhythmias in chronic HF. METHODS AND RESULTS: We applied multiple approaches to characterize SAN structure, SAN function, and adenosine A1 receptor expression in control (n=17) and 4-month tachypacing-induced chronic HF (n=18) dogs. Novel intramural optical mapping of coronary-perfused right atrial preparations revealed that adenosine (10 µmol/L) markedly prolonged postpacing SAN conduction time in HF by 206 ± 99 milliseconds (versus 66 ± 21 milliseconds in controls; P=0.02). Adenosine induced SAN intranodal conduction block or microreentry in 6 of 8 dogs with HF versus 0 of 7 controls (P=0.007). Adenosine-induced SAN conduction abnormalities and automaticity depression caused postpacing atrial pauses in HF versus control dogs (17.1 ± 28.9 versus 1.5 ± 1.3 seconds; P<0.001). Furthermore, 10 µmol/L adenosine shortened atrial repolarization and led to pacing-induced atrial fibrillation in 6 of 7 HF versus 0 of 7 control dogs (P=0.002). Adenosine-induced SAN dysfunction and atrial fibrillation were abolished or prevented by adenosine A1 receptor antagonists (50 µmol/L theophylline/1 µmol/L 8-cyclopentyl-1,3-dipropylxanthine). Adenosine A1 receptor protein expression was significantly upregulated during HF in the SAN (by 47 ± 19%) and surrounding atrial myocardium (by 90 ± 40%). Interstitial fibrosis was significantly increased within the SAN in HF versus control dogs (38 ± 4% versus 23 ± 4%; P<0.001). CONCLUSIONS: In chronic HF, adenosine A1 receptor upregulation in SAN pacemaker and atrial cardiomyocytes may increase cardiac sensitivity to adenosine. This effect may exacerbate conduction abnormalities in the structurally impaired SAN, leading to SAN dysfunction, and potentiate atrial repolarization shortening, thereby facilitating atrial fibrillation. Atrial fibrillation may further depress SAN function and lead to tachy-brady arrhythmias in HF.


Assuntos
Bradicardia/fisiopatologia , Insuficiência Cardíaca/fisiopatologia , Receptor A1 de Adenosina/biossíntese , Nó Sinoatrial/fisiopatologia , Taquicardia/fisiopatologia , Imagens com Corantes Sensíveis à Voltagem/métodos , Potenciais de Ação/efeitos dos fármacos , Adenosina/administração & dosagem , Adenosina/farmacologia , Adenosina/toxicidade , Antagonistas do Receptor A1 de Adenosina/farmacologia , Antagonistas do Receptor A1 de Adenosina/uso terapêutico , Animais , Fibrilação Atrial/etiologia , Fibrilação Atrial/fisiopatologia , Bradicardia/etiologia , Estimulação Cardíaca Artificial/efeitos adversos , Cães , Relação Dose-Resposta a Droga , Fibrose , Sistema de Condução Cardíaco/efeitos dos fármacos , Sistema de Condução Cardíaco/fisiopatologia , Insuficiência Cardíaca/genética , Receptor A1 de Adenosina/genética , Receptor A1 de Adenosina/fisiologia , Nó Sinoatrial/efeitos dos fármacos , Nó Sinoatrial/patologia , Taquicardia/etiologia , Teofilina/farmacologia , Teofilina/uso terapêutico , Regulação para Cima , Xantinas/farmacologia , Xantinas/uso terapêutico
5.
Biochem Biophys Res Commun ; 449(4): 477-82, 2014 Jul 11.
Artigo em Inglês | MEDLINE | ID: mdl-24845382

RESUMO

Several diseases are related to retinal ganglion cell death, such as glaucoma, diabetes and other retinopathies. Many studies have attempted to identify factors that could increase neuroprotection after axotomy of these cells. Interleukin-6 has been shown to be able to increase the survival and regeneration of retinal ganglion cells (RGC) in mixed culture as well as in vivo. In this work we show that the trophic effect of IL-6 is mediated by adenosine receptor (A2aR) activation and also by the presence of extracellular BDNF. We also show that there is a complex cross-talk between IL-6, BDNF, the Adenosine A1 and A2a receptors that results in neuroprotection of retinal ganglion cells.


Assuntos
Fator Neurotrófico Derivado do Encéfalo/metabolismo , Interleucina-6/fisiologia , Receptor A1 de Adenosina/fisiologia , Receptor A2A de Adenosina/fisiologia , Células Ganglionares da Retina/patologia , Adenina/análogos & derivados , Adenina/farmacologia , Adenosina/análogos & derivados , Adenosina/farmacologia , Animais , Axotomia , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/fisiologia , Células Cultivadas , Fármacos Neuroprotetores/farmacologia , Fenetilaminas/farmacologia , Ratos , Receptor A1 de Adenosina/biossíntese , Receptor A2A de Adenosina/biossíntese
6.
Biochem Biophys Res Commun ; 430(2): 512-8, 2013 Jan 11.
Artigo em Inglês | MEDLINE | ID: mdl-23232112

RESUMO

IL-6 is a pleiotropic cytokine classically denominated pro-inflammatory. It has been already demonstrated that IL-6 can increase the survival of retinal ganglion cells (RGC) in culture. In this work, we show that the trophic effect of IL-6 is mediated by adenosine receptor (A1R) activation. The neutralization of extracellular BDNF abolished the IL-6 effect and the treatment with IL-6 and CHA (an agonist of A1R) modulated BDNF expression as well as pCREB and pTrkB levels.


Assuntos
Interleucina-6/farmacologia , Receptor A1 de Adenosina/fisiologia , Células Ganglionares da Retina/efeitos dos fármacos , Adenosina/farmacologia , Agonistas do Receptor A1 de Adenosina/farmacologia , Animais , Fator Neurotrófico Derivado do Encéfalo/metabolismo , Sobrevivência Celular/efeitos dos fármacos , Células Cultivadas , Ratos , Ratos Endogâmicos , Receptor A1 de Adenosina/biossíntese , Células Ganglionares da Retina/metabolismo
7.
J Neurochem ; 123(2): 239-49, 2012 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-22862679

RESUMO

Little is known about the mechanisms that regulate the expression of adenosine receptors during CNS development. We demonstrate here that retinas from chick embryos injected in ovo with selective adenosine receptor ligands show changes in A1 receptor expression after 48 h. Exposure to A1 agonist N6-cyclohexyladenosine (CHA) or antagonist 8-Cyclopentyl-1, 3-dipropylxanthine (DPCPX) reduced or increased, respectively, A1 receptor protein and [³H]DPCPX binding, but together, CHA+DPCPX had no effect. Interestingly, treatment with A(2A) agonist 3-[4-[2-[[6-amino-9-[(2R,3R,4S,5S)-5-(ethylcarbamoyl)-3,4-dihydroxy-oxolan-2-yl]purin-2-yl]amino] ethyl]phenyl] propanoic acid (CGS21680) increased A1 receptor protein and [³H]DPCPX binding, and reduced A(2A) receptors. The A(2A) antagonists 7-(2-phenylethyl)-5-amino-2-(2-furyl)-pyrazolo-[4,3-e]-1,2,4-trizolo[1,5-c] pyrimidine (SCH58261) and 4-(2-[7-amino-2-[2-furyl][1,2,4]triazolo[2,3-a][1,3,5]triazo-5-yl-amino]ethyl)phenol (ZM241385) had opposite effects on A1 receptor expression. Exposure to CGS21680 + CHA did not change A1 receptor levels, whereas CHA + ZM241385 or CGS21680 + DPCPX had no synergic effect. The blockade of adenosine transporter with S-(4-nitrobenzyl)-6-thioinosine (NBMPR) also reduced [³H]DPCPX binding, an effect blocked by DPCPX, but not enhanced by ZM241385. [³H]DPCPX binding kinetics showed that treatment with CHA reduced and CGS21680 increased the Bmax, but did not affect Kd values. CHA, DPCPX, CGS21680, and ZM241385 had no effect on A1 receptor mRNA. These data demonstrated an in vivo regulation of A1 receptor expression by endogenous adenosine or long-term treatment with A1 and A(2A) receptors modulators.


Assuntos
Adenosina/biossíntese , Regulação da Expressão Gênica no Desenvolvimento , Receptor A1 de Adenosina/biossíntese , Receptor A2A de Adenosina/fisiologia , Retina/embriologia , Retina/metabolismo , Agonistas do Receptor A1 de Adenosina/farmacologia , Antagonistas do Receptor A1 de Adenosina/farmacologia , Antagonistas do Receptor A2 de Adenosina/farmacologia , Animais , Embrião de Galinha , Retina/efeitos dos fármacos
8.
J Neurochem ; 121(2): 217-27, 2012 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-22335470

RESUMO

The extension of microglial processes toward injured sites in the brain is triggered by the stimulation of the purinergic receptor P2Y(12) by extracellular ATP. We recently showed that P2Y(12) stimulation by ATP induces microglial process extension in collagen gels. In the present study, we found that a P2Y(12) agonist, 2-methylthio-ADP (2MeSADP), failed to induce the process extension of microglia in collagen gels and that co-stimulation with adenosine, a phosphohydrolytic derivative of ATP, and 2MeSADP restored the chemotactic process extension. An adenosine A3 receptor (A3R)-selective agonist restored the chemotactic process extension, but other receptor subtype agonists did not. The removal of adenosine by adenosine deaminase and the blocking of A3R by an A3R-selective antagonist inhibited ADP-induced process extension. The A3R antagonist inhibited ADP-induced microglial migration, and an A3R agonist promoted 2MeSADP-stimulated migration. ADP and the A3R agonist activated Jun N-terminal kinase in microglia, and a Jun N-terminal kinase inhibitor inhibited the ADP-induced process extension. An RT-PCR analysis showed that A1R and A3R were expressed by microglia sorted from adult rat brains and that the A2AR expression level was very low. These results suggested that A3R signaling may be involved in the ADP-induced process extension and migration of microglia.


Assuntos
Difosfato de Adenosina/farmacologia , Movimento Celular/efeitos dos fármacos , Microglia/efeitos dos fármacos , Receptor A3 de Adenosina/fisiologia , Adenosina/farmacologia , Agonistas do Receptor A3 de Adenosina/farmacologia , Inibidores de Adenosina Desaminase/farmacologia , Difosfato de Adenosina/análogos & derivados , Animais , Animais Recém-Nascidos , Células Cultivadas , Córtex Cerebral/citologia , Córtex Cerebral/efeitos dos fármacos , Quimiotaxia/efeitos dos fármacos , Colágeno , Citometria de Fluxo , Indicadores e Reagentes , Proteínas Quinases JNK Ativadas por Mitógeno/fisiologia , Agonistas do Receptor Purinérgico P2Y/farmacologia , Ratos , Ratos Wistar , Reação em Cadeia da Polimerase em Tempo Real , Receptor A1 de Adenosina/biossíntese , Receptor A3 de Adenosina/efeitos dos fármacos , Receptores Purinérgicos P2Y12/efeitos dos fármacos , Tionucleotídeos/farmacologia
9.
Basic Res Cardiol ; 106(6): 953-66, 2011 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-21681579

RESUMO

In rat atrial myocytes GIRK (Kir3) channels can be activated by acetylcholine and adenosine via M(2) and A(1) receptors coupled to Pertussis-toxin-sensitive G proteins, such as M(2)R or A(1)R. Owing to the lower density of A(1)R, the amplitude of current activated by a saturating concentration (10 µM) of Ado (I(K(Ado))) amounts to about 40% of maximum I(K(ACh)). Adenovirus-driven overexpression of A(1)R results in an increase in I(K(Ado)). In a fraction of A(1)R-overexpressing cells, both ACh and Ado failed to activate GIRK channels. These cells had a large constitutive Ba(2+)-sensitive inward rectifying background K(+) current, which was insensitive to the GIRK channel inhibitor tertiapin (200 nM), suggesting this current component to be carried by I(K1) (Kir) channels. This effect of A(1)R overexpression was reduced by treatment (48 h) with the A(1)R antagonist DPCPX. siRNA-mediated knockdown of Kir2.1, simultaneously with A(1)R overexpression, substantially reduced I(K1). The mechanisms underlying the upregulation of functional I(K1) channels involve activation of the phosphatidylinositol 3-kinase (Pi3K)/Akt (protein kinase B) pathway. Kir2.1 transcripts are not increased in myocytes overexpressing A(1)R. These data demonstrate that manipulation of the expression level of a G protein-coupled receptor has unpredictable effects on functional expression of proteins that are supposed to be unrelated to the pathway controlled by that GPCR.


Assuntos
Canais de Potássio Corretores do Fluxo de Internalização Acoplados a Proteínas G/metabolismo , Átrios do Coração/metabolismo , Miócitos Cardíacos/metabolismo , Receptor A1 de Adenosina/biossíntese , Transdução de Sinais/fisiologia , Animais , Feminino , Técnicas de Silenciamento de Genes , Masculino , Técnicas de Cultura de Órgãos , Técnicas de Patch-Clamp , Interferência de RNA , Ratos , Ratos Endogâmicos WKY , Reação em Cadeia da Polimerase em Tempo Real , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transdução Genética , Regulação para Cima
10.
Neuropathol Appl Neurobiol ; 37(5): 484-99, 2011 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-21114681

RESUMO

AIM: Neuropathological changes classically associated with sheep scrapie do not always correlate with clinical disease. We aimed to determine if selected neuromodulatory responses were altered during the course of the infection as it has been described in Creutzfeldt-Jakob disease and experimental bovine spongiform encephalopathy. METHODS: Hemi-brains from healthy sheep and natural scrapie cases at two stages of infection were examined for biochemical alterations related to the expression of type I metabotropic glutamatergic receptors (mGluR(1) ) and type I adenosine receptors I (A(1) R), and of selected downstream intermediate signalling targets. Immunohistochemistry for different scrapie-related neuropathological changes was performed in the contralateral hemi-brains. RESULTS: PrP(d) deposition, spongiform change, astrocytosis and parvalbumin expression were significantly altered in brains from clinically affected sheep compared with preclinical cases and negative controls; the latter also showed significantly higher immunoreactivity for synaptophysin than clinical cases. Between clinically affected and healthy sheep, no differences were found in the protein levels of mGluR(1) , while phospholipase Cß1 expression in terminally ill sheep was increased in some brain areas but decreased in others. Adenyl cyclase 1 and A(1) R levels were significantly lower in various brain areas of affected sheep. No abnormal biochemical expression levels of these markers were found in preclinically infected sheep. CONCLUSIONS: These findings point towards an involvement of mGluR(1) and A(1) R downstream pathways in natural scrapie. While classical prion disease lesions and neuromodulatory responses converge in some affected regions, they do not do so in others suggesting that there are independent regulatory factors for distinct degenerative and neuroprotective responses.


Assuntos
Receptor A1 de Adenosina/biossíntese , Receptores de Glutamato Metabotrópico/biossíntese , Scrapie/metabolismo , Scrapie/patologia , Animais , Western Blotting , Encéfalo/metabolismo , Encéfalo/patologia , Imuno-Histoquímica , Ovinos
11.
J Neurochem ; 113(3): 661-73, 2010 May.
Artigo em Inglês | MEDLINE | ID: mdl-20163523

RESUMO

The expression of A1 and A2a adenosine receptors is developmentally regulated in the chick retina, but little is known about the factors important for this regulation. Here, we show that cell aggregation and cAMP analogs promote a dramatic increase in A1 receptor expression. Importantly, a long-term stimulation of A2a receptors also promotes an increase of A1 receptor expression accompanied by a down-regulation of A2a receptors. Chick embryo retina cultures grown in the form of aggregates or dispersed cells accumulate cAMP when stimulated with dopamine or the adenosine agonist 2-chloroadenosine. However, inhibition of dopamine-dependent cAMP accumulation by 2-chloroadenosine was observed in aggregate cultures but not in dispersed cell cultures. Accordingly, A1 receptor binding sites were detected in aggregate cultures, but were low or absent from dispersed cell cultures. Interestingly, an increase of A1 binding sites was detected when dispersed cell cultures were treated for 5 days with permeable cAMP analogs, the adenylyl cyclase activator forskolin or A2a receptor agonists. Although a significant amount of A1 receptor protein was detected in dispersed cell cultures by western blot or immunocytochemistry, the long-term stimulation of A2a receptors also promoted an increase of the A1 receptor protein and mRNA, indicating that A2a receptors and cAMP were regulating transcription and/or translation of A1 receptors. We also found an increase of A1 receptors in locations in or near the membrane after treatment with A2a agonist. The long-term stimulation of retinal explants with A2a agonist also promoted an increase of A1 receptor protein. The results indicate that A2a receptors and the cAMP-dependent protein kinase pathway are involved in the regulation of A1 receptor expression during retinal development.


Assuntos
Proteínas Quinases Dependentes de AMP Cíclico/fisiologia , AMP Cíclico/fisiologia , Receptor A1 de Adenosina/biossíntese , Receptor A2A de Adenosina/biossíntese , Retina/metabolismo , 2-Cloroadenosina/farmacologia , Animais , Western Blotting , Agregação Celular , Células Cultivadas , Embrião de Galinha , Dopamina/farmacologia , Dopamina/fisiologia , Expressão Gênica/fisiologia , Imuno-Histoquímica , RNA Mensageiro/biossíntese , RNA Mensageiro/genética , Retina/citologia , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Xantinas/metabolismo
12.
J Neurochem ; 114(6): 1667-77, 2010 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-20598020

RESUMO

Neuroprotection is one of the prominent functions of the interleukin (IL)-6-type cytokine family, for which the underlying mechanism(s) are not fully understood. We have previously shown that neuroprotection and neuromodulation mediated by IL-6 require neuronal adenosine A(1) receptor (A(1) R) function. We now have investigated whether two other IL-6-type cytokines [oncostatin M (OSM) and leukemia inhibitory factor (LIF)] use a similar mechanism. It is presented here that OSM but not LIF, enhanced the expression of A(1) Rs (both mRNA and protein levels) in cultured neurons. Whereas the neuroprotective effect of LIF was unchanged in A(1) R deficient neurons, OSM failed to protect neurons in the absence of A(1) R. In addition, OSM pre-treatment for 4 h potentiated the A(1) R-mediated inhibition of electrically evoked excitatory post-synaptic currents recorded from hippocampal slices either under normal or hypoxic conditions. No such effect was observed after LIF pre-treatment. Our findings thus strongly suggest that, despite known structural and functional similarities, OSM and LIF use different mechanisms to achieve neuroprotection and neuromodulation.


Assuntos
Fator Inibidor de Leucemia/fisiologia , Neurônios/fisiologia , Oncostatina M/fisiologia , Receptor A1 de Adenosina/fisiologia , Animais , Hipóxia Celular , Sobrevivência Celular , Células Cultivadas , Estimulação Elétrica , Potenciais Pós-Sinápticos Excitadores , Hipocampo/citologia , Hipocampo/fisiologia , Técnicas In Vitro , Fator Inibidor de Leucemia/farmacologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Neurônios/citologia , Neurônios/efeitos dos fármacos , Fármacos Neuroprotetores/farmacologia , Oncostatina M/farmacologia , Técnicas de Patch-Clamp , RNA Mensageiro/biossíntese , Receptor A1 de Adenosina/biossíntese , Receptor A1 de Adenosina/genética , Proteínas Recombinantes/farmacologia , Transmissão Sináptica
13.
J Neurochem ; 115(3): 782-94, 2010 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-20807311

RESUMO

Insomnia is a severe symptom of alcohol withdrawal; however, the underlying neuronal mechanism is yet unknown. We hypothesized that chronic ethanol exposure will impair basal forebrain (BF) adenosinergic mechanism resulting in insomnia-like symptoms. We performed a series of experiments in Sprague-Dawley rats to test our hypothesis. We used Majchrowicz's chronic binge ethanol protocol to induce ethanol dependency. Our first experiment verified the effects of ethanol withdrawal on sleep-wakefulness. Significant increase in wakefulness was observed during ethanol withdrawal. Next, we examined c-Fos expression (marker of neuronal activation) in BF wake-promoting neurons during ethanol withdrawal. There was a significant increase in the number of BF wake-promoting neurons with c-Fos immunoreactivity. Our third experiment examined the effects of ethanol withdrawal on sleep deprivation induced increase in BF adenosine levels. Sleep deprivation did not increase BF adenosine levels in ethanol dependent rats. Our last experiment examined the effects of ethanol withdrawal on equilibrative nucleoside transporter 1 and A1 receptor expression in the BF. There was a significant reduction in A1 receptor and equilibrative nucleoside transporter 1 expression in the BF of ethanol dependent rats. Based on these results, we suggest that insomnia observed during ethanol withdrawal is caused because of impaired adenosinergic mechanism in the BF.


Assuntos
Adenosina/fisiologia , Alcoolismo/complicações , Alcoolismo/fisiopatologia , Prosencéfalo/fisiopatologia , Distúrbios do Início e da Manutenção do Sono/etiologia , Distúrbios do Início e da Manutenção do Sono/fisiopatologia , Transtornos do Sono-Vigília/etiologia , Transtornos do Sono-Vigília/fisiopatologia , Vigília/fisiologia , Adenosina/metabolismo , Animais , Proteínas de Transporte/biossíntese , Contagem de Células , Transportador Equilibrativo 1 de Nucleosídeo , Imuno-Histoquímica , Masculino , Microdiálise , Proteínas Proto-Oncogênicas c-fos/biossíntese , RNA/biossíntese , RNA/genética , Ratos , Ratos Sprague-Dawley , Receptor A1 de Adenosina/biossíntese , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Sono/fisiologia , Privação do Sono , Sono REM/fisiologia , Síndrome de Abstinência a Substâncias/psicologia
14.
Circ Res ; 102(3): 356-63, 2008 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-18032734

RESUMO

Intracoronary delivery of endothelial progenitor cells (EPCs) is an emerging concept for the treatment of cardiovascular disease. Enhancement of EPC adhesion to vascular endothelium could improve cell retention within targeted organs. Because extracellular adenosine is elevated at sites of ischemia and stimulates neovascularization, we examined the potential role of adenosine in augmenting EPC retention to cardiac microvascular endothelium. Stimulation of adenosine receptors in murine embryonic EPCs (eEPCs) and cardiac endothelial cells (cECs) rapidly, within minutes, increased eEPC adhesion to cECs under static and flow conditions. Similarly, adhesion of human adult culture-expanded EPCs to human cECs was increased by stimulation of adenosine receptors. Furthermore, adenosine increased eEPC retention in isolated mouse hearts perfused with eEPCs. We determined that eEPCs and cECs preferentially express functional A1 and A2B adenosine receptor subtypes, respectively, and that both subtypes are involved in the regulation of eEPC adhesion to cECs. We documented that the interaction between P-selectin and its ligand (P-selectin glycoprotein ligand-1) plays a role in adenosine-dependent eEPC adhesion to cECs and that stimulation of adenosine receptors in cECs induces rapid cell surface expression of P-selectin. Our results suggest a role for adenosine in vasculogenesis and its potential use to stimulate engraftment in cell-based therapies.


Assuntos
Adenosina/farmacologia , Células Endoteliais/metabolismo , Neovascularização Fisiológica/fisiologia , Receptor A1 de Adenosina/biossíntese , Receptor A2B de Adenosina/biossíntese , Células-Tronco/metabolismo , Vasodilatadores/farmacologia , Adenosina/metabolismo , Agonistas do Receptor A1 de Adenosina , Agonistas do Receptor A2 de Adenosina , Animais , Adesão Celular/efeitos dos fármacos , Células Cultivadas , Células Endoteliais/citologia , Células Endoteliais/transplante , Endotélio Vascular/citologia , Endotélio Vascular/metabolismo , Glicoproteínas de Membrana/biossíntese , Camundongos , Isquemia Miocárdica/metabolismo , Isquemia Miocárdica/terapia , Miocárdio/citologia , Miocárdio/metabolismo , Neovascularização Fisiológica/efeitos dos fármacos , Selectina-P/biossíntese , Transplante de Células-Tronco , Células-Tronco/citologia , Vasodilatadores/metabolismo
15.
Eur J Pharmacol ; 887: 173431, 2020 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-32758568

RESUMO

Intraocular pressure (IOP) lowering in glaucomatous eyes is currently achieved mainly by improved aqueous outflow via alternate drainage pathways. However, the focus is now shifting to trabecular meshwork (TM), the site or major pathological changes including increased extracellular matrix (ECM) deposition and reduced matrix metalloproteinases (MMPs) secretion by TM cells. Trans-resveratrol was previously shown to lower IOP and reduce ECM deposition; however, the mechanisms of action remain unclear. Therefore, we determined the effect of trans-resveratrol on MMP-2 and -9 expression by human TM cells (HTMCs) in the presence of dexamethasone and whether it also affects adenosine A1 receptors (A1AR) expression and nuclear factor kappa B (NFkB) activation. We observed that trans-resveratrol, 12.5 µM, increased MMP-2 and -9 protein expression by HTMCs despite exposure to dexamethasone (1.89- and 1.53-fold, respectively; P < 0.001). Further it was observed that trans-resveratrol increases A1AR expression in HTMC in the presence of dexamethasone (1.55-fold; P < 0.01). Trans-resveratrol also increased NFkB activation in the presence of dexamethasone and A1AR antagonist (P < 0.01 versus dexamethasone group). These effects of trans-resveratrol were associated with increased MMP -2 and -9 expression. It could be concluded that trans-resveratrol prevents dexamethasone-induced reduction in MMP-2 and -9 secretion by NFkB activation in HTMCs. This effect of trans-resveratrol is likely to involve increased A1AR expression.


Assuntos
Dexametasona/toxicidade , Metaloproteinases da Matriz/biossíntese , NF-kappa B/biossíntese , Receptor A1 de Adenosina/biossíntese , Resveratrol/farmacologia , Malha Trabecular/metabolismo , Antioxidantes/farmacologia , Células Cultivadas , Regulação Enzimológica da Expressão Gênica , Humanos , Inibidores de Metaloproteinases de Matriz/toxicidade , NF-kappa B/antagonistas & inibidores , Malha Trabecular/efeitos dos fármacos
16.
Am J Physiol Renal Physiol ; 297(5): F1256-64, 2009 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-19741017

RESUMO

Adenosine 1 receptors (A1AR) in the kidney are expressed in the vasculature and the tubular system. Pharmacological inhibition or global genetic deletion of A1AR causes marked reductions or abolishment of tubuloglomerular feedback (TGF) responses. To assess the function of vascular A1AR in TGF, we generated transgenic mouse lines in which A1AR expression in smooth muscle was augmented by placing A1AR under the control of a 5.38-kb fragment of the rat smooth muscle alpha-actin promoter and first intron (12). Two founder lines with highest expression in the kidney [353 +/- 42 and 575 +/- 43% compared with the wild type (WT)] were used in the experiments. Enhanced expression of A1AR at the expected site in these lines was confirmed by augmented constrictor responses of isolated afferent arterioles to administration of the A1AR agonist N6-cyclohexyladenosine. Maximum TGF responses (0-30 nl/min flow step) were increased from 8.4 +/- 0.9 mmHg in WT (n = 21) to 14.2 +/- 0.7 mmHg in A1AR-transgene (tg) 4 (n = 22; P < 0.0001), and to 12.6 +/- 1.2 mmHg in A1AR-tg7 (n = 12; P < 0.02). Stepwise changes in perfusion flow caused greater numerical TGF responses in A1AR-tg than WT in all flow ranges with differences reaching levels of significance in the intermediate flow ranges of 7.5-10 and 10-15 nl/min. Proximal-distal single-nephron glomerular filtration rate (SNGFR) differences (free-flow micropuncture) were also increased in A1AR-tg, averaging 6.25 +/- 1.5 nl/min compared with 2.6 +/- 0.51 nl/min in WT (P = 0.034). Basal plasma renin concentrations as well as the suppression of renin secretion after volume expansion were similar in A1AR-tg and WT mice, suggesting lack of transgene expression in juxtaglomerular cells. These data indicate that A1AR expression in vascular smooth muscle cells is a critical component for TGF signaling and that changes in renal vascular A1AR expression may determine the magnitude of TGF responses.


Assuntos
Vasos Sanguíneos/metabolismo , Retroalimentação/fisiologia , Glomérulos Renais/metabolismo , Receptor A1 de Adenosina/biossíntese , Agonistas do Receptor A1 de Adenosina , Animais , Arteríolas/citologia , Arteríolas/metabolismo , Pressão Sanguínea/fisiologia , DNA Complementar/biossíntese , DNA Complementar/genética , Taxa de Filtração Glomerular , Rim/patologia , Glomérulos Renais/patologia , Camundongos , Camundongos Transgênicos , Músculo Liso Vascular/citologia , Músculo Liso Vascular/metabolismo , Concentração Osmolar , RNA Mensageiro/biossíntese , RNA Mensageiro/genética , Ratos , Receptor A1 de Adenosina/genética , Renina/sangue , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transdução de Sinais/fisiologia , Fator de Crescimento Transformador alfa/fisiologia
17.
Anat Rec (Hoboken) ; 301(11): 1882-1905, 2018 11.
Artigo em Inglês | MEDLINE | ID: mdl-30315630

RESUMO

In the brain, purines such as ATP and adenosine can function as neurotransmitters and co-transmitters, or serve as signals in neuron-glial interactions. In thalamocortical (TC) projections to sensory cortex, adenosine functions as a negative regulator of glutamate release via activation of the presynaptic adenosine A1 receptor (A1 R). In the auditory forebrain, restriction of A1 R-adenosine signaling in medial geniculate (MG) neurons is sufficient to extend LTP, LTD, and tonotopic map plasticity in adult mice for months beyond the critical period. Interfering with adenosine signaling in primary auditory cortex (A1) does not contribute to these forms of plasticity, suggesting regional differences in the roles of A1 R-mediated adenosine signaling in the forebrain. To advance understanding of the circuitry, in situ hybridization was used to localize neuronal and glial cell types in the auditory forebrain that express A1 R transcripts (Adora1), based on co-expression with cell-specific markers for neuronal and glial subtypes. In A1, Adora1 transcripts were concentrated in L3/4 and L6 of glutamatergic neurons. Subpopulations of GABAergic neurons, astrocytes, oligodendrocytes, and microglia expressed lower levels of Adora1. In MG, Adora1 was expressed by glutamatergic neurons in all divisions, and subpopulations of all glial classes. The collective findings imply that A1 R-mediated signaling broadly extends to all subdivisions of auditory cortex and MG. Selective expression by neuronal and glial subpopulations suggests that experimental manipulations of A1 R-adenosine signaling could impact several cell types, depending on their location. Strategies to target Adora1 in specific cell types can be developed from the data generated here. Anat Rec, 301:1882-1905, 2018. © 2018 The Authors. The Anatomical Record published by Wiley Periodicals, Inc. on behalf of American Association of Anatomists.


Assuntos
Córtex Auditivo/metabolismo , Neuroglia/metabolismo , Neurônios/metabolismo , RNA Mensageiro/biossíntese , Receptor A1 de Adenosina/biossíntese , Animais , Córtex Auditivo/citologia , Feminino , Expressão Gênica , Corpos Geniculados/citologia , Corpos Geniculados/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Neuroglia/citologia , Prosencéfalo/citologia , Prosencéfalo/metabolismo , RNA Mensageiro/genética , Receptor A1 de Adenosina/genética
18.
Life Sci ; 81(13): 1031-41, 2007 Sep 08.
Artigo em Inglês | MEDLINE | ID: mdl-17869311

RESUMO

The striatal dopamine D2 receptor (D2R) and adenosine A2A receptor (A2AAR) exhibit mutually antagonistic effects through physical interactions and by differential modulation of post-receptor signaling pathways. The expression of the A2AAR and the D2R is differentially regulated by nuclear factor-kappaB (NF-kappaB). In this report, we determined the role of NF-kappaB in regulation of these receptors by comparing mice deficient in the NF-kappaB p50 subunit (p50 KO) with genetically intact B6129PF2/J (F2) mice. Quantification of adenosine receptor (AR) subtypes in mouse striatum by real time PCR, immunocytochemistry and radioligand binding assays showed more A2AAR but less A1AR in p50 KO mice as compared with F2 mice. Striata from p50 KO mice also had less D2R mRNA and [(3)H]-methylspiperone binding than did striata from F2 mice. G(alphaolf) and G(alphas) proteins, which are transducers of A2AAR signals, were also present at a higher level in striata from the p50 KO versus F2 mice. In contrast, the G(alphai1) protein, which transduces signals from the A1AR and D2R, was significantly reduced in striata from p50 KO mice. Behaviorally, p50 KO mice exhibited increased locomotor activity relative to that of F2 mice after caffeine ingestion. These data are consistent with a role for the NF-kappaB in the regulation of A1AR, A2AAR, D2R and possibly their coupling G proteins in the striatum. Dysregulation of these receptors in the striata of p50 KO mice might sensitize these animals to locomotor stimulatory action of caffeine.


Assuntos
Gânglios da Base/metabolismo , Regulação da Expressão Gênica/fisiologia , Subunidade p50 de NF-kappa B/metabolismo , Receptor A2A de Adenosina/biossíntese , Receptores de Dopamina D2/biossíntese , Transdução de Sinais/fisiologia , Animais , Gânglios da Base/citologia , Comportamento Animal/efeitos dos fármacos , Comportamento Animal/fisiologia , Cafeína/farmacologia , Estimulantes do Sistema Nervoso Central/farmacologia , Subunidades alfa de Proteínas de Ligação ao GTP/metabolismo , Regulação da Expressão Gênica/efeitos dos fármacos , Camundongos , Camundongos Knockout , Atividade Motora/efeitos dos fármacos , Atividade Motora/fisiologia , Subunidade p50 de NF-kappa B/genética , RNA Mensageiro/biossíntese , Receptor A1 de Adenosina/biossíntese , Transdução de Sinais/efeitos dos fármacos
19.
J Clin Endocrinol Metab ; 91(5): 1882-6, 2006 May.
Artigo em Inglês | MEDLINE | ID: mdl-16507638

RESUMO

CONTEXT: African-American women (AAW) lose less weight and at a slower rate than Caucasian women (CAW) under the same weight-loss regimens. A potential cause of this finding is inhibition of lipolysis. OBJECTIVE: Because alpha-2 and adenosine receptors are directly involved in inhibition of lipolysis, differences in alpha-2 or adenosine A1 receptors in visceral adipose tissue (VAT) and sc adipose tissue (SAT) from obese AAW and CAW were determined. DESIGN: Measurements of maximal binding capacity (Bmax) of alpha-2 and adenosine A1 receptors as well as protein and mRNA levels of the adenosine receptor in VAT and SAT from AAW and CAW were taken. SETTING: The study was conducted in the general community. PATIENTS: Patients were selected by body mass index greater than 40 and age matched. MAIN OUTCOME MEASURES: Bmax (density) of the two receptors and protein and mRNA levels of adenosine receptors were determined in adipose tissue of AAW and CAW. RESULTS: No differences were found for alpha-2 receptor Bmax in either VAT or SAT from AAW and CAW. Bmax (but not the dissociation constant, Kd) for the adenosine A1 receptor in VAT from AAW was higher (P < 0.05) than in VAT from CAW. Adenosine receptor protein and mRNA levels were significantly higher in VAT from AAW than VAT from CAW. No racial differences in these parameters were observed in SAT. CONCLUSIONS: These data suggest that inhibition of lipolysis by adenosine has the potential to be greater in obese AAW, and this could possibly be one explanation for the observation that obese AAW have more difficulty in losing weight than obese CAW.


Assuntos
Tecido Adiposo/metabolismo , Obesidade Mórbida/metabolismo , Receptor A1 de Adenosina/biossíntese , Adulto , População Negra , Ácidos Graxos não Esterificados/sangue , Feminino , Humanos , Insulina/sangue , Membranas/metabolismo , RNA Mensageiro/biossíntese , RNA Mensageiro/genética , Ensaio Radioligante , Receptor A1 de Adenosina/genética , Receptores Adrenérgicos alfa 2/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , População Branca
20.
Toxicology ; 339: 19-33, 2016 Jan 02.
Artigo em Inglês | MEDLINE | ID: mdl-26478469

RESUMO

The aim of the present study was to investigate the molecular effects of perinatal exposure to lead (Pb) on protein and mRNA expression of purine receptors: P2X4, P2X7, adenosine receptor A1; and astrocytes (GFAP mRNA expression) and on microglia activation (Iba1 mRNA expression) in several structures of the mesolimbic system (striatum, hippocampus, prefrontal cortex) in rats expressing tolerance to the antinociceptive effect of morphine. Rat mothers were orally treated with 0.1% lead acetate from conception, through gestation, and postnatally, as well as to offspring up to day (PND) 28; subsequently molecular studies were conducted on adult (PND 60) male rats. Morphine tolerance developed more strongly in rats perinatally exposed to Pb. The analysis revealed a significant up-regulation of protein and mRNA P2X4 receptor expression in the striatum and prefrontal cortex but not in the hippocampus; P2X7 protein and mRNA receptor expression in the striatum and hippocampus, but not in the prefrontal cortex; A1 protein receptor expression in all investigated structures and A1 mRNA expression in the striatum and hippocampus; Iba1 mRNA expression in the striatum and hippocampus; and GFAP mRNA expression in the striatum and prefrontal cortex. Immunohistochemical analysis has also revealed significant alterations. Strong expressions of P2X4, P2X7, A1 receptors, astrocytes and microglia activation were observed in the hippocampus in Pb and/or morphine treated rats. The higher expression of purine receptors and glial cell activation are important markers of neuroinflammatory processes. Therefore, we conclude that Pb-induced neuroinflammation may be responsible for the intensification of morphine tolerance in the Pb-treated rats. Additionally, the dysregulation of A1 adenosine receptors, mainly in the hippocampus, may also be involved in the intensification of morphine tolerance in Pb-treated rats. Our study demonstrates the significant participation of environmental factors in addictive process; additionally, it shows the necessity of modification of addictive disorder with neuroprotective agents.


Assuntos
Analgésicos Opioides/farmacologia , Química Encefálica/efeitos dos fármacos , Gliose/induzido quimicamente , Intoxicação por Chumbo/metabolismo , Morfina/farmacologia , Receptores Purinérgicos/biossíntese , Animais , Astrócitos/efeitos dos fármacos , Astrócitos/metabolismo , Tolerância a Medicamentos , Feminino , Gliose/patologia , Chumbo/metabolismo , Masculino , Degeneração Neural/induzido quimicamente , Degeneração Neural/patologia , Neuroglia/efeitos dos fármacos , Neuroglia/metabolismo , Gravidez , Ratos , Ratos Wistar , Receptor A1 de Adenosina/biossíntese , Receptores Purinérgicos P2X4/biossíntese , Receptores Purinérgicos P2X7/biossíntese , Regulação para Cima
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa