Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 912
Filtrar
1.
Toxicology ; 411: 172-180, 2019 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-30359672

RESUMO

Our previous study showed that both Kupffer cell eliminator (GdCl3) and tumor necrosis factor α (TNF-α) receptor antagonist (etanercept) could partially attenuate binge drinking-induced liver steatosis. Herein, we extended the study by directly investigating the roles of TNF-α on the hepatic fat levels in mice and in HepG2 cells, and explored the underlying mechanisms. SPF male ICR mice were exposed to TNF-α (0.166 mg/kg body weight) with or without phenylisopropyl adenosine (PIA, an anti-lipolytic drug) for 1.5, 3, 6, and 24 h. We found that TNF-α treatment resulted in hepatic triglyceride (TG) elevation at 6 h time point, which was blocked by PIA. TNF-α led to the activation of extrahepatic lipolysis demonstrated by the increase of serum free fatty acid (FFA) level, and the increased protein levels of adipose triglyceride lipase (ATGL) and phosphorylated hormone-sensitive lipase (HSL) in mice epididymal adipose tissues, but had no significant effects on the protein levels of sterol regulatory element binding protein 1c (SREBP-1c) and peroxisomal proliferator activation receptor α (PPAR-α) in mice liver. The in vitro study showed TNF-α treatment could not result in elevation of TG in HepG2 cells, although it indeed brought about a slight activation of SREBP-1c. These results support the hypothesis that TNF-α might make a small contribution to ethanol-induced fatty liver by stimulating extrahepatic lipolysis.


Assuntos
Ácidos Graxos/metabolismo , Fígado Gorduroso/induzido quimicamente , Fígado Gorduroso/metabolismo , Lipólise/efeitos dos fármacos , Fígado/metabolismo , Fator de Necrose Tumoral alfa/toxicidade , Animais , Ácidos Graxos/sangue , Fígado Gorduroso/patologia , Células Hep G2 , Humanos , Fígado/patologia , Masculino , Camundongos , Camundongos Endogâmicos ICR , PPAR alfa/metabolismo , Fenilisopropiladenosina/toxicidade , Proteína de Ligação a Elemento Regulador de Esterol 1/biossíntese , Proteína de Ligação a Elemento Regulador de Esterol 1/genética , Triglicerídeos/metabolismo
2.
Epilepsy Behav ; 61: 168-173, 2016 08.
Artigo em Inglês | MEDLINE | ID: mdl-27371881

RESUMO

Adenosine is an endogenous anticonvulsant that activates pre- and postsynaptic adenosine A1 receptors. A1 receptor agonists increase the latency for the development of seizures and status epilepticus following pilocarpine administration. Although hippocampal adenosine is increased in the chronic phase of the pilocarpine model, it is not known whether the modulation of A1 receptors may influence the frequency of spontaneous recurrent seizures (SRS). Here, we tested the hypothesis that the A1 receptor agonist RPia ([R]-N-phenylisopropyladenosine) and the A1 antagonist DPCPX (8-Cyclopentyl-1,3-dipropylxanthine) administered to chronic pilocarpine epileptic rats would respectively decrease and increase the frequency of SRS and hippocampal excitability. Four months after Pilo-induced SE, chronic epileptic rats were video-monitored for the recording of SRS before (basal) and after a 2-week treatment with RPia (25µg/kg) or DPCPX (50µg/kg). Following sacrifice, brain slices were studied with electrophysiology. We found that rats given RPia had a 93% nonsignificant reduction in the frequency of seizures compared with their own pretreatment baseline. In contrast, the administration of DPCPX resulted in an 87% significant increase in seizure rate. Nontreated epileptic rats had a similar frequency of seizures along the study. Corroborating our behavioral data, in vitro recordings showed that slices from animals previously given DPCPX had a shorter latency to develop epileptiform activity, longer and higher DC shifts, and higher spike amplitude compared with slices from nontreated Pilo controls. In contrast, smaller spike amplitude was recorded in slices from animals given RPia. In summary, the administration of A1 agonists reduced hippocampal excitability but not the frequency of spontaneous recurrent seizures in chronic epileptic rats, whereas A1 receptor antagonists increased both.


Assuntos
Agonistas do Receptor A1 de Adenosina/farmacologia , Antagonistas do Receptor A1 de Adenosina/farmacologia , Convulsivantes/farmacologia , Epilepsia/induzido quimicamente , Agonistas Muscarínicos/farmacologia , Pilocarpina/farmacologia , Convulsões/induzido quimicamente , Convulsões/prevenção & controle , Animais , Encéfalo/fisiopatologia , Eletroencefalografia/efeitos dos fármacos , Epilepsia/fisiopatologia , Masculino , Fenilisopropiladenosina/farmacologia , Ratos , Ratos Wistar , Convulsões/fisiopatologia , Xantinas/farmacologia
3.
J Neurosci ; 32(8): 2683-95, 2012 Feb 22.
Artigo em Inglês | MEDLINE | ID: mdl-22357852

RESUMO

Endogenous adenosine is an essential protective agent against neural damage by various insults to the brain. However, the therapeutic potential of adenosine receptor-directed ligands for neuroprotection is offset by side effects in peripheral tissues and organs. An increase in adenosine receptor responsiveness to endogenous adenosine would enhance neuroprotection while avoiding the confounding effects of exogenous ligands. Here we report novel regulation of adenosine-evoked responses by a neural tissue-specific protein, neurabin. Neurabin attenuated adenosine A(1) receptor (A1R) signaling by assembling a complex between the A1R and the regulator of G-protein signaling 4 (RGS4), a protein known to turn off G-protein signaling. Inactivation of the neurabin gene enhanced A1R signaling and promoted the protective effect of adenosine against excitotoxic seizure and neuronal death in mice. Furthermore, administration of a small molecule inhibitor of RGS4 significantly attenuated seizure severity in mice. Notably, the dose of kainate capable of inducing an ∼50% rate of death in wild-type (WT) mice did not affect neurabin-null mice or WT mice cotreated with an RGS4 inhibitor. The enhanced anti-seizure and neuroprotective effect achieved by disruption of the A1R/neurabin/RGS4 complex is elicited by the on-site and on-demand release of endogenous adenosine, and does not require administration of A1R ligands. These data identify neurabin-RGS4 as a novel tissue-selective regulatory mechanism for fine-tuning adenosine receptor function in the nervous system. Moreover, these findings implicate the A1R/neurabin/RGS4 complex as a valid therapeutic target for specifically manipulating the neuroprotective effects of endogenous adenosine.


Assuntos
Adenosina/metabolismo , Proteínas dos Microfilamentos/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Proteínas RGS/metabolismo , Convulsões/metabolismo , Antagonistas do Receptor A1 de Adenosina/farmacologia , Animais , Animais Recém-Nascidos , Morte Celular/efeitos dos fármacos , Células Cultivadas , Cricetinae , Cricetulus , Modelos Animais de Doenças , Eletroencefalografia , Fluoresceínas , Hipocampo/patologia , Marcação In Situ das Extremidades Cortadas , Ácido Caínico/toxicidade , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteínas dos Microfilamentos/deficiência , Modelos Biológicos , Proteínas do Tecido Nervoso/deficiência , Compostos Orgânicos/metabolismo , Fenilisopropiladenosina/farmacologia , Proteínas RGS/antagonistas & inibidores , Receptor A1 de Adenosina/genética , Receptor A1 de Adenosina/metabolismo , Convulsões/induzido quimicamente , Convulsões/tratamento farmacológico , Convulsões/patologia , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/fisiologia , Sulfonamidas/farmacologia , Fatores de Tempo , Transfecção , Xantinas/farmacologia , Xantinas/uso terapêutico
4.
J Neurosci ; 31(44): 15629-39, 2011 Nov 02.
Artigo em Inglês | MEDLINE | ID: mdl-22049406

RESUMO

Astrocytes play a key role in modulating synaptic transmission by controlling the available extracellular GABA via the GAT-1 and GAT-3 GABA transporters (GATs). Using primary cultures of rat astrocytes, we show here that an additional level of regulation of GABA uptake occurs via modulation of the GATs by the adenosine A(1) (A(1)R) and A(2A) (A(2A)R) receptors. This regulation occurs through a complex of heterotetramers (two interacting homodimers) of A(1)R-A(2A)R that signal via two different G-proteins, G(s) and G(i/o), and either enhances (A(2A)R) or inhibits (A(1)R) GABA uptake. These results provide novel mechanistic insight into how G-protein-coupled receptor heteromers signal. Furthermore, we uncover a previously unknown mechanism in which adenosine, in a concentration-dependent manner, acts via a heterocomplex of adenosine receptors in astrocytes to significantly contribute to neurotransmission at the tripartite (neuron-glia-neuron) synapse.


Assuntos
Proteínas da Membrana Plasmática de Transporte de GABA/metabolismo , Subunidades alfa Gi-Go de Proteínas de Ligação ao GTP/metabolismo , Subunidades alfa Gs de Proteínas de Ligação ao GTP/metabolismo , Receptores A2 de Adenosina/metabolismo , Ácido gama-Aminobutírico/metabolismo , Análise de Variância , Animais , Animais Recém-Nascidos , Astrócitos/efeitos dos fármacos , Astrócitos/metabolismo , Proteínas de Bactérias/genética , Biotinilação , Células Cultivadas , Córtex Cerebral/citologia , Relação Dose-Resposta a Droga , Interações Medicamentosas , Inibidores Enzimáticos/farmacologia , GABAérgicos/farmacologia , Proteínas da Membrana Plasmática de Transporte de GABA/genética , Subunidades alfa Gi-Go de Proteínas de Ligação ao GTP/genética , Subunidades alfa Gs de Proteínas de Ligação ao GTP/genética , Regulação da Expressão Gênica/efeitos dos fármacos , Guanosina 5'-O-(3-Tiotrifosfato)/farmacocinética , Humanos , Proteínas Luminescentes/genética , Modelos Biológicos , Ácidos Nipecóticos/farmacologia , Fenilisopropiladenosina/metabolismo , Ligação Proteica/efeitos dos fármacos , Purinérgicos/farmacologia , Ratos , Ratos Wistar , Receptores A2 de Adenosina/genética , Proteínas Recombinantes/metabolismo , Fatores de Tempo , Transfecção/métodos , Trítio/metabolismo
5.
Epilepsy Behav ; 22(2): 207-13, 2011 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-21852200

RESUMO

Aiming at a better understanding of the role of A(2A) in temporal lobe epilepsy (TLE), we characterized the effects of the A(2A) antagonist SCH58261 (7-(2-phenylethyl)-5-amino-2(2-furyl)-pyrazolo-[4,3-e]-1,2,4-triazolo[1,5-c]pyrimidine) on seizures and neuroprotection in the pilocarpine model. The effects of SCH58261 were further analyzed in combination with the A(1) agonist R-Pia (R(-)-N(6)-(2)-phenylisopropyl adenosine). Eight groups were studied: pilocarpine (Pilo), SCH+Pilo, R-Pia+Pilo, R-Pia+SCH+Pilo, Saline, SCH+Saline, R-Pia+Saline, and R-Pia+SCH+Saline. The administration of SCH58261, R-Pia, and R-Pia+SCH58261 prior to pilocarpine increased the latency to SE, and decreased either the incidence of or rate of mortality from SE compared with controls. Administration of R-Pia and R-Pia+SCH58261 prior to pilocarpine reduced the number of Fluoro-Jade B-stained cells in the hippocampus and piriform cortex when compared with control. This study showed that pretreatment with R-Pia and SCH58261 reduces seizure occurrence, although only R-Pia has neuroprotective properties. Further studies are needed to clarify the neuroprotective role of A(2A) in TLE.


Assuntos
Receptor A1 de Adenosina/metabolismo , Receptor A2A de Adenosina/metabolismo , Estado Epiléptico/metabolismo , Adenosina/farmacologia , Análise de Variância , Animais , Encéfalo/patologia , Contagem de Células , Modelos Animais de Doenças , Interações Medicamentosas , Fluoresceínas , Masculino , Degeneração Neural/etiologia , Degeneração Neural/metabolismo , Degeneração Neural/patologia , Fármacos Neuroprotetores/farmacologia , Fármacos Neuroprotetores/uso terapêutico , Compostos Orgânicos/metabolismo , Fenilisopropiladenosina/farmacologia , Fenilisopropiladenosina/uso terapêutico , Pilocarpina/toxicidade , Pirimidinas/uso terapêutico , Ratos , Ratos Wistar , Tempo de Reação/efeitos dos fármacos , Estado Epiléptico/induzido quimicamente , Estado Epiléptico/tratamento farmacológico , Estado Epiléptico/patologia , Triazóis/uso terapêutico
6.
J Lipid Res ; 51(5): 1193-200, 2010 May.
Artigo em Inglês | MEDLINE | ID: mdl-19965580

RESUMO

High fatty acid (FA) flux is associated with systemic insulin resistance, and African-American (AA) women tend to be more insulin resistant. We assessed possible depot and race difference in the antilipolytic effect of insulin in adipocytes isolated from abdominal (Abd) and gluteal (Glt) subcutaneous (sc) adipose tissue of overweight, postmenopausal AA and Caucasian (C) women. Percent body fat, fasting insulin, visceral adiposity, and adipocyte size was higher in AA women. Disinhibited lipolysis (presence of adenosine deaminase) per unit adipocyte surface area was similar in Abd and Glt and in AA and C. However, rates of 'basal' [submaximal phenylisopropyl adenosine (PIA)-suppressed] and insulin-suppressed lipolysis were higher in Abd of AA compared with C women even after adjustment for percent fat and visceral fat area. The race difference in rates of PIA- and insulin-suppressed lipolysis in AA were correlated with their hyperinsulinemia, but AA race, independent of fasting insulin, was associated with lower responsiveness (percent suppression) to submaximal insulin concentrations, although sensitivity (ED50) was not affected. Overall, these data are consistent with the hypothesis that decreased responsiveness of Abd adipocytes to antilipolytic effectors may contribute to higher FA availability and thereby to racial differences in insulin resistance.


Assuntos
Adipócitos/metabolismo , Negro ou Afro-Americano , Resistência à Insulina/etnologia , Insulina/metabolismo , Lipólise , Pós-Menopausa/metabolismo , População Branca , Gordura Abdominal/efeitos dos fármacos , Gordura Abdominal/metabolismo , Adipócitos/efeitos dos fármacos , Adulto , Idoso , Nádegas , Ácidos Graxos não Esterificados/metabolismo , Feminino , Humanos , Insulina/farmacologia , Isoproterenol/farmacologia , Lipólise/efeitos dos fármacos , Pessoa de Meia-Idade , Fenilisopropiladenosina/farmacologia
7.
Clin Exp Pharmacol Physiol ; 37(1): 46-50, 2010 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-19566843

RESUMO

1. The effect of the adenosine A(2) receptor (AdoA(2)R) agonist N(6)-[2-(3,5-dimethoxyphenyl)-2-(2-methylphenyl)-ethyl]adenosine (DPMA) on adenosine A(1) receptor (AdoA(1)R)-mediated negative inotropic responses was investigated in rat heart. 2. Hearts from male Wistar rats (250-350 g) were perfused with Krebs'-Henseleit solution at constant flow in non-recirculating Langendorff mode. Hearts were paced at 5 Hz (5 ms duration, supramaximal voltage) via ventricular electrodes. After 30 min equilibration, (R)-N(6)-phenylisopropyl adenosine (R-PIA) concentration-response curves were constructed in the absence or presence of DPMA. 3. In paced hearts, R-PIA induced concentration-dependent decreases in triple product (heart rate x peak systolic developed pressure x dP / dt(max)), which were significantly attenuated by 1 nmol / L DPMA with a shift in pEC(50) from 8.0 +/- 0.5 (n = 9) in control hearts to 6.63 +/- 1.03 (n = 5) in treated tissues (P < 0.05). The AdoA(2A)R antagonist 8-(3-chlorostyryl)caffeine (1 micromol / L) and the adenylyl cyclase inhibitor cis-N-(2-phenylcyclopentyl)-azacyclotridec-1-en-2-amine hydrochloride (MDL12330A; 100 nmol / L) reversed the effects of DPMA on AdoA(1)R-mediated negative inotropic actions, whereas the AdoA(2B)R antagonist alloxazine (3 micromol / L) had no effect on DPMA activity. 4. The results of the present study show that stimulation of the AdoA(2)R attenuates AdoA(1)R-dependent reductions in inotropic state. The receptor involved appears to be the AdoA(2A)R and its action involves stimulation of adenylyl cyclase activity.


Assuntos
Adenosina/análogos & derivados , Contração Miocárdica/fisiologia , Fenilisopropiladenosina/farmacologia , Receptor A1 de Adenosina/fisiologia , Receptores A2 de Adenosina/fisiologia , Adenosina/farmacologia , Agonistas do Receptor A2 de Adenosina , Antagonistas do Receptor A2 de Adenosina , Inibidores de Adenilil Ciclases , Adenilil Ciclases/fisiologia , Animais , Cafeína/análogos & derivados , Cafeína/farmacologia , Depressão Química , Relação Dose-Resposta a Droga , Interações Medicamentosas , Inibidores Enzimáticos/farmacologia , Flavinas/farmacologia , Iminas/farmacologia , Técnicas In Vitro , Masculino , Contração Miocárdica/efeitos dos fármacos , Fenilisopropiladenosina/antagonistas & inibidores , Ratos , Ratos Wistar , Receptor A1 de Adenosina/efeitos dos fármacos , Estimulação Química
8.
Am J Physiol Cell Physiol ; 298(1): C56-65, 2010 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-19828838

RESUMO

Exposure of cells to adenosine receptor (AR) agonists leads to receptor uncoupling from G proteins and downregulation of the A(1)AR. The receptor levels on the cell surface generally recover on withdrawal of the agonist, because of either translocation of the sequestered A(1)AR back to plasma membrane or de novo synthesis of A(1)AR. To examine the mechanism(s) underlying A(1)AR downregulation and recovery, we treated ductus deferens tumor (DDT(1) MF-2) cells with the agonist R-phenylisopropyladenosine (R-PIA) and showed a decrease in membrane A(1)AR levels by 24 h, which was associated with an unexpected 11-fold increase in A(1)AR mRNA. Acute exposure of these cells to R-PIA resulted in a rapid translocation of beta-arrestin1 to the plasma membrane. Knockdown of beta-arrestin1 by short interfering RNA (siRNA) blocked R-PIA-mediated downregulation of the A(1)AR, suppressed R-PIA-dependent ERK1/2 and activator protein-1 (AP-1) activity, and reduced the induction of A(1)AR mRNA. Withdrawal of the agonist after a 24-h exposure resulted in rapid recovery of plasma membrane A(1)AR. This was dependent on the de novo protein synthesis and on the activity of ERK1/2 but independent of beta-arrestin1 and nuclear factor-kappaB. Together, these data suggest that exposure to A(1)AR agonist stimulates ERK1/2 activity via beta-arrestin1, which subserves receptor uncoupling and downregulation, in addition to the induction of A(1)AR expression. We propose that such a pathway ensures both the termination of the agonist signal and recovery by priming the cell for rapid de novo synthesis of A(1)AR once the drug is terminated.


Assuntos
Arrestinas/fisiologia , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Receptor A1 de Adenosina/fisiologia , Animais , Arrestinas/genética , Western Blotting , Técnicas de Cultura de Células , Divisão Celular , Cricetinae , Imuno-Histoquímica , Masculino , Mesocricetus , Músculo Liso/citologia , Músculo Liso/fisiologia , Fenilisopropiladenosina/farmacologia , RNA Interferente Pequeno/genética , Receptor A1 de Adenosina/efeitos dos fármacos , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transfecção , Ducto Deferente/citologia , Ducto Deferente/fisiologia , beta-Arrestinas
9.
Neuroscience ; 163(2): 524-32, 2009 Oct 06.
Artigo em Inglês | MEDLINE | ID: mdl-19559760

RESUMO

Little is known about the G protein-coupled receptor desensitization process during pregnancy. Wistar pregnant rats were treated with (-)N(6)-phenylisopropyladenosine (R-PIA), an adenosine A(1) receptor (A(1)R) agonist, in their drinking water during pregnancy, and the effect on A(1)R/adenylyl cyclase system was studied in both maternal and fetal brain. In maternal brain, binding assays revealed a significant decrease in total receptor numbers in plasma membranes (27%, P<0.05), with no significant changes in receptor affinity. The effect of R-PIA on plasma membranes from fetal brains was more marked, with approximately 42% (P<0.05) of the total receptors detected in control fetuses. Real time reverse transcriptase polymerase chain reaction (RT-PCR) analyses showed that chronic R-PIA treatment during the whole gestational period only decreased significantly mRNA level coding A(1)R in maternal brain (P<0.05). alpha Gi(1,2) and alpha Gi(3) subunits were not affected in mothers or fetuses as revealed by immunoblotting. mRNA levels coding these subunits were also unaffected in mothers and fetuses. On the other hand, forskolin- and forskolin-plus guanosine-5'-O-(3-thiotriphosphate) (GTP gamma S)-stimulated adenylyl cyclase activity was decreased in maternal (P<.01) and fetal brain (P<.001). Furthermore, adenylyl cyclase inhibition elicited by N(6)-cyclohexyladenosine (CHA), a selective A(1)R agonist, was significantly decreased in both maternal (P<0.05) and fetal brain (P<.01), suggesting a desensitization of the A(1)R/adenylyl cyclase pathway. Therefore, these results suggest that R-PIA intake during pregnancy causes desensitization of the A(1)R-mediated inhibitory transduction pathway in both maternal and fetal brain, probably due to the decreased density of A(1)R at the cell surface.


Assuntos
Adenilil Ciclases/metabolismo , Encéfalo/efeitos dos fármacos , Encéfalo/metabolismo , Fármacos do Sistema Nervoso Central/farmacologia , Fenilisopropiladenosina/farmacologia , Receptor A1 de Adenosina/metabolismo , Adenosina/análogos & derivados , Adenosina/farmacologia , Agonistas do Receptor A1 de Adenosina , Inibidores de Adenilil Ciclases , Animais , Encéfalo/embriologia , Membrana Celular/efeitos dos fármacos , Membrana Celular/metabolismo , Colforsina/farmacologia , Feminino , Subunidade alfa Gi2 de Proteína de Ligação ao GTP/metabolismo , Subunidades alfa Gi-Go de Proteínas de Ligação ao GTP/metabolismo , Gravidez , RNA Mensageiro/metabolismo , Ratos , Ratos Wistar , Transdução de Sinais/efeitos dos fármacos
10.
Anesthesiology ; 107(5): 797-806, 2007 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-18073555

RESUMO

BACKGROUND: Activation of A1 adenosine receptors (A1Rs) causes antinociception after nerve injury and inflammation. However, the role of A2a adenosine receptors (A2aRs) for pain processing is less clear. In the current study, the authors investigated the role of spinal adenosine A1Rs and A2aRs for the maintenance of mechanical hyperalgesia in an animal model for postoperative pain. METHODS: Rats with intrathecal catheters were anesthetized and underwent plantar incision. Spontaneous pain behavior and withdrawal threshold to punctuate stimulation were measured before and after administration of intrathecal R-phenylisopropyl-adenosine (R-PIA; A1R agonist), 2-w p-2-carbonyl-ethyl-phenylethylaminox-5X-N-ethylcarboxami-doadenosine (CGS21680; A2aR agonist), or vehicle. In separate groups of animals, the effects of pertussis toxin, forskolin, glibenclamide, 4-aminopyridine, tetraethylammonium, apamin, charybdotoxin, or margatoxin on R-PIA-induced antinociception were examined. RESULTS: Intrathecal administration of 5 nmol R-PIA but not 10 nmol CGS21680 decreased nonevoked spontaneous pain behavior. Furthermore, intrathecal administration of R-PIA but not of CGS21680 increased withdrawal thresholds after incision. Pretreatment with pertussis toxin and administration of forskolin, glibenclamide, 4-aminopyridine, and tetraethylammonium inhibited R-PIA-induced antinociception. In addition, intrathecal administration of apamin, charybdotoxin, or margatoxin did not modify mechanical hypoalgesia mediated by R-PIA. CONCLUSIONS: Spinal A1Rs but not A2aRs play an important role in the maintenance of nonevoked and evoked pain behaviors after an incision. Furthermore, A1R-induced spinal antinociception is mediated by interactions with pertussis toxin-sensitive G proteins. In addition, the opening of adenosine triphosphate-sensitive K channels but not of calcium-activated potassium channels and voltage-gated Kv1.3 or Kv1.6 channels contribute to the antinociceptive effect of A1R agonists.


Assuntos
Agonistas do Receptor A1 de Adenosina , Agonistas do Receptor A2 de Adenosina , Adenosina/análogos & derivados , Proteínas de Ligação ao GTP/efeitos dos fármacos , Hiperalgesia/tratamento farmacológico , Toxina Pertussis/farmacologia , Procedimentos Cirúrgicos Operatórios/efeitos adversos , Adenosina/administração & dosagem , Adenosina/farmacologia , Adenosina-5'-(N-etilcarboxamida)/administração & dosagem , Adenosina-5'-(N-etilcarboxamida)/farmacologia , Animais , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Proteínas de Ligação ao GTP/metabolismo , Hiperalgesia/etiologia , Masculino , Dor Pós-Operatória/tratamento farmacológico , Fenilisopropiladenosina/administração & dosagem , Fenilisopropiladenosina/farmacologia , Estimulação Física , Bloqueadores dos Canais de Potássio/administração & dosagem , Bloqueadores dos Canais de Potássio/farmacologia , Canais de Potássio/efeitos dos fármacos , Ratos , Ratos Sprague-Dawley , Vasodilatadores/administração & dosagem , Vasodilatadores/farmacologia
11.
J Lipid Res ; 48(5): 1099-107, 2007 May.
Artigo em Inglês | MEDLINE | ID: mdl-17267946

RESUMO

Adipocyte lipolysis is dependent on an increase in the intracellular concentration of cAMP. Intracellular phosphodiesterases (PDEs) hydrolyze cAMP and limit stimulation of lipolysis. In the present study, the mRNA expression of PDE4 subtypes and the antilipolytic role of PDE4 in rat adipocytes were investigated. Fragments encoding PDE4A (233 bp), PDE4B (786 bp), PDE4C (539 bp), and PDE4D (262 bp) sequences were amplified by RT-PCR. The mRNA expression of PDE4 subtypes (A, B, C, D) determined by real-time quantitative PCR was 7, 18.7, 18.9, and 7.2% relative to PDE3B. Inhibition of PDE4 by rolipram increased basal lipolysis and reversed in part prostaglandin E2 antilipolysis. The combination of PDE3 and PDE4 inhibitors synergistically reversed both prostaglandin E2 and phenylisopropyl adenosine antilipolysis. Stimulation of adipocytes with prostaglandin E2 increased total PDE activity and PDE3 activity measured by hydrolysis of 3[H]cAMP by the particulate fraction of adipocytes. The present study confirmed that mRNAs for all four PDE4 subtypes were expressed in rat adipocytes, with PDE4B and PDE4C predominant. Moreover, PDE4 not only limits the rate of basal lipolysis but also contributes to prostaglandin E2 antilipolysis in rat adipocytes.


Assuntos
3',5'-AMP Cíclico Fosfodiesterases/metabolismo , Adipócitos/enzimologia , Regulação Enzimológica da Expressão Gênica , Lipólise , 3',5'-AMP Cíclico Fosfodiesterases/antagonistas & inibidores , 3',5'-AMP Cíclico Fosfodiesterases/classificação , 3',5'-AMP Cíclico Fosfodiesterases/genética , Adipócitos/efeitos dos fármacos , Animais , Sequência de Bases , Nucleotídeo Cíclico Fosfodiesterase do Tipo 3 , Nucleotídeo Cíclico Fosfodiesterase do Tipo 4 , Dinoprostona/farmacologia , Inibidores Enzimáticos/farmacologia , Insulina/farmacologia , Isoproterenol/farmacologia , Lipólise/efeitos dos fármacos , Masculino , Dados de Sequência Molecular , Fenilisopropiladenosina/farmacologia , RNA Mensageiro/genética , Ratos , Ratos Sprague-Dawley , Alinhamento de Sequência
12.
Hum Exp Toxicol ; 24(7): 369-75, 2005 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-16119251

RESUMO

The objective of this study was to evaluate the effects of an adenosine A1 agonist, phenylisopropyl adenosine (PIA), on metamidophos poisoning compared to specific antidotes. Rats were poisoned with metamidophos (30 mg/kg, oral) and observed for 24 hours. One group received sodium chloride (1 mL/kg) and four experimental groups received atropine (5 mg/kg), pralidoxime (PAM, 20 mg/kg), atropine/PAM (5/20 mg/kg) or PIA (1 mg/kg) intraperitoneally. Atropine reduced salivation and prevented respiratory distress when compared to sodium chloride-treated rats. Treatment with PAM did not cause any suppression of cholinergic signs. Atropine and PAM combination prevented salivation, convulsion and respiratory distress. PIA delayed initial time of the salivation, convulsion and time to death. However, PIA was found ineffective against the metamidophos-induced cholinergic symptoms and mortality. All treatments, except PIA, lead to survival of these animals. Acetylcholinesterase (AChE) activity was not normalized by PIA or PAM. PIA prevented metamidophos-induced diaphragmatic muscle necrosis as much as PAM. In conclusion, a single dose of PIA was unable to protect the rats from metamidophos toxicity. Further studies are needed involving a combination of PAM and/or atropine with repeated doses of PIA to clarify the efficacy of adenosine agonists in OP poisoning.


Assuntos
Agonistas do Receptor A1 de Adenosina , Antídotos/farmacologia , Inseticidas/envenenamento , Compostos Organotiofosforados/envenenamento , Fenilisopropiladenosina/farmacologia , Animais , Antídotos/administração & dosagem , Atropina/administração & dosagem , Reativadores da Colinesterase/administração & dosagem , Colinesterases/sangue , Colinesterases/metabolismo , Diafragma/efeitos dos fármacos , Diafragma/patologia , Interações Medicamentosas , Masculino , Modelos Animais , Antagonistas Muscarínicos/administração & dosagem , Fenilisopropiladenosina/administração & dosagem , Intoxicação/sangue , Intoxicação/mortalidade , Compostos de Pralidoxima/administração & dosagem , Ratos , Ratos Wistar , Respiração/efeitos dos fármacos , Salivação/efeitos dos fármacos , Taxa de Sobrevida , Fatores de Tempo
13.
Mol Cell Biochem ; 270(1-2): 209-14, 2005 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-15792369

RESUMO

This study investigated the interactive effects of acute exercise and adenosine receptor agonist and antagonist on antioxidant enzyme activities, glutathione and lipid peroxidation in the heart of the rat. Male Fisher-344 rats were divided into six groups and treated as follows: (1) saline control; (2) acute exercise (100% VO2max); (3) R-Phenyl isopropyl adenosine (R-PIA) (3.46 micromol/kg, i.p.); (4) theophylline (1.70 micromol/kg, i.p.) plus acute exercise; (5) theophylline plus R-PIA; and (6) theophylline. Animals were sacrificed 1 h after treatments; hearts were isolated and analyzed. The results show that acute exercise as well as adenosine receptor agonist R-PIA significantly enhanced cardiac superoxide dismutase (SOD), catalase (CAT), glutathione peroxidase (GSH-Px), and glutathione reductase (GR) activity by 36-135% and 16-51%, respectively. Adenosine receptor agonist R-PIA significantly decreased cardiac GSSG concentration and enhanced GSH/GSSG ratio by 22 and 30%, respectively. Whereas theophylline treatment blocked the activation of antioxidant enzyme activities enhanced by acute exercise and R-PIA. Theophylline treatment significantly increased lipid peroxidation by 43% in the heart of exercised rats. The study concluded that the adenosine receptors are involved in the upregulation of cardiac antioxidant defense system and attenuation of lipid peroxidation due to acute exercise in rats.


Assuntos
Antioxidantes/farmacologia , Miocárdio/patologia , Condicionamento Físico Animal , Agonistas do Receptor Purinérgico P1 , Animais , Antioxidantes/metabolismo , Catalase/metabolismo , Dissulfetos/química , Glutationa/metabolismo , Glutationa Peroxidase/metabolismo , Glutationa Redutase/metabolismo , Peroxidação de Lipídeos , Masculino , Miocárdio/enzimologia , Consumo de Oxigênio , Fenilisopropiladenosina/farmacologia , Ratos , Ratos Endogâmicos F344 , Superóxido Dismutase/metabolismo , Teofilina/farmacologia
14.
Acta Pharmacol Sin ; 26(2): 181-5, 2005 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-15663896

RESUMO

AIM: To investigate the antinociceptive effect of adenosine agonist R-phenylisopropyl-adenosine (R-PIA) given to conscious rats by intracerebroventricular (ICV) and intrathecal (IT), and identify the effect of R-PIA on minimum alveolar concentration (MAC) of halothane with pretreatment of A1 receptor antagonist 8-cyclopentyl-1,3-dipropylxanthine (DPCPX) or K+ channel blocker 4-aminopyridine (4-AP). METHODS: Sprague-Dawley rats were implanted with 24-gauge stainless steel guide cannula using stereotaxic apparatus and ICV method, and an IT catheter (PE-10, 8.5 cm) was inserted into the lumbar subarachnoid space, while the rats were under pentobarbital anesthesia. After one week of recovery from surgery, rats were randomly assigned to one of the following protocols: MAC of halothane, or tail-flick latency. All measurements were performed after R-PIA (0.8-2.0 microg) microinjection into ICV and IT with or without pretreatment of DPCPX or 4-AP. RESULTS: Microinjection of adenosine agonist R-PIA in doses of 0.8-2.0 microg into ICV and IT produced a significant dose- and time-dependent antinociceptive action as reflected by increasing latency times and ICV administration of adenosine agonist R-PIA (0.8 microg) reducing halothane anesthetic requirements (by 29%). The antinociception and reducing halothane requirements effected by adenosine agonist R-PIA was abolished by DPCPX and 4-AP. CONCLUSION: ICV and IT administration of adenosine agonist R-PIA produced an antinociceptive effect in a dose-dependent manner and decreased halothane MAC with painful stimulation through activation of A1 receptor subtype, and the underlying mechanism involves K+ channel activation.


Assuntos
Analgesia , Analgésicos/farmacologia , Anestesia , Halotano/administração & dosagem , Fenilisopropiladenosina/farmacologia , 4-Aminopiridina/farmacologia , Adenosina/agonistas , Antagonistas do Receptor A1 de Adenosina , Analgésicos/administração & dosagem , Anestésicos Inalatórios/administração & dosagem , Animais , Relação Dose-Resposta a Droga , Injeções Intraventriculares , Injeções Espinhais , Masculino , Microinjeções , Fenilisopropiladenosina/administração & dosagem , Fenilisopropiladenosina/antagonistas & inibidores , Bloqueadores dos Canais de Potássio/farmacologia , Distribuição Aleatória , Ratos , Ratos Sprague-Dawley , Xantinas/farmacologia
15.
Anesth Analg ; 100(2): 461-468, 2005 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-15673876

RESUMO

Nerve ligation injury may produce a tactile allodynia. Intrathecal adenosine receptor agonists or morphine have an antiallodynic effect. In this study, we examined the effect of intrathecal morphine on the antiallodynic state induced by the adenosine A1 receptor agonist, N(6)-(2-phenylisopropyl)-adenosine R-(-)isomer (R-PIA), in a rat model of nerve ligation injury. Rats were prepared with ligation of left L5-6 spinal nerves and intrathecal catheter implantation. Tactile allodynia was measured by applying von Frey filaments to the lesioned hindpaw. Thresholds for withdrawal response were assessed. Morphine and R-PIA were administered to obtain the dose-response curve and the 50% effective dose (ED(50)). Fractions of ED(50)s were administered concurrently to establish the ED(50) of the drug combination. The drug interaction was analyzed using the isobolographic method. Intrathecal 8-cyclopentyl-1,3-dipropylxanthine (DPCPX), an A1 receptor antagonist, and naloxone were administered to examine the reversal of the antiallodynic effect. Side effects were also observed. Intrathecal morphine and R-PIA and their combination produced a dose-dependent antagonism without severe side effects. Intrathecal morphine synergistically enhanced the antiallodynic effect of R-PIA when coadministered. Intrathecal naloxone and DPCPX reversed the maximal antiallodynic effect in the combination group. These results suggest that activation of mu-opioid and A1 receptors at the spinal level is required for the synergistic interaction on tactile allodynia.


Assuntos
Agonistas do Receptor A1 de Adenosina , Analgésicos Opioides/uso terapêutico , Morfina/uso terapêutico , Dor/tratamento farmacológico , Doenças do Sistema Nervoso Periférico/complicações , Fenilisopropiladenosina/uso terapêutico , Analgésicos Opioides/administração & dosagem , Animais , Ritmo Circadiano , Relação Dose-Resposta a Droga , Sinergismo Farmacológico , Injeções Espinhais , Ligadura , Masculino , Morfina/administração & dosagem , Naloxona/farmacologia , Antagonistas de Entorpecentes/farmacologia , Dor/etiologia , Medição da Dor/efeitos dos fármacos , Fenilisopropiladenosina/administração & dosagem , Ratos , Ratos Sprague-Dawley , Xantinas/uso terapêutico
16.
Life Sci ; 73(4): 393-401, 2003 Jun 13.
Artigo em Inglês | MEDLINE | ID: mdl-12759134

RESUMO

Prolonged treatment of adipocytes with certain inhibitors of lipolysis, including N(6)-phenylisopropyl adenosine (PIA) and prostaglandin E(1) (PGE(1)) leads to down-regulation of G(i). Prolonged treatment with PIA increases the rate of lipolysis, and we have reported that tumor necrosis factor-alpha (TNF alpha) stimulates lipolysis by down-regulating G(i). To determine the relationship between G(i) concentration and lipolysis, we have investigated the effect of two other acute inhibitors of lipolysis; PGE(1), which down-regulates G(i), and nicotinic acid (NA), which does not down-regulate G(i). Rat adipocytes were incubated with PIA (300 nM), PGE(1) (3 microM) or nicotinic acid (1 mM) for 24 h. The rate of lipolysis (glycerol release) was increased approximately 2 to 3-fold in PIA-treated cells, and in PGE(1)-treated cells. Conversely, the rate of lipolysis was not altered by the prolonged nicotinic acid treatment. These findings support the hypothesis that the rate of lipolysis in adipocytes is determined, at least partly, by the cellular concentration of G(i) proteins.


Assuntos
Adipócitos/metabolismo , Alprostadil/farmacologia , Animais , Relação Dose-Resposta a Droga , Regulação para Baixo , Ácidos Graxos/metabolismo , Subunidades alfa Gi-Go de Proteínas de Ligação ao GTP/metabolismo , Resistência à Insulina , Lipólise , Niacina/metabolismo , Niacina/farmacologia , Fenilisopropiladenosina/farmacologia , Ratos , Fatores de Tempo
17.
Phytother Res ; 17(3): 279-81, 2003 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-12672162

RESUMO

The antihypoxic activity of Allium sativum clove (garlic) aqueous and methanolic extracts was studied in mice. The extracts of garlic showed that the antihypoxic effect was dose-dependent. The minimum effective doses of aqueous and methanolic extracts were 0.2 g/kg and 5.12 g/kg, respectively. Phenytoin, 50 mg/kg, and R-phenylisopropyladenosine (R-PIA), 1.6 mg/kg (R-PIA) as positive controls increased survival time up to 52.5 +/- 2.9 min and 120.5 +/- 6 min, respectively, compared to normal saline (34.73 +/- 0.71 min). The high doses of aqueous (16.9 g/kg) and methanolic (12.8 g/kg) extracts increased survival time up to 73.17 +/- 4.9 and 68.41 +/- 3.7, respectively. These results indicated that the extracts of A. sativum cloves have a protective effect against hypoxia-induced lethality in mice.


Assuntos
Alho , Hipóxia Encefálica/metabolismo , Fármacos Neuroprotetores/farmacologia , Fitoterapia , Extratos Vegetais/farmacologia , Animais , Relação Dose-Resposta a Droga , Feminino , Injeções Intraperitoneais , Masculino , Camundongos , Fármacos Neuroprotetores/administração & dosagem , Fármacos Neuroprotetores/uso terapêutico , Fenilisopropiladenosina/administração & dosagem , Fenilisopropiladenosina/farmacologia , Fenilisopropiladenosina/uso terapêutico , Fenitoína/administração & dosagem , Fenitoína/farmacologia , Fenitoína/uso terapêutico , Extratos Vegetais/administração & dosagem , Extratos Vegetais/uso terapêutico
18.
Exp Nephrol ; 10(1): 43-50, 2002.
Artigo em Inglês | MEDLINE | ID: mdl-11803204

RESUMO

We have compared renal A(1) adenosine receptor (AR) regulations in rats after chronic agonist and antagonist treatments. In one group, R-phenylisopropyladenosine (R-PIA), a selective A(1) AR agonist, was infused subcutaneously for 7 days. Another group was fed theophylline, a non-selective AR antagonist, for 2 weeks. Renal cortex membrane A(1) AR binding with 1,3-[(3)H]-dipropyl-8-cyclopentylxanthine demonstrated approximately 40% reduction in the B(max ) for the R-PIA group without any changes in the K(d) values. Neither the B(max) nor the K(d) changed following chronic theophylline treatment. Renal cortex G(i)alpha-proteins from the R-PIA treated rats decreased by approximately 30%. Renal G(i)alpha levels did not change in theophylline-treated rats. Consistent with the A(1) AR desensitization, R-PIA-treated rats had significantly higher basal renin release and showed attenuated A(1) AR-mediated inhibition of renin release. These data suggest that prolonged A(1) AR stimulation results in downregulation of renal A(1) ARs and G(i)alpha, accompanied by desensitization of A(1) AR-mediated inhibitory effects on renin release. Unlike cardiac and brain A(1) ARs, renal A(1) receptors are not subject to up-regulation following chronic antagonist treatment.


Assuntos
Rim/metabolismo , Receptores Purinérgicos P1/metabolismo , Adenosina/análogos & derivados , Adenosina/farmacologia , Animais , Membrana Celular/metabolismo , Proteínas de Ligação ao GTP/imunologia , Proteínas de Ligação ao GTP/metabolismo , Immunoblotting , Rim/efeitos dos fármacos , Córtex Renal/metabolismo , Cinética , Masculino , Fenilisopropiladenosina/farmacologia , Inibidores de Fosfodiesterase/farmacologia , Agonistas do Receptor Purinérgico P1 , Antagonistas de Receptores Purinérgicos P1 , Ratos , Ratos Wistar , Renina/metabolismo , Teofilina/farmacologia , Fatores de Tempo , Xantinas/metabolismo
19.
Endocrinology ; 142(12): 5069-75, 2001 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-11713199

RESUMO

Prolonged treatment of rat adipocytes with TNF alpha increases lipolysis through a mechanism mediated, in part, by down-regulation of inhibitory G proteins (G(i)). Separately, down-regulation of G(i) by prolonged treatment with an A(1)-adenosine receptor agonist, N(6)-phenylisopropyl adenosine (PIA) increases lipolysis. To investigate the role of proteolysis in TNF alpha and PIA-mediated G(i) down-regulation and stimulation of lipolysis, we used the protease inhibitors lactacystin (proteasome inhibitor) and calpeptin (calpain inhibitor). Rat adipocytes were preincubated for 1 h with lactacystin (10 microM) or calpeptin (50 microM), before 30-h treatment with either TNF alpha (50 ng/ml) or PIA (300 nM). We then measured lipolysis (glycerol release), abundance of alpha-subunits of G(i)1 and G(i)2 in plasma membranes (Western blotting) and protease activities (in specific fluorogenic assays). TNF alpha and PIA stimulated lipolysis approximately 2-fold and caused G(i) down-regulation. Although neither lactacystin nor calpeptin affected basal lipolysis, lactacystin completely inhibited both TNF alpha and PIA-stimulated lipolysis (the 50% inhibitory concentration was approximately 2 microM), whereas calpeptin had no effect. Similarly, lactacystin but not calpeptin blocked both PIA and TNF alpha-induced G(i) down-regulation. These findings provide further evidence that the chronic lipolytic effect of TNF alpha and PIA is secondary to G(i) down-regulation and suggest that the mechanism involves proteolytic degradation mediated through the proteasome pathway.


Assuntos
Acetilcisteína/análogos & derivados , Adipócitos/metabolismo , Cisteína Endopeptidases/fisiologia , Subunidades alfa Gi-Go de Proteínas de Ligação ao GTP/metabolismo , Lipólise/fisiologia , Complexos Multienzimáticos/fisiologia , Fator de Necrose Tumoral alfa/fisiologia , Acetilcisteína/farmacologia , Animais , Calpaína/antagonistas & inibidores , Calpaína/fisiologia , Células Cultivadas , Inibidores de Cisteína Proteinase/farmacologia , Dipeptídeos/farmacologia , Regulação para Baixo/fisiologia , Lipólise/efeitos dos fármacos , Masculino , Complexos Multienzimáticos/antagonistas & inibidores , Fenilisopropiladenosina/farmacologia , Complexo de Endopeptidases do Proteassoma , Ratos , Ratos Sprague-Dawley
20.
J Appl Physiol (1985) ; 91(2): 552-60, 2001 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-11457765

RESUMO

The purpose of this study was to pharmacologically characterize the adenosine receptor subtype(s) that mediates adenosine-induced increases in macromolecular efflux from the intact hamster cheek pouch. Using intravital microscopy, we found that 1,3-dipropyl-8-(2-amino-4-chlorophenyl)-xanthine (PACPX), a selective adenosine receptor-1 antagonist, but not 3,7-dimethyl-1-propargylxanthine (DMPX), a selective adenosine receptor-2 antagonist, significantly attenuated adenosine-induced leaky site formation and increased clearance of fluorescein isothiocyanate-labeled dextran (molecular mass, 70 kDa) from the intact hamster cheek pouch (P < 0.05). Both compounds had no significant effects on bradykinin-induced responses. Nanomolar concentrations of R(-)-N(6)-(2-phenylisopropyl)-adenosine [R(-)-PIA], a selective adenosine A(1) agonist, evoked significant, concentration-dependent increases in macromolecular efflux. This response was significantly attenuated by PACPX but not by DMPX. In contrast, CGS-21680, a selective adenosine A(2) agonist, increased macromolecular efflux but only at micromolar concentrations. This response was significantly attenuated by DMPX but not by PACPX. Suffusion of nitroglycerin had no significant effects on R(-)-PIA- and CGS-21680-induced responses. In addition, suffusion of N(G)-nitro-L-arginine methyl ester, a nitric oxide synthase inhibitor, had no significant effects on adenosine-induced responses. Indomethacin had no significant effects on adenosine-, R(-)-PIA-, and CGS-21680-induced increases in macromolecular efflux. Collectively, these data indicate that adenosine increases macromolecular efflux from the intact hamster cheek pouch by stimulating high-affinity adenosine A(1) receptors in a specific, nitric oxide- and prostaglandin-independent fashion.


Assuntos
Exsudatos e Transudatos/metabolismo , Mucosa Bucal/fisiologia , Receptores Purinérgicos P1/fisiologia , Teobromina/farmacologia , Xantinas/farmacologia , Adenosina/análogos & derivados , Adenosina/farmacologia , Animais , Bradicinina/farmacologia , Cricetinae , Dextranos/farmacocinética , Exsudatos e Transudatos/efeitos dos fármacos , Fluoresceína-5-Isotiocianato/análogos & derivados , Fluoresceína-5-Isotiocianato/farmacocinética , Indometacina/farmacologia , Masculino , Mesocricetus , Mucosa Bucal/efeitos dos fármacos , NG-Nitroarginina Metil Éster/farmacologia , Nitroglicerina/farmacologia , Fenetilaminas/farmacologia , Fenilisopropiladenosina/farmacologia , Agonistas do Receptor Purinérgico P1 , Antagonistas de Receptores Purinérgicos P1 , Teobromina/análogos & derivados
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...