Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 57
Filtrar
1.
Biochem Pharmacol ; 192: 114739, 2021 10.
Artigo em Inglês | MEDLINE | ID: mdl-34418353

RESUMO

Hepatic insulin resistance (IR) and enhanced hepatic glucose production (HGP) are key features of type 2 diabetes (T2D), contributing to fasting hyperglycemia. Adenosine receptors (ARs) are G protein-coupled and expressed in hepatocytes. Here, we explored the role of hepatic Gi/o-coupled A1AR on insulin resistance and glucose fluxes associated with obesity. We generated a mouse model with hepatocyte-specific deletion of A1AR (A1LΔ/Δ), which was compared with whole body knockout of A1AR or A1AR/A3AR (both Gi-coupled). Selective deletion of hepatic A1AR resulted in a modest improvement in insulin sensitivity. In addition, HFD A1LΔ/Δ mice showed decreased fasting glucose levels. Hyperinsulinemic-euglycemic clamp studies demonstrated enhanced insulin sensitivity with no change in HGP in HFD A1LΔ/Δ mice. Similar to A1LΔ/Δ, fasting blood glucose levels were significantly reduced in whole body A1Δ/Δ and A1Δ/ΔA3Δ/Δ compared to wild-type mice. Taken together, our data support the concept that blocking hepatic A1AR may decrease fasting blood glucose levels without directly affecting hepatocyte glucose metabolism and insulin sensitivity.


Assuntos
Diabetes Mellitus Experimental/metabolismo , Glucose/metabolismo , Hepatócitos/metabolismo , Resistência à Insulina/fisiologia , Receptor A1 de Adenosina/deficiência , Animais , Diabetes Mellitus Experimental/genética , Dieta Hiperlipídica/efeitos adversos , Insulina/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Receptor A1 de Adenosina/genética
2.
J Neurophysiol ; 122(2): 721-728, 2019 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-31242045

RESUMO

Adenosine receptors are widely expressed in the brain, and adenosine is a key bioactive substance for neuroprotection. In this article, we clarify systematically the role of adenosine A1 receptors during a range of timescales and conditions when a significant amount of adenosine is released. Using acute hippocampal slices obtained from mice that were wild type or null mutant for the adenosine A1 receptor, we quantified and characterized the impact of varying durations of experimental ischemia, hypoxia, and hypoglycemia on synaptic transmission in the CA1 subregion. In normal tissue, these three stressors rapidly and markedly reduced synaptic transmission, and only treatment of sufficient duration led to incomplete recovery. In contrast, inactivation of adenosine A1 receptors delayed and/or lessened the reduction in synaptic transmission during all three stressors and reduced the magnitude of the recovery significantly. We reproduced the responses to hypoxia and hypoglycemia by applying an adenosine A1 receptor antagonist, validating the clear effects of genetic receptor inactivation on synaptic transmission. We found activation of adenosine A1 receptor inhibited hippocampal synaptic transmission during the acute phase of ischemia, hypoxia, or hypoglycemia and caused the recovery from synaptic impairment after these three stressors using genetic mutant. These studies quantify the neuroprotective role of the adenosine A1 receptor during a variety of metabolic stresses within the same recording system.NEW & NOTEWORTHY Deprivation of oxygen and/or glucose causes a rapid adenosine A1 receptor-mediated decrease in synaptic transmission in mouse hippocampus. We quantified adenosine A1 receptor-mediated inhibition during and synaptic recovery after ischemia, hypoxia, and hypoglycemia of varying durations using a genetic mutant and confirmed these findings using pharmacology. Overall, using the same recording conditions, we found the acute response and the neuroprotective ability of the adenosine A1 receptor depended on the type and duration of deprivation event.


Assuntos
Região CA1 Hipocampal/metabolismo , Hipoglicemia/metabolismo , Hipóxia/metabolismo , Isquemia/metabolismo , Receptor A1 de Adenosina/fisiologia , Estresse Fisiológico/fisiologia , Transmissão Sináptica/fisiologia , Antagonistas do Receptor A1 de Adenosina/farmacologia , Animais , Região CA1 Hipocampal/efeitos dos fármacos , Feminino , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Receptor A1 de Adenosina/deficiência , Estresse Fisiológico/efeitos dos fármacos , Transmissão Sináptica/efeitos dos fármacos
3.
Am J Physiol Renal Physiol ; 314(5): F788-F797, 2018 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-29117994

RESUMO

Sepsis and acute kidney injury (AKI) synergistically increase morbidity and mortality in the ICU. How sepsis reduces glomerular filtration rate (GFR) and causes AKI is poorly understood; one proposed mechanism includes tubuloglomerular feedback (TGF). When sodium reabsorption by the proximal tubules is reduced in normal animals, the macula densa senses increased luminal sodium chloride, and then adenosine-1a receptor (A1aR) signaling triggers tubuloglomerular feedback, reducing GFR through afferent arteriole vasoconstriction. We measured GFR and systemic hemodynamics early during cecal ligation and puncture-induced sepsis in wild-type and A1aR-knockout mice. A miniaturized fluorometer was attached to the back of each mouse and recorded the clearance of FITC-sinistrin via transcutaneous fluorescence to monitor GFR. Clinical organ injury markers and cytokines were measured and hemodynamics monitored using implantable transducer telemetry devices. In wild-type mice, GFR was stable within 1 h after surgery, declined by 43% in the next hour, and then fell to less than 10% of baseline after 2 h and 45 min. In contrast, in A1aR-knockout mice GFR was 37% below baseline immediately after surgery and then gradually declined over 4 h. A1aR-knockout mice had similar organ injury and inflammatory responses, albeit with lower heart rate. We conclude that transcutaneous fluorescence can accurately monitor GFR and detect changes rapidly during sepsis. Tubuloglomerular feedback plays a complex role in sepsis; initially, TGF helps maintain GFR in the 1st hour, and over the subsequent 3 h, TGF causes GFR to plummet. By 18 h, TGF has no cumulative effect on renal or extrarenal organ damage.


Assuntos
Injúria Renal Aguda/metabolismo , Taxa de Filtração Glomerular , Rim/metabolismo , Receptor A1 de Adenosina/metabolismo , Sepse/metabolismo , Injúria Renal Aguda/etiologia , Injúria Renal Aguda/genética , Injúria Renal Aguda/fisiopatologia , Animais , Modelos Animais de Doenças , Retroalimentação Fisiológica , Fluoresceínas/administração & dosagem , Corantes Fluorescentes/administração & dosagem , Corantes Fluorescentes/metabolismo , Fluorometria/métodos , Hemodinâmica , Injeções Intravenosas , Rim/fisiopatologia , Camundongos Endogâmicos C57BL , Camundongos Knockout , Oligossacarídeos/administração & dosagem , Oligossacarídeos/sangue , Receptor A1 de Adenosina/deficiência , Receptor A1 de Adenosina/genética , Sepse/complicações , Sepse/genética , Sepse/fisiopatologia , Transdução de Sinais , Fatores de Tempo
4.
Hear Res ; 345: 43-51, 2017 03.
Artigo em Inglês | MEDLINE | ID: mdl-28034618

RESUMO

Our previous studies have shown that the stimulation of A1 adenosine receptors in the inner ear can mitigate the loss of sensory hair cells and hearing loss caused by exposure to traumatic noise. Here, we focus on the role of adenosine receptors (AR) in the development of noise-induced neural injury in the cochlea using A1AR and A2AAR null mice (A1AR-/- and A2AAR-/-). Wildtype (WT) and AR deficient mice were exposed to octave band noise (8-16 kHz, 100 dB SPL) for 2 h to induce cochlear injury and hearing loss. Auditory thresholds and input/output functions were assessed using auditory brainstem responses (ABR) before and two weeks post-exposure. The loss of outer hair cells (OHC), afferent synapses and spiral ganglion neurons (SGN) were assessed by quantitative histology. A1AR-/- mice (6-8 weeks old) displayed a high frequency hearing loss (ABR threshold shift and reduced ABR wave I and II amplitudes). This hearing loss was further aggravated by acute noise exposure and exceeded the hearing loss in the WT and A2AAR-/- mice. All mice experienced the loss of OHC, synaptic ribbons and SGN after noise exposure, but the loss of SGN was significantly higher in A1AR-/- mice than in the A2AAR-/- and WT genotypes. The A2AAR-/- demonstrated better preservation of OHC and afferent synapses and the minimal loss of SGN after noise exposure. The findings suggest that the loss of A1AR expression results in an increased susceptibility to cochlear neural injury and hearing loss, whilst absence of A2AAR increases cochlear resistance to acoustic trauma.


Assuntos
Cóclea/metabolismo , Perda Auditiva Provocada por Ruído/metabolismo , Audição , Ruído/efeitos adversos , Receptor A1 de Adenosina/metabolismo , Receptor A2A de Adenosina/metabolismo , Animais , Limiar Auditivo , Cóclea/lesões , Cóclea/patologia , Cóclea/fisiopatologia , Modelos Animais de Doenças , Potenciais Evocados Auditivos do Tronco Encefálico , Feminino , Predisposição Genética para Doença , Células Ciliadas Auditivas Externas/metabolismo , Células Ciliadas Auditivas Externas/patologia , Perda Auditiva Provocada por Ruído/etiologia , Perda Auditiva Provocada por Ruído/fisiopatologia , Perda Auditiva Provocada por Ruído/prevenção & controle , Masculino , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fenótipo , Fatores de Proteção , Receptor A1 de Adenosina/deficiência , Receptor A1 de Adenosina/genética , Receptor A2A de Adenosina/deficiência , Receptor A2A de Adenosina/genética , Fatores de Risco , Gânglio Espiral da Cóclea/lesões , Gânglio Espiral da Cóclea/metabolismo , Gânglio Espiral da Cóclea/patologia , Sinapses/metabolismo , Sinapses/patologia , Fatores de Tempo
5.
Neuron ; 87(3): 549-62, 2015 Aug 05.
Artigo em Inglês | MEDLINE | ID: mdl-26247862

RESUMO

Major depressive disorder is among the most commonly diagnosed disabling mental diseases. Several non-pharmacological treatments of depression upregulate adenosine concentration and/or adenosine A1 receptors (A1R) in the brain. To test whether enhanced A1R signaling mediates antidepressant effects, we generated a transgenic mouse with enhanced doxycycline-regulated A1R expression, specifically in forebrain neurons. Upregulating A1R led to pronounced acute and chronic resilience toward depressive-like behavior in various tests. Conversely, A1R knockout mice displayed an increased depressive-like behavior and were resistant to the antidepressant effects of sleep deprivation (SD). Various antidepressant treatments increase homer1a expression in medial prefrontal cortex (mPFC). Specific siRNA knockdown of homer1a in mPFC enhanced depressive-like behavior and prevented the antidepressant effects of A1R upregulation, SD, imipramine, and ketamine treatment. In contrast, viral overexpression of homer1a in the mPFC had antidepressant effects. Thus, increased expression of homer1a is a final common pathway mediating the antidepressant effects of different antidepressant treatments.


Assuntos
Proteínas de Transporte/biossíntese , Depressão/metabolismo , Imipramina/uso terapêutico , Ketamina/uso terapêutico , Receptor A1 de Adenosina/biossíntese , Privação do Sono/metabolismo , Animais , Depressão/psicologia , Depressão/terapia , Proteínas de Arcabouço Homer , Humanos , Imipramina/farmacologia , Ketamina/farmacologia , Camundongos , Camundongos Knockout , Camundongos Transgênicos , Córtex Pré-Frontal/efeitos dos fármacos , Córtex Pré-Frontal/metabolismo , Ratos , Receptor A1 de Adenosina/deficiência , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/fisiologia , Privação do Sono/psicologia
6.
Acta Physiol (Oxf) ; 213(1): 259-67, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25182861

RESUMO

AIM: Differences in genetic background between control mice and mice with targeted gene mutations have been recognized as a potential cause for phenotypic differences. In this study, we have used A1AR-deficient mice in a C57Bl/6 and SWR/J congenic background to assess the influence of background on the effect of A1AR-deficiency on cardiovascular and renal functional parameters. METHODS: In A1AR+/+ and A1AR-/- mice in C57Bl/6 and SWR/J congenic backgrounds, we assessed blood pressure and heart rate using radio-telemetry, plasma renin concentrations and tubuloglomerular feedback. RESULTS: We did not detect significant differences in arterial blood pressure (MAP) and heart rates (HR) between A1AR+/+ and A1AR-/- mice in either C57Bl/6, SWR/J or mixed backgrounds. MAP and HR were significantly higher in SWR/J than in C57Bl/6 mice. A high NaCl intake increased MAP in A1AR-/- mice on C57Bl/6 background while there was less or no salt sensitivity in the SWR/J background. No significant differences in plasma renin concentration were detected between A1AR-/- and A1AR+/+ mice in any of the strains. Tubuloglomerular feedback was found to be absent in A1AR-/- mice with SWR/J genetic background. CONCLUSIONS: While this study confirmed important differences between inbred mouse strains, we did not identify phenotypic modifications of A1AR-related effects on blood pressure, heart rate and plasma renin by differences in genetic background.


Assuntos
Pressão Sanguínea/fisiologia , Frequência Cardíaca/fisiologia , Rim/metabolismo , Receptor A1 de Adenosina/metabolismo , Animais , Pressão Sanguínea/genética , Taxa de Filtração Glomerular/genética , Taxa de Filtração Glomerular/fisiologia , Frequência Cardíaca/genética , Camundongos da Linhagem 129 , Camundongos Endogâmicos C57BL , Camundongos Knockout , Receptor A1 de Adenosina/deficiência , Renina/metabolismo
7.
J Lipid Res ; 55(11): 2254-60, 2014 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-25170119

RESUMO

A high-fat low-carbohydrate ketogenic diet (KD) is an effective treatment for refractory epilepsy, yet myriad metabolic effects in vivo have not been reconciled clearly with neuronal effects. A KD limits blood glucose and produces ketone bodies from ß-oxidation of lipids. Studies have explored changes in ketone bodies and/or glucose in the effects of the KD, and glucose is increasingly implicated in neurological conditions. To examine the interaction between altered glucose and the neural effects of a KD, we fed rats and mice a KD and restricted glucose in vitro while examining the seizure-prone CA3 region of acute hippocampal slices. Slices from KD-fed animals were sensitive to small physiological changes in glucose, and showed reduced excitability and seizure propensity. Similar to clinical observations, reduced excitability depended on maintaining reduced glucose. Enhanced glucose sensitivity and reduced excitability were absent in slices obtained from KD-fed mice lacking adenosine A1 receptors (A1Rs); in slices from normal animals effects of the KD could be reversed with blockers of pannexin-1 channels, A1Rs, or KATP channels. Overall, these studies reveal that a KD sensitizes glucose-based regulation of excitability via purinergic mechanisms in the hippocampus and thus link key metabolic and direct neural effects of the KD.


Assuntos
Dieta Cetogênica , Glucose/metabolismo , Hipocampo/fisiologia , Animais , Região CA3 Hipocampal/metabolismo , Região CA3 Hipocampal/fisiologia , Região CA3 Hipocampal/fisiopatologia , Conexinas/metabolismo , Feminino , Técnicas de Inativação de Genes , Hipocampo/metabolismo , Hipocampo/fisiopatologia , Canais KATP/metabolismo , Masculino , Camundongos , Proteínas do Tecido Nervoso/metabolismo , Ratos , Receptor A1 de Adenosina/deficiência , Receptor A1 de Adenosina/genética , Convulsões/metabolismo , Convulsões/fisiopatologia , Convulsões/prevenção & controle
8.
J Virol ; 88(17): 10214-27, 2014 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-24965449

RESUMO

UNLABELLED: We have shown that bronchoalveolar epithelial A1-adenosine receptors (A1-AdoR) are activated in influenza A virus-infected mice. Alveolar macrophages and neutrophils also express A1-AdoRs, and we hypothesized that activation of A1-AdoRs on these cells will promote macrophage and neutrophil chemotaxis and activation and thereby play a role in the pathogenesis of influenza virus-induced acute lung injury. Wild-type (WT) C57BL/6 mice, congenic A1-AdoR knockout (A1-KO) mice, and mice that had undergone reciprocal bone marrow transfer were inoculated intranasally with 10,000 PFU/mouse influenza A/WSN/33 (H1N1) virus. Alternatively, WT mice underwent daily treatment with the A1-AdoR antagonist 8-cyclopentyl-1,3-dipropylxanthine (DPCPX) from 1 day prior to inoculation. Infection increased bronchoalveolar lining fluid (BALF) adenosine comparably in WT and A1-KO mice. Infection of WT mice resulted in reduced carotid arterial O2 saturation (hypoxemia), lung pathology, pulmonary edema, reduced lung compliance, increased basal airway resistance, and hyperresponsiveness to methacholine. These effects were absent or significantly attenuated in A1-KO mice. Levels of BALF leukocytes, gamma interferon (IFN-γ), and interleukin 10 (IL-10) were significantly reduced in infected A1-KO mice, but levels of KC, IP-10, and MCP-1 were increased. Reciprocal bone marrow transfer resulted in WT-like lung injury severity, but BALF leukocyte levels increased only in WT and A1-KO mice with WT bone barrow. Hypoxemia, pulmonary edema, and levels of BALF alveolar macrophages, neutrophils, IFN-γ, and IL-10 were reduced in DPCPX-treated WT mice. Levels of viral replication did not differ between mouse strains or treatment groups. These findings indicate that adenosine activation of leukocyte A1-AdoRs plays a significant role in their recruitment to the infected lung and contributes to influenza pathogenesis. A1-AdoR inhibitor therapy may therefore be beneficial in patients with influenza virus-induced lung injury. IMPORTANCE: Because antiviral drugs are of limited efficacy in patients hospitalized for influenza virus-induced respiratory failure, there is an urgent need for new therapeutics that can limit the progression of lung injury and reduce influenza death rates. We show that influenza A virus infection results in increased production of the nucleoside adenosine in the mouse lung and that activation of A1-subtype adenosine receptors by adenosine contributes significantly to both recruitment of innate immune cells to the lung and development of acute lung injury following influenza virus infection. We also show that treatment with an A1-adenosine receptor antagonist reduces the severity of lung injury in influenza virus-infected mice. Our findings indicate that adenosine plays an important and previously unrecognized role in the innate immune response to influenza virus infection and suggest that drugs which can inhibit either generation of adenosine or activation of A1-adenosine receptors may be beneficial in treating influenza patients hospitalized for respiratory failure.


Assuntos
Lesão Pulmonar Aguda/imunologia , Vírus da Influenza A Subtipo H1N1/fisiologia , Leucócitos/imunologia , Pulmão/imunologia , Infecções por Orthomyxoviridae/imunologia , Receptor A1 de Adenosina/metabolismo , Lesão Pulmonar Aguda/patologia , Transferência Adotiva , Animais , Movimento Celular , Modelos Animais de Doenças , Vírus da Influenza A Subtipo H1N1/imunologia , Leucócitos/fisiologia , Pulmão/patologia , Camundongos Endogâmicos C57BL , Camundongos Knockout , Infecções por Orthomyxoviridae/patologia , Receptor A1 de Adenosina/deficiência
9.
Cardiovasc Res ; 102(1): 157-65, 2014 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-24525840

RESUMO

AIMS: The goal of this study was to determine whether the A1 adenosine receptor (AR) plays a role in atherosclerosis development and to explore its potential mechanisms. METHODS AND RESULTS: Double knockout (DKO) mice, deficient in the genes encoding A1 AR and apolipoprotein E (apoE), demonstrated reduced atherosclerotic lesions in aortic arch (en face), aortic root, and innominate arteries when compared with apoE-deficient mice (APOE-KO) of the same age. Treating APOE-KO with an A1 AR antagonist (DPCPX) also led to a concentration-dependent reduction in lesions. The total plasma cholesterol and triglyceride levels were not different between DKO and APOE-KO; however, higher triglyceride was observed in DKO fed a high-fat diet. DKO also had higher body weights than APOE-KO. Plasma cytokine concentrations (IL-5, IL-6, and IL-13) were significantly lower in DKO. Proliferating cell nuclear antigen expression was also significantly reduced in the aorta from DKO. Despite smaller lesions in DKO, the composition of the innominate artery lesion and cholesterol loading and efflux from bone marrow-derived macrophages of DKO were not different from APOE-KO. CONCLUSION: The A1 AR may play a role in the development of atherosclerosis, possibly due to its pro-inflammatory and mitogenic properties.


Assuntos
Apolipoproteínas E/deficiência , Aterosclerose/metabolismo , Colesterol/metabolismo , Receptor A1 de Adenosina/metabolismo , Animais , Aorta/metabolismo , Modelos Animais de Doenças , Inflamação/metabolismo , Macrófagos/metabolismo , Camundongos Endogâmicos C57BL , Camundongos Knockout , Receptor A1 de Adenosina/deficiência
10.
Acta Physiol (Oxf) ; 210(2): 440-5, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-23901799

RESUMO

AIM: Glomerular hyperfiltration is commonly observed in diabetics early after the onset of the disease and predicts the progression of nephropathy. Sustained hyperglycaemia is also closely associated with kidney hypertrophy and increased electrolyte and glucose reabsorption in the proximal tubule. In this study, we investigated the role of the increased tubular sodium/glucose cotransport for diabetes-induced glomerular hyperfiltration. To eliminate any potential confounding effect of the tubuloglomerular feedback (TGF) mechanism, we used adenosine A1-receptor deficient (A1AR(-/-)) mice known to lack a functional TGF mechanism and compared the results to corresponding wild-type animals (A1AR(+/+)). METHODS: Diabetes was induced by an intravenous bolus injection of alloxan. Glomerular filtration rate (GFR) was determined in conscious mice by a single bolus injection of inulin. The sodium/glucose cotransporters were inhibited by phlorizin 30 min prior to GFR measurements. RESULTS: Normoglycaemic animals had a similar GFR independent of genotype (A1AR(+/+) 233 ± 11 vs. A1AR(-/-) 241 ± 25 µL min(-1)), and induction of diabetes resulted in glomerular hyperfiltration in both groups (A1AR(+/+) 380 ± 25 vs. A1AR(-/-) 336 ± 35 µL min(-1); both P < 0.05). Phlorizin had no effect on GFR in normoglycaemic mice, whereas it reduced GFR in both genotypes during diabetes (A1AR(+/+) 365 ± 18 to 295 ± 19, A1AR(-/-) 354 ± 38 to 199 ± 15 µL min(-1); both P < 0.05). Notably, the reduction was more pronounced in the A1AR(-/-) (P < 0.05). CONCLUSION: This study demonstrates that increased tubular sodium/glucose reabsorption is important for diabetes-induced hyperfiltration, and that the TGF mechanism is not involved in these alterations, but rather functions to reduce any deviations from a new set-point.


Assuntos
Diabetes Mellitus Experimental/fisiopatologia , Taxa de Filtração Glomerular/fisiologia , Glucose/metabolismo , Glomérulos Renais/fisiopatologia , Sódio/metabolismo , Animais , Estado de Consciência , Diabetes Mellitus Experimental/metabolismo , Hiperglicemia/fisiopatologia , Glomérulos Renais/metabolismo , Masculino , Camundongos , Camundongos Knockout , Receptor A1 de Adenosina/deficiência
11.
Am J Physiol Heart Circ Physiol ; 305(11): H1668-79, 2013 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-24043252

RESUMO

We previously demonstrated that A2A, but not A2B, adenosine receptors (ARs) mediate coronary reactive hyperemia (RH), possibly by producing H2O2 and, subsequently, opening ATP-dependent K(+) (KATP) channels in coronary smooth muscle cells. In this study, A1 AR knockout (KO), A3 AR KO, and A1 and A3 AR double-KO (A1/A3 DKO) mice were used to investigate the roles and mechanisms of A1 and A3 ARs in modulation of coronary RH. Coronary flow of isolated hearts was measured using the Langendorff system. A1 KO and A1/A3 DKO, but not A3 KO, mice showed a higher flow debt repayment [~30% more than wild-type (WT) mice, P < 0.05] following a 15-s occlusion. SCH-58261 (a selective A2A AR antagonist, 1 µM) eliminated the augmented RH, suggesting the involvement of enhanced A2A AR-mediated signaling in A1 KO mice. In isolated coronary arteries, immunohistochemistry showed an upregulation of A2A AR (1.6 ± 0.2 times that of WT mice, P < 0.05) and a higher magnitude of adenosine-induced H2O2 production in A1 KO mice (1.8 ± 0.3 times that of WT mice, P < 0.05), which was blocked by SCH-58261. Catalase (2,500 U/ml) and glibenclamide (a KATP channel blocker, 5 µM), but not N(G)-nitro-l-arginine methyl ester, also abolished the enhanced RH in A1 KO mice. Our data suggest that A1, but not A3, AR counteracts the A2A AR-mediated CF increase and that deletion of A1 AR results in upregulation of A2A AR and/or removal of the negative modulatory effect of A1 AR, thus leading to an enhanced A2A AR-mediated H2O2 production, KATP channel opening, and coronary vasodilation during RH. This is the first report implying that A1 AR has a role in coronary RH.


Assuntos
Circulação Coronária , Vasos Coronários/metabolismo , Peróxido de Hidrogênio/metabolismo , Hiperemia/metabolismo , Canais KATP/metabolismo , Receptor A1 de Adenosina/metabolismo , Receptor A2A de Adenosina/metabolismo , Vasodilatação , Antagonistas do Receptor A2 de Adenosina/farmacologia , Animais , Antioxidantes/farmacologia , Circulação Coronária/efeitos dos fármacos , Vasos Coronários/efeitos dos fármacos , Vasos Coronários/fisiopatologia , Feminino , Hiperemia/genética , Hiperemia/fisiopatologia , Canais KATP/antagonistas & inibidores , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Perfusão , Bloqueadores dos Canais de Potássio/farmacologia , Receptor A1 de Adenosina/deficiência , Receptor A1 de Adenosina/genética , Receptor A2A de Adenosina/efeitos dos fármacos , Receptor A3 de Adenosina/genética , Receptor A3 de Adenosina/metabolismo , Transdução de Sinais/efeitos dos fármacos , Fatores de Tempo , Vasodilatação/efeitos dos fármacos
12.
Toxicology ; 309: 100-6, 2013 Jul 05.
Artigo em Inglês | MEDLINE | ID: mdl-23692951

RESUMO

Previous studies have indicated a critical role of adenosine and its receptors in the pathogenesis of liver diseases. The aim of this study was to determine the contribution of A1 adenosine receptor (A1AR) to acute ethanol-induced hepatotoxicity. Wild-type (WT) and A1AR(-/-) mice were intragastrically administered with ethanol (5 g/kg), and hepatic injury was evaluated 6h thereafter. Mice lacking A1AR were more susceptible to ethanol-induced liver damage than WT mice, as evidenced by higher serum transaminase levels and increased extent of histopathological changes. Ethanol induced triglycerides accumulation in the serum and liver, and this accumulation was augmented in A1AR(-/-) mice. Analysis of gene expression in the liver revealed up-regulated mRNA levels of genes related to lipogenesis (including: FAS, SCD1, ACC1, DGAT2, and PPARγ) in A1AR(-/-) mice after ethanol treatment. In addition, lack of A1AR aggravated lipid peroxidation and superoxide dismutase depletion caused by acute ethanol exposure. A subsequent study revealed that, pretreatment with A1AR antagonist DPCPX increases the sensitivity of mice to ethanol-induced liver injury. In conclusion, these results indicated that endogenous A1AR activation protects mice against acute ethanol-induced liver injury by reducing oxidative stress and decreasing lipid accumulation.


Assuntos
Etanol/toxicidade , Fígado/metabolismo , Receptor A1 de Adenosina/fisiologia , Animais , Fígado/efeitos dos fármacos , Fígado/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Estresse Oxidativo/efeitos dos fármacos , Estresse Oxidativo/fisiologia , Receptor A1 de Adenosina/deficiência , Fatores de Tempo
13.
Eur J Pharmacol ; 698(1-3): 213-9, 2013 Jan 05.
Artigo em Inglês | MEDLINE | ID: mdl-23142373

RESUMO

The present study explored a link between spinal 5-HT(7) and adenosine A(1) receptors in antinociception by systemic amitriptyline in normal and adenosine A(1) receptor knock-out mice using the 2% formalin test. In normal mice, antinociception by systemic amitriptyline 3mg/kg was blocked by intrathecal administration of the selective adenosine A(1) receptor antagonist 8-cyclopentyl-1,3-dipropylxanthine (DPCPX) 10 nmol. Blockade was also seen in adenosine A(1) receptor +/+ mice, but not in -/- mice lacking these receptors. In both normal and adenosine A(1) receptor +/+ mice, the selective 5-HT(7) receptor antagonist (2R)-1-[(3-hydroxyphenyl)sulfonyl]-2-[2-(4-methyl-1-piperidinyl)ethyl]pyrrolidine hydrochloride (SB269970) 3 µg blocked antinociception by systemic amitriptyline, but it did not prevent antinociception in adenosine A(1) receptor -/- mice. In normal mice, flinching was unaltered when the selective 5-HT(7) receptor agonist (2S)-(+)-5-(1,3,5-trimethylpyrazol-4-yl)-2-(dimethylamino)tetralin (AS-19) 20 µg was administered alone, but increased when co-administered intrathecally with DPCPX 10 nmol or SB269970 3 µg. Intrathecal AS-19 decreased flinching in adenosine A(1) receptor +/+ mice compared to -/- mice. Systemic amitriptyline appears to reduce nociception by activating spinal adenosine A(1) receptors secondarily to 5-HT(7) receptors. Spinal actions constitute only one aspect of antinociception by amitriptyline, as intraplantar DPCPX 10 nmol blocked antinociception by systemic amitriptyline in normal and adenosine A(1) receptor +/+, but not -/- mice. Adenosine A(1) receptor interactions are worthy of attention, as chronic oral caffeine (0.1, 0.3g/L, doses considered relevant to human intake levels) blocked antinociception by systemic amitriptyline in normal mice. In conclusion, adenosine A(1) receptors contribute to antinociception by systemic amitriptyline in both spinal and peripheral compartments.


Assuntos
Amitriptilina/administração & dosagem , Amitriptilina/farmacologia , Sistema Nervoso Periférico/metabolismo , Receptor A1 de Adenosina/metabolismo , Receptores de Serotonina/metabolismo , Medula Espinal/metabolismo , Antagonistas do Receptor A1 de Adenosina/farmacologia , Amitriptilina/antagonistas & inibidores , Analgésicos/administração & dosagem , Analgésicos/antagonistas & inibidores , Analgésicos/farmacologia , Animais , Cafeína/farmacologia , Feminino , Técnicas de Silenciamento de Genes , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Sistema Nervoso Periférico/efeitos dos fármacos , Fenóis/farmacologia , Pirazóis/farmacologia , Receptor A1 de Adenosina/deficiência , Receptor A1 de Adenosina/genética , Antagonistas da Serotonina/farmacologia , Agonistas do Receptor de Serotonina/farmacologia , Medula Espinal/efeitos dos fármacos , Sulfonamidas/farmacologia , Tetra-Hidronaftalenos/farmacologia , Fatores de Tempo , Xantinas/farmacologia
14.
Eur J Pharmacol ; 674(2-3): 248-54, 2012 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-22085758

RESUMO

The present study examined effects of caffeine on antinociception by acetaminophen in the formalin test in mice. It demonstrates that caffeine 10mg/kg inhibits antinociception produced by acetaminophen 300 mg/kg i.p. against phase 2 flinches. Chronic administration of caffeine in the drinking water (0.1, 0.3g/l) for 8 days also inhibits the action of acetaminophen. The selective adenosine A(1) receptor antagonist DPCPX 1mg/kg i.p. mimics the action of caffeine, but the selective adenosine A(2A) receptor antagonist SCH58261 3mg/kg i.p. does not. While acetaminophen produced the same effect in mice that were +/+, +/- and -/- for adenosine A(1) receptors, inhibition of antinociception by caffeine was seen only in +/+ and +/- mice. A higher dose of caffeine, 40 mg/kg, produced an intrinsic antinociception against formalin-evoked flinches, an effect also seen when caffeine was administered intrathecally. SCH58261 30 nmol, but not DPCPX 10 nmol, also produced antinociception when administered intrathecally indicating involvement of adenosine A(2A) receptors in spinal antinociception. Caffeine reversal of acetaminophen results from actions in the spinal cord, as intrathecal DPCPX 10 nmol inhibited antinociception by systemic acetaminophen; this was also observed in +/+ but not in -/- adenosine A(1) receptor mice. We propose that spinal adenosine A(1) receptors contribute to the action of acetaminophen secondarily to involvement of descending serotonin pathways and release of adenosine within the spinal cord. Inhibition of acetaminophen antinociception by doses of caffeine relevant to dietary human intake levels suggests a more detailed consideration of acetaminophen-caffeine interactions in humans is warranted.


Assuntos
Acetaminofen/antagonistas & inibidores , Antagonistas do Receptor A1 de Adenosina/farmacologia , Analgésicos/antagonistas & inibidores , Cafeína/farmacologia , Medição da Dor/efeitos dos fármacos , Receptor A1 de Adenosina/metabolismo , Medula Espinal/metabolismo , Acetaminofen/administração & dosagem , Antagonistas do Receptor A2 de Adenosina/farmacologia , Analgésicos/administração & dosagem , Animais , Cafeína/administração & dosagem , Deleção de Genes , Masculino , Camundongos , Receptor A1 de Adenosina/deficiência , Receptor A1 de Adenosina/genética , Receptor A2A de Adenosina/metabolismo , Medula Espinal/efeitos dos fármacos , Fatores de Tempo
15.
J Neurosci ; 31(44): 16012-25, 2011 Nov 02.
Artigo em Inglês | MEDLINE | ID: mdl-22049443

RESUMO

Thalamocortical (TC) projections provide the major pathway for ascending sensory information to the mammalian neocortex. Arrays of these projections form synaptic inputs on thalamorecipient neurons, thus contributing to the formation of receptive fields (RFs) in sensory cortices. Experience-dependent plasticity of RFs persists throughout an organism's life span but in adults requires activation of cholinergic inputs to the cortex. In contrast, synaptic plasticity at TC projections is limited to the early postnatal period. This disconnect led to the widespread belief that TC synapses are the principal site of RF plasticity only in neonatal sensory cortices, but that they lose this plasticity upon maturation. Here, we tested an alternative hypothesis that mature TC projections do not lose synaptic plasticity but rather acquire gating mechanisms that prevent the induction of synaptic plasticity. Using whole-cell recordings and direct measures of postsynaptic and presynaptic activity (two-photon glutamate uncaging and two-photon imaging of the FM 1-43 assay, respectively) at individual synapses in acute mouse brain slices that contain the auditory thalamus and cortex, we determined that long-term depression (LTD) persists at mature TC synapses but is gated presynaptically. Cholinergic activation releases presynaptic gating through M(1) muscarinic receptors that downregulate adenosine inhibition of neurotransmitter release acting through A(1) adenosine receptors. Once presynaptic gating is released, mature TC synapses can express LTD postsynaptically through group I metabotropic glutamate receptors. These results indicate that synaptic plasticity at TC synapses is preserved throughout the life span and, therefore, may be a cellular substrate of RF plasticity in both neonate and mature animals.


Assuntos
Córtex Cerebral/citologia , Depressão Sináptica de Longo Prazo/fisiologia , Terminações Pré-Sinápticas/fisiologia , Sinapses/fisiologia , Transmissão Sináptica/fisiologia , Tálamo/citologia , Animais , Estimulação Elétrica , Potenciais Pós-Sinápticos Excitadores/efeitos dos fármacos , Potenciais Pós-Sinápticos Excitadores/genética , Glutamatos/farmacologia , Técnicas In Vitro , Indóis/farmacologia , Ativação do Canal Iônico/efeitos dos fármacos , Ativação do Canal Iônico/genética , Depressão Sináptica de Longo Prazo/efeitos dos fármacos , Depressão Sináptica de Longo Prazo/genética , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Técnicas de Patch-Clamp , Terminações Pré-Sinápticas/efeitos dos fármacos , Compostos de Piridínio/metabolismo , Compostos de Amônio Quaternário/metabolismo , Receptor A1 de Adenosina/deficiência , Transmissão Sináptica/genética
16.
Am J Physiol Heart Circ Physiol ; 301(3): H1127-34, 2011 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-21685268

RESUMO

The presence of sex differences in myocardial ß-adrenergic responsiveness is controversial, and limited studies have addressed the mechanism underlying these differences. Studies were performed using isolated perfused hearts from male, intact female and ovariectomized female mice to investigate sex differences and the effects of ovarian hormone withdrawal on ß-adrenergic receptor function. Female hearts exhibited blunted contractile responses to the ß-adrenergic receptor agonist isoproterenol (ISO) compared with males but not ovariectomized females. There were no sex differences in ß(1)-adrenergic receptor gene or protein expression. To investigate the role of adenylyl cyclase, phosphodiesterase, and the cAMP-signaling cascade in generating sex differences in the ß-adrenergic contractile response, dose-response studies were performed in isolated perfused male and female hearts using forskolin, 3-isobutyl-1-methylxanthine (IBMX), and 8-(4-chlorophenylthio)adenosine 3',5'-cyclic monophosphate (CPT-cAMP). Males showed a modestly enhanced contractile response to forskolin at 300 nM and 5 µM compared with females, but there were no sex differences in the response to IBMX or CPT-cAMP. The role of the A(1) adenosine receptor (A(1)AR) in antagonizing the ß-adrenergic contractile response was investigated using both the A(1)AR agonist 2-chloro-N(6)-cyclopentyl-adenosine and A(1)AR knockout (KO) mice. Intact females showed an enhanced A(1)AR anti-adrenergic effect compared with males and ovariectomized females. The ß-adrenergic contractile response was potentiated in both male and female A(1)ARKO hearts, with sex differences no longer present above 1 nM ISO. The ß-adrenergic contractile response is greater in male hearts than females, and minor differences in the action of adenylyl cyclase or the A(1)AR may contribute to these sex differences.


Assuntos
Agonistas Adrenérgicos beta/farmacologia , Isoproterenol/farmacologia , Contração Miocárdica/efeitos dos fármacos , Miocárdio/metabolismo , Ovariectomia , Receptores Adrenérgicos beta/efeitos dos fármacos , Agonistas do Receptor A1 de Adenosina/farmacologia , Adenilil Ciclases/metabolismo , Análise de Variância , Animais , Peso Corporal , AMP Cíclico/metabolismo , Relação Dose-Resposta a Droga , Ativadores de Enzimas/farmacologia , Feminino , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Perfusão , Inibidores de Fosfodiesterase/farmacologia , Diester Fosfórico Hidrolases/metabolismo , RNA Mensageiro/metabolismo , Receptor A1 de Adenosina/deficiência , Receptor A1 de Adenosina/efeitos dos fármacos , Receptor A1 de Adenosina/genética , Receptores Adrenérgicos beta/genética , Receptores Adrenérgicos beta/metabolismo , Fatores Sexuais , Transdução de Sinais/efeitos dos fármacos
17.
J Pharmacol Exp Ther ; 336(1): 77-86, 2011 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-20876230

RESUMO

Ghrelin, a potent orexigenic hormone released from the stomach, is important in regulating energy metabolism. Abnormal ghrelin levels are associated with eating disorders and metabolic diseases. However, factors involved in the regulation of ghrelin release remain unclear. Here, we examined the involvement of adenosine signaling in the control of ghrelin release from the perfused mouse stomach. Adenosine stimulated ghrelin release concentration-dependently, and the A(2A) receptor-selective antagonists 4-(2-[7-amino-2-(2-furyl)[1,2,4]triazolo[2,3-a][1,3,5]triazin-5-ylamino]ethyl)phenol (ZM 241385) and 2-(2-furanyl)-7-(2-phenylethyl)-7H-pyrazolo[4,3-e][1,2,4]triazolo[1,5-c]pyrimidin-5-amine (SCH 58261) abolished the increased release. The A(2A) receptor-selective agonist 2-p-(2-carboxyethyl)phenethylamino-5-N-ethylcarboxamidoadenosine hydrochloride (CGS 21680) augmented ghrelin release concentration-dependently, whereas the A(1) receptor-selective agonist 2-chloro-N(6)-cyclopentyladenosine inhibited ghrelin release. In A(2A) receptor knockout mice, adenosine inhibited ghrelin release, and the A(1) receptor-selective antagonist 8-cyclopentyl-1,3-dipropylxanthine blocked this inhibition. The adenosine deaminase inhibitor erythro-9-(2-hydroxy-3-nonyl)adenine hydrochloride increased ghrelin release in wild-type and A(1) receptor knockout mice but not in A(2A) receptor knockout mice. Colocalization of ghrelin immunoreactivity with A(1) and A(2A) receptor immunoreactivities in the gastric nerve fibers were observed. Colocalization was also detected for ghrelin and A(1) receptor immunoreactivities in the gastric mucosa. Blockade of neural activities with tetrodotoxin abolished the stimulatory effect of adenosine on ghrelin release. In conclusion, adenosine exerts predominantly a tonic A(2A) receptor-mediated stimulatory action on gastric ghrelin release, whereas an A(1) receptor-mediated inhibitory action is also apparent when the tonic excitatory effect was removed.


Assuntos
Adenosina/fisiologia , Mucosa Gástrica/metabolismo , Grelina/metabolismo , Receptor A1 de Adenosina/fisiologia , Receptor A2A de Adenosina/fisiologia , Transdução de Sinais/fisiologia , Animais , Relação Dose-Resposta a Droga , Mucosa Gástrica/efeitos dos fármacos , Masculino , Camundongos , Camundongos Congênicos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Perfusão , Receptor A1 de Adenosina/deficiência , Receptor A2A de Adenosina/deficiência , Transdução de Sinais/efeitos dos fármacos
18.
Brain Res ; 1367: 310-8, 2011 Jan 07.
Artigo em Inglês | MEDLINE | ID: mdl-20828543

RESUMO

Adenosine A(2A) receptor antagonism provides a promising approach to developing nondopaminergic therapy for Parkinson's disease (PD). Clinical trials of A(2A) antagonists have targeted PD patients with L-3,4-dihydroxyphenylalanine (L-DOPA)-induced dyskinesia (LID) in an effort to improve parkinsonian symptoms. The role of adenosine in the development of LID is little known, especially regarding its actions via A1 receptors. We aimed to examine the effects of genetic deletion and pharmacological blockade of A1 and/or A(2A) receptors on the development of LID, on the induction of molecular markers of LID including striatal preprodynorphin and preproenkephalin (PPE), and on the integrity of dopaminergic nigrostriatal neurons in hemiparkinsonian mice. Following a unilateral 6-hydroxydopamine lesion A1, A(2A) and double A1-A(2A) knockout (KO) and wild-type littermate mice, and mice pretreated with caffeine (an antagonist of both A1 and A(2A) receptors) or saline were treated daily for 18-21 days with a low dose of L-DOPA. Total abnormal involuntary movements (AIMs, a measure of LID) were significantly attenuated (p<0.05) in A1 and A(2A) KOs, but not in A1-A(2A) KOs and caffeine-pretreated mice. An elevation of PPE mRNA ipsilateral to the lesion in WT mice was reduced in all KO mice. In addition, neuronal integrity assessed by striatal dopamine content was similar in all KOs and caffeine-pretreated mice following 6-hydroxydopamine lesioning. Our findings raise the possibility that A1 or A(2A) receptors blockade might also confer a disease-modifying benefit of reduced risk of disabling LID, whereas the effect of their combined inactivation is less clear.


Assuntos
Antiparkinsonianos/efeitos adversos , Discinesia Induzida por Medicamentos/genética , Discinesia Induzida por Medicamentos/prevenção & controle , Levodopa/efeitos adversos , Receptor A1 de Adenosina/deficiência , Receptor A2A de Adenosina/deficiência , Adrenérgicos/toxicidade , Animais , Cafeína/administração & dosagem , Corpo Estriado/efeitos dos fármacos , Modelos Animais de Doenças , Dinorfinas/genética , Dinorfinas/metabolismo , Discinesia Induzida por Medicamentos/etiologia , Encefalinas/genética , Encefalinas/metabolismo , Regulação da Expressão Gênica/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Oxidopamina/toxicidade , Doença de Parkinson/tratamento farmacológico , Doença de Parkinson/etiologia , Doença de Parkinson/genética , Precursores de Proteínas/genética , Precursores de Proteínas/metabolismo , Antagonistas de Receptores Purinérgicos P1/administração & dosagem , RNA Mensageiro/metabolismo , Estatísticas não Paramétricas , Fatores de Tempo
19.
Am J Physiol Regul Integr Comp Physiol ; 299(5): R1263-8, 2010 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-20739607

RESUMO

A high protein intake is associated with increased glomerular filtration rate (GFR), which has been suggested to be mediated by reduced signaling of the tubuloglomerular feedback (TGF) mechanism. Nitric oxide (NO) has been shown to contribute to high protein-induced glomerular hyperfiltration, but the specific NO synthase (NOS) isoform responsible is not clear. In this study, a model for high-protein-induced hyperfiltration in conscious mice was developed. Using this model, we investigated the role of TGF using adenosine A(1)-receptor knockout mice lacking the TGF mechanism. Furthermore, the role of the different NOS isoforms was studied using neuronal-, inducible-, and endothelial-NOS knockout mice, and furthermore, wild-type mice acutely administered with the unspecific NOS inhibitor N(ω)-nitro-l-arginine methyl ester (100 mg/kg). GFR was measured consecutively in mice given a low-protein diet (8% casein) for 10 days, followed by a high-protein diet (50% casein) for 10 days. All mice developed high protein-induced hyperfiltration to a similar degree. These results demonstrate that high protein-induced glomerular hyperfiltration is independent of the TGF mechanism and NOS isoforms.


Assuntos
Taxa de Filtração Glomerular , Nefropatias/fisiopatologia , Rim/fisiopatologia , Óxido Nítrico Sintase/metabolismo , Animais , Proteínas Alimentares , Modelos Animais de Doenças , Inibidores Enzimáticos/farmacologia , Retroalimentação Fisiológica , Feminino , Taxa de Filtração Glomerular/efeitos dos fármacos , Rim/efeitos dos fármacos , Rim/enzimologia , Nefropatias/enzimologia , Nefropatias/etiologia , Glomérulos Renais/enzimologia , Glomérulos Renais/fisiopatologia , Túbulos Renais/enzimologia , Túbulos Renais/fisiopatologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , NG-Nitroarginina Metil Éster/farmacologia , Óxido Nítrico Sintase/antagonistas & inibidores , Óxido Nítrico Sintase/deficiência , Óxido Nítrico Sintase/genética , Óxido Nítrico Sintase Tipo I/metabolismo , Óxido Nítrico Sintase Tipo II/metabolismo , Óxido Nítrico Sintase Tipo III/metabolismo , Receptor A1 de Adenosina/deficiência , Receptor A1 de Adenosina/genética , Fatores de Tempo
20.
J Neuroimmunol ; 224(1-2): 85-92, 2010 Jul 27.
Artigo em Inglês | MEDLINE | ID: mdl-20570369

RESUMO

We characterized the role of adenosine receptor (AR) subtypes in the modulation of glutamatergic neurotransmission by the chemokine fractalkine (CX3CL1) in mouse hippocampal CA1 neurons. CX(3)CL1 causes a reversible depression of excitatory postsynaptic current (EPSC), which is abolished by the A(3)R antagonist MRS1523, but not by A(1)R (DPCPX) or A(2A)R (SCH58261) antagonists. Consistently, CX3CL1-induced EPSC depression is absent in slices from A(3)R(-/-) but not A(1)R(-/-) or A(2A)R(-/-) mice. Further, A(3)R stimulation causes similar EPSC depression. In cultured neurons, CX3CL1-induced depression of AMPA current shows A(1)R-A(3)R pharmacology. We conclude that glutamatergic depression induced by released adenosine requires the stimulation of different ARs.


Assuntos
Região CA1 Hipocampal/imunologia , Região CA1 Hipocampal/metabolismo , Quimiocina CX3CL1/fisiologia , Potenciais Pós-Sinápticos Excitadores/imunologia , Inibição Neural/imunologia , Receptores Purinérgicos P1/fisiologia , Transmissão Sináptica/imunologia , Antagonistas do Receptor A1 de Adenosina , Antagonistas do Receptor A2 de Adenosina , Antagonistas do Receptor A3 de Adenosina , Animais , Região CA1 Hipocampal/ultraestrutura , Células Cultivadas , Potenciais Pós-Sinápticos Excitadores/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Inibição Neural/genética , Técnicas de Cultura de Órgãos , Técnicas de Patch-Clamp , Terminações Pré-Sinápticas/imunologia , Terminações Pré-Sinápticas/metabolismo , Receptor A1 de Adenosina/deficiência , Receptor A1 de Adenosina/fisiologia , Receptor A3 de Adenosina/deficiência , Receptor A3 de Adenosina/fisiologia , Receptores A2 de Adenosina/deficiência , Receptores A2 de Adenosina/fisiologia , Receptores Purinérgicos P1/deficiência , Receptores Purinérgicos P1/genética , Transmissão Sináptica/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA