Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 110
Filtrar
1.
Handb Exp Pharmacol ; 253: 359-381, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-28646346

RESUMO

The classic endogenous somnogen adenosine promotes sleep via A1 and A2A receptors. In this chapter, we present an overview of the current knowledge regarding the regulation of adenosine levels, adenosine receptors, and available pharmacologic and genetic tools to manipulate the adenosine system. This is followed by a summary of current knowledge of the role of adenosine and its receptors in the regulation of sleep and wakefulness. Despite strong data implicating numerous brain areas, including the basal forebrain, the tuberomammillary nucleus, the lateral hypothalamus, and the nucleus accumbens, in the adenosinergic control of sleep, the complete neural circuitry in the brain involved in the sleep-promoting effects of adenosine remains unclear. Moreover, the popular demand for natural sleep aids has led to a search for natural compounds that can promote sleep via adenosine receptor activation. Finally, we discuss the effects of caffeine in man and the possible use of more selective adenosine receptor drugs for the treatment of sleep disorders.


Assuntos
Adenosina , Sono , Adenosina/metabolismo , Encéfalo/fisiologia , Vigília/fisiologia
2.
Purinergic Signal ; 2022 Oct 21.
Artigo em Inglês | MEDLINE | ID: mdl-36269528
3.
Purinergic Signal ; 12(1): 89-101, 2016 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-26608888

RESUMO

Neonatal brain hypoxic ischemia (HI) often results in long-term motor and cognitive impairments. Post-ischemic inflammation greatly effects outcome and adenosine receptor signaling modulates both HI and immune cell function. Here, we investigated the influence of adenosine A1 receptor deficiency (A1R(-/-)) on key immune cell populations in a neonatal brain HI model. Ten-day-old mice were subjected to HI. Functional outcome was assessed by open locomotion and beam walking test and infarction size evaluated. Flow cytometry was performed on brain-infiltrating cells, and semi-automated analysis of flow cytometric data was applied. A1R(-/-) mice displayed larger infarctions (+33%, p < 0.05) and performed worse in beam walking tests (44% more mistakes, p < 0.05) than wild-type (WT) mice. Myeloid cell activation after injury was enhanced in A1R(-/-) versus WT brains. Activated B lymphocytes expressing IL-10 infiltrated the brain after HI in WT, but were less activated and did not increase in relative frequency in A1R(-/-). Also, A1R(-/-) B lymphocytes expressed less IL-10 than their WT counterparts, the A1R antagonist DPCPX decreased IL-10 expression whereas the A1R agonist CPA increased it. CD4(+) T lymphocytes including FoxP3(+) T regulatory cells, were unaffected by genotype, whereas CD8(+) T lymphocyte responses were smaller in A1R(-/-) mice. Using PCA to characterize the immune profile, we could discriminate the A1R(-/-) and WT genotypes as well as sham operated from HI-subjected animals. We conclude that A1R signaling modulates IL-10 expression by immune cells, influences the activation of these cells in vivo, and affects outcome after HI.


Assuntos
Isquemia Encefálica/imunologia , Encéfalo/imunologia , Hipóxia Encefálica/imunologia , Receptor A1 de Adenosina/imunologia , Agonistas do Receptor A1 de Adenosina/farmacologia , Antagonistas do Receptor A1 de Adenosina/farmacologia , Animais , Animais Recém-Nascidos , Linfócitos T CD4-Positivos/efeitos dos fármacos , Linfócitos T CD4-Positivos/metabolismo , Infarto Cerebral/patologia , Feminino , Hipóxia Encefálica/congênito , Interleucina-10/biossíntese , Interleucina-10/genética , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Atividade Motora , Células Mieloides/efeitos dos fármacos , Células Mieloides/imunologia , Células Mieloides/metabolismo , Equilíbrio Postural , Gravidez , Receptor A1 de Adenosina/efeitos dos fármacos
4.
Diabetologia ; 58(7): 1610-20, 2015 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-25835725

RESUMO

AIMS/HYPOTHESIS: Adenosine is an important regulator of metabolism; however, the role of the A1 receptor during ageing and obesity is unclear. The aim of this study was to investigate the effects of A1 signalling in modulating metabolic function during ageing. METHODS: Age-matched young and aged A 1 (also known as Adora1)-knockout (A1(-/-)) and wild-type (A1(+/+)) mice were used. Metabolic regulation was evaluated by body composition, and glucose and insulin tolerance tests. Isolated islets and islet arterioles were used to detect islet endocrine and vascular function. Oxidative stress and inflammation status were measured in metabolic organs and systemically. RESULTS: Advanced age was associated with both reduced glucose clearance and insulin sensitivity, as well as increased visceral adipose tissue (VAT) in A1(+/+) compared with A1(-/-) mice. Islet morphology and insulin content were similar between genotypes, but relative changes in in vitro insulin release following glucose stimulation were reduced in aged A1(+/+) compared with A1(-/-) mice. Islet arteriolar responses to angiotensin II were stronger in aged A1(+/+) mice, this being associated with increased NADPH oxidase activity. Ageing resulted in multiple changes in A1(+/+) compared with A1(-/-) mice, including enhanced NADPH oxidase-derived O2(-) formation and NADPH oxidase isoform 2 (Nox2) protein expression in pancreas and VAT; elevated levels of circulating insulin, leptin and proinflammatory cytokines (TNF-α, IL-1ß, IL-6 and IL-12); and accumulation of CD4(+) T cells in VAT. This was associated with impaired insulin signalling in VAT from aged A1(+/+) mice. CONCLUSIONS/INTERPRETATION: These studies emphasise that A1 receptors regulate metabolism and islet endocrine and vascular functions during ageing, including via the modulation of oxidative stress and inflammatory responses, among other things.


Assuntos
Inflamação/genética , Estresse Oxidativo/genética , Receptor A1 de Adenosina/genética , Tecido Adiposo/metabolismo , Envelhecimento/metabolismo , Angiotensina II/farmacologia , Animais , Composição Corporal/genética , Linfócitos T CD4-Positivos/metabolismo , Citocinas/metabolismo , Feminino , Intolerância à Glucose/genética , Insulina/metabolismo , Resistência à Insulina , Ilhotas Pancreáticas/irrigação sanguínea , Masculino , Glicoproteínas de Membrana/metabolismo , Metabolismo/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , NADPH Oxidase 2 , NADPH Oxidases/metabolismo , Fluxo Sanguíneo Regional/fisiologia , Transdução de Sinais/genética
5.
Pharmacol Rev ; 63(1): 1-34, 2011 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-21303899

RESUMO

In the 10 years since our previous International Union of Basic and Clinical Pharmacology report on the nomenclature and classification of adenosine receptors, no developments have led to major changes in the recommendations. However, there have been so many other developments that an update is needed. The fact that the structure of one of the adenosine receptors has recently been solved has already led to new ways of in silico screening of ligands. The evidence that adenosine receptors can form homo- and heteromultimers has accumulated, but the functional significance of such complexes remains unclear. The availability of mice with genetic modification of all the adenosine receptors has led to a clarification of the functional roles of adenosine, and to excellent means to study the specificity of drugs. There are also interesting associations between disease and structural variants in one or more of the adenosine receptors. Several new selective agonists and antagonists have become available. They provide improved possibilities for receptor classification. There are also developments hinting at the usefulness of allosteric modulators. Many drugs targeting adenosine receptors are in clinical trials, but the established therapeutic use is still very limited.


Assuntos
Receptores Purinérgicos P1/classificação , Receptores Purinérgicos P1/fisiologia , Animais , Animais Geneticamente Modificados , Humanos , Agências Internacionais , Terapia de Alvo Molecular , Agonistas do Receptor Purinérgico P1/farmacologia , Agonistas do Receptor Purinérgico P1/uso terapêutico , Antagonistas de Receptores Purinérgicos P1/farmacologia , Antagonistas de Receptores Purinérgicos P1/uso terapêutico , Receptores Purinérgicos P1/química , Receptores Purinérgicos P1/genética , Terminologia como Assunto
6.
J Neurosci ; 31(45): 16327-35, 2011 Nov 09.
Artigo em Inglês | MEDLINE | ID: mdl-22072684

RESUMO

The chemokine CX3CL1 and its receptor CX3CR1 are constitutively expressed in the nervous system. In this study, we used in vivo murine models of permanent middle cerebral artery occlusion (pMCAO) to investigate the protective potential of CX3CL1. We report that exogenous CX3CL1 reduced ischemia-induced cerebral infarct size, neurological deficits, and caspase-3 activation. CX3CL1-induced neuroprotective effects were long lasting, being observed up to 50 d after pMCAO in rats. The neuroprotective action of CX3CL1 in different models of brain injuries is mediated by its inhibitory activity on microglia and, in vitro, requires the activation of adenosine receptor 1 (A1R). We show that, in the presence of the A1R antagonist 1,3-dipropyl-8-cyclopentylxanthine and in A1R⁻/⁻ mice, the neuroprotective effect of CX3CL1 on pMCAO was abolished, indicating the critical importance of the adenosine system in CX3CL1 protection also in vivo. In apparent contrast with the above reported data but in agreement with previous findings, cx3cl1⁻/⁻ and cx3cr1(GFP/GFP) mice, respectively, deficient in CX3CL1 or CX3CR1, had less severe brain injury on pMCAO, and the administration of exogenous CX3CL1 increased brain damage in cx3cl1⁻/⁻ ischemic mice. We also report that CX3CL1 induced a different phagocytic activity in wild type and cx3cl1⁻/⁻ microglia in vitro during cotreatment with the medium conditioned by neurons damaged by oxygen-glucose deprivation. Together, these data suggest that acute administration of CX3CL1 reduces ischemic damage via an adenosine-dependent mechanism and that the absence of constitutive CX3CL1-CX3CR1 signaling changes the outcome of microglia-mediated effects during CX3CL1 administration to ischemic brain.


Assuntos
Quimiocina CX3CL1/metabolismo , Quimiocina CX3CL1/uso terapêutico , Infarto da Artéria Cerebral Média/metabolismo , Infarto da Artéria Cerebral Média/prevenção & controle , Antagonistas do Receptor A1 de Adenosina/uso terapêutico , Análise de Variância , Animais , Animais Geneticamente Modificados , Animais Recém-Nascidos , Infarto Encefálico/etiologia , Infarto Encefálico/prevenção & controle , Receptor 1 de Quimiocina CX3C , Células Cultivadas , Córtex Cerebral/citologia , Quimiocina CX3CL1/deficiência , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Ensaio de Imunoadsorção Enzimática/métodos , Glucose/deficiência , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Humanos , Hipóxia/prevenção & controle , Infarto da Artéria Cerebral Média/complicações , Imageamento por Ressonância Magnética , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Doenças do Sistema Nervoso/etiologia , Doenças do Sistema Nervoso/metabolismo , Doenças do Sistema Nervoso/terapia , Neurônios/efeitos dos fármacos , Fagocitose/efeitos dos fármacos , Ratos , Receptores de Quimiocinas/deficiência , Receptores Purinérgicos P1/deficiência , Xantinas/uso terapêutico
7.
J Neurosci ; 31(27): 10067-75, 2011 Jul 06.
Artigo em Inglês | MEDLINE | ID: mdl-21734299

RESUMO

Caffeine, the most widely used psychoactive compound, is an adenosine receptor antagonist. It promotes wakefulness by blocking adenosine A(2A) receptors (A(2A)Rs) in the brain, but the specific neurons on which caffeine acts to produce arousal have not been identified. Using selective gene deletion strategies based on the Cre/loxP technology in mice and focal RNA interference to silence the expression of A(2A)Rs in rats by local infection with adeno-associated virus carrying short-hairpin RNA, we report that the A(2A)Rs in the shell region of the nucleus accumbens (NAc) are responsible for the effect of caffeine on wakefulness. Caffeine-induced arousal was not affected in rats when A(2A)Rs were focally removed from the NAc core or other A(2A)R-positive areas of the basal ganglia. Our observations suggest that caffeine promotes arousal by activating pathways that traditionally have been associated with motivational and motor responses in the brain.


Assuntos
Nível de Alerta/efeitos dos fármacos , Cafeína/farmacologia , Estimulantes do Sistema Nervoso Central/farmacologia , Núcleo Accumbens/efeitos dos fármacos , Núcleo Accumbens/metabolismo , Receptor A2A de Adenosina/metabolismo , Análise de Variância , Animais , Gânglios da Base/efeitos dos fármacos , Gânglios da Base/metabolismo , Linhagem Celular Transformada , Colina O-Acetiltransferase/metabolismo , Relação Dose-Resposta a Droga , Eletroencefalografia/métodos , Eletromiografia/métodos , Proteínas de Fluorescência Verde/genética , Humanos , Locomoção/efeitos dos fármacos , Locomoção/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Mutagênese , Mutação/genética , Fosfopiruvato Hidratase/metabolismo , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/metabolismo , Ratos , Ratos Sprague-Dawley , Receptor A2A de Adenosina/deficiência , Receptor A2A de Adenosina/genética , Receptores de Dopamina D2/metabolismo , Transfecção/métodos
8.
Am J Physiol Endocrinol Metab ; 303(2): E180-90, 2012 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-22550063

RESUMO

Type 2 diabetes mellitus (T2DM) is characterized by the inability of the pancreatic ß-cells to secrete enough insulin to meet the demands of the body. Therefore, research of potential therapeutic approaches to treat T2DM has focused on increasing insulin output from ß-cells or improving systemic sensitivity to circulating insulin. In this study, we examined the role of the A(1) receptor in glucose homeostasis with the use of A(1) receptor knockout mice (A(1)R(-/-)). A(1)R(-/-) mice exhibited superior glucose tolerance compared with wild-type controls. However, glucose-stimulated insulin release, insulin sensitivity, weight gain, and food intake were comparable between the two genotypes. Following a glucose challenge, plasma glucagon levels in wild-type controls decreased, but this was not observed in A(1)R(-/-) mice. In addition, pancreas perfusion with oscillatory glucose levels of 10-min intervals produced a regular pattern of pulsatile insulin release with a 10-min cycling period in wild-type controls and 5 min in A(1)R(-/-) mice. When the mice were fed a high-fat diet (HFD), both genotypes exhibited impaired glucose tolerance and insulin resistance. Increased insulin release was observed in HFD-fed mice in both genotypes, but increased glucagon release was observed only in HFD-fed A(1)R(-/-) mice. In addition, the regular patterns of insulin release following oscillatory glucose perfusion were abolished in HFD-fed mice in both genotypes. In conclusion, A(1) receptors in the pancreas are involved in regulating the temporal patterns of insulin release, which could have implications in the development of glucose intolerance seen in T2DM.


Assuntos
Glicemia/metabolismo , Insulina/metabolismo , Receptor A1 de Adenosina/metabolismo , Animais , Dieta Hiperlipídica , Ingestão de Alimentos , Glucagon/sangue , Glucose/farmacologia , Teste de Tolerância a Glucose , Insulina/sangue , Resistência à Insulina , Secreção de Insulina , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Receptor A1 de Adenosina/genética , Aumento de Peso
9.
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
10.
Am J Physiol Regul Integr Comp Physiol ; 301(6): R1669-81, 2011 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-21975649

RESUMO

Adenosine mediates tubuloglomerular feedback responses via activation of A(1)-receptors on the renal afferent arteriole. Increased preglomerular reactivity, due to reduced nitric oxide (NO) production or increased levels of ANG II and reactive oxygen species (ROS), has been linked to hypertension. Using A(1)-receptor knockout (A(1)(-/-)) and wild-type (A(1)(+/+)) mice we investigated the hypothesis that A(1)-receptors modulate arteriolar and blood pressure responses during NO synthase (NOS) inhibition or ANG II treatment. Blood pressure and renal afferent arteriolar responses were measured in nontreated mice and in mice with prolonged N(ω)-nitro-L-arginine methyl ester hydrochloride (L-NAME) or ANG II treatment. The hypertensive responses to L-NAME and ANG II were clearly attenuated in A(1)(-/-) mice. Arteriolar contractions to L-NAME (10(-4) mol/l; 15 min) and cumulative ANG II application (10(-12) to 10(-6) mol/l) were lower in A(1)(-/-) mice. Simultaneous treatment with tempol (10(-4) mol/l; 15 min) attenuated arteriolar responses in A(1)(+/+) but not in A(1)(-/-) mice, suggesting differences in ROS formation. Chronic treatment with L-NAME or ANG II did not alter arteriolar responses in A(1)(-/-) mice, but enhanced maximal contractions in A(1)(+/+) mice. In addition, chronic treatments were associated with higher plasma levels of dimethylarginines (asymmetrical and symmetrical) and oxidative stress marker malondialdehyde in A(1)(+/+) mice, and gene expression analysis showed reduced upregulation of NOS-isoforms and greater upregulation of NADPH oxidases. In conclusion, adenosine A(1)-receptors enhance preglomerular responses during NO inhibition and ANG II treatment. Interruption of A(1)-receptor signaling blunts l-NAME and ANG II-induced hypertension and oxidative stress and is linked to reduced responsiveness of afferent arterioles.


Assuntos
Angiotensina II/farmacologia , Arteríolas/efeitos dos fármacos , Pressão Sanguínea/efeitos dos fármacos , Óxido Nítrico/antagonistas & inibidores , Receptor A1 de Adenosina/genética , Animais , Arginina/análogos & derivados , Arginina/sangue , Pressão Sanguínea/genética , Pressão Sanguínea/fisiologia , Regulação da Expressão Gênica/efeitos dos fármacos , Regulação da Expressão Gênica/fisiologia , Rim/irrigação sanguínea , Rim/metabolismo , Camundongos , NG-Nitroarginina Metil Éster/farmacologia , Receptor A1 de Adenosina/metabolismo
11.
FASEB J ; 24(7): 2325-33, 2010 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-20181934

RESUMO

Adenosine regulates a wide variety of physiological processes via interaction with one or more G-protein-coupled receptors (A(1)R, A(2A)R, A(2B)R, and A(3)R). Because A(1)R occupancy promotes fusion of human monocytes to form giant cells in vitro, we determined whether A(1)R occupancy similarly promotes osteoclast function and formation. Bone marrow cells (BMCs) were harvested from C57Bl/6 female mice or A(1)R-knockout mice and their wild-type (WT) littermates and differentiated into osteoclasts in the presence of colony stimulating factor-1 and receptor activator of NF-kappaB ligand in the presence or absence of the A(1)R antagonist 1,3-dipropyl-8-cyclopentyl xanthine (DPCPX). Osteoclast morphology was analyzed in tartrate-resistant acid phosphatase or F-actin-stained samples, and bone resorption was evaluated by toluidine blue staining of dentin. BMCs from A(1)R-knockout mice form fewer osteoclasts than BMCs from WT mice, and the A(1)R antagonist DPCPX inhibits osteoclast formation (IC(50)=1 nM), with altered morphology and reduced ability to resorb bone. A(1)R blockade increased ubiquitination and degradation of TRAF6 in RAW264.7 cells induced to differentiate into osteoclasts. These studies suggest a critical role for adenosine in bone homeostasis via interaction with adenosine A(1)R and further suggest that A(1)R may be a novel pharmacologic target to prevent the bone loss associated with inflammatory diseases and menopause.


Assuntos
Osteoclastos/citologia , Receptor A1 de Adenosina/fisiologia , Animais , Células da Medula Óssea/citologia , Reabsorção Óssea , Diferenciação Celular , Forma Celular , Feminino , Homeostase , Camundongos , Osteoclastos/fisiologia , Fator 6 Associado a Receptor de TNF/metabolismo
12.
Exp Cell Res ; 316(8): 1284-8, 2010 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-20153317

RESUMO

There are four adenosine receptors, A(1), A(2A), A(2B) and A(3), together forming a defined subgroup of G protein coupled receptors. They are well conserved and widely expressed. The endogenous agonist, adenosine, has a minimal concentration in body fluids (20-200 nM) that is sufficient to slightly activate the receptors where they are very highly expressed-as in the basal ganglia, on fat cells and in the kidney. Here adenosine can play a physiological role and here antagonists such as caffeine can have effects in healthy individuals. Adenosine levels rise in stress and distress (up to 30 microM in ischemia) and tend to minimize the risk for adverse outcomes by increasing energy supply and decreasing cellular work, by stimulating angiogenesis, mediating preconditioning and having multiple effects on immune competent cells. These pathophysiological roles of adenosine also offer some potential drug targets, but the fact that adenosine receptors are involved in so many processes does not simplify drug development.


Assuntos
Antagonistas de Receptores Purinérgicos P1 , Animais , Humanos , Receptores Purinérgicos P1/metabolismo
13.
Handb Exp Pharmacol ; (200): 1-9, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-20859791

RESUMO

As behooves something so deeply entrenched in culture, the historical origins of the use of methylxanthines are unknown and dressed in myth. This is true for coffee as well as tea, and for both it is interesting to note that their common use is really very recent. For coffee we know that its use became more widespread in the fifteenth and sixteenth centuries, and in Europe this occurred in the eighteenth and nineteenth centuries. The use of tea became more common during the Ming Dynasty in China and during the eighteenth century in Britain. Coffee was mostly an upper-class drink in Arabia, and remained a relative luxury in Europe until quite recently. The use of other methylxanthine-containing beverages, such as maté, is even less well known. It is interesting to note that before these drinks were commonly used on a daily basis they were used for medicinal purposes, indicating that their pharmacological actions had long been noted.


Assuntos
Cafeína/história , Cacau/história , Café/história , História do Século XV , História do Século XVI , História do Século XVII , História do Século XVIII , História do Século XIX , Humanos , Chá/história
14.
Proc Natl Acad Sci U S A ; 105(50): 19992-7, 2008 Dec 16.
Artigo em Inglês | MEDLINE | ID: mdl-19066225

RESUMO

Adenosine has been proposed to promote sleep through A(1) receptors (A(1)R's) and/or A(2A) receptors in the brain. We previously reported that A(2A) receptors mediate the sleep-promoting effect of prostaglandin D(2), an endogenous sleep-inducing substance, and that activation of these receptors induces sleep and blockade of them by caffeine results in wakefulness. On the other hand, A(1)R has been suggested to increase sleep by inhibition of the cholinergic region of the basal forebrain. However, the role and target sites of A(1)R in sleep-wake regulation remained controversial. In this study, immunohistochemistry revealed that A(1)R was expressed in histaminergic neurons of the rat tuberomammillary nucleus (TMN). In vivo microdialysis showed that the histamine release in the frontal cortex was decreased by microinjection into the TMN of N(6)-cyclopentyladenosine (CPA), an A(1)R agonist, adenosine or coformycin, an inhibitor of adenosine deaminase, which catabolizes adenosine to inosine. Bilateral injection of CPA into the rat TMN significantly increased the amount and the delta power density of non-rapid eye movement (non-REM; NREM) sleep but did not affect REM sleep. CPA-promoted sleep was observed in WT mice but not in KO mice for A(1)R or histamine H(1) receptor, indicating that the NREM sleep promoted by A(1)R-specific agonist depended on the histaminergic system. Furthermore, the bilateral injection of adenosine or coformycin into the rat TMN increased NREM sleep, which was completely abolished by coadministration of 1,3-dimethyl-8-cyclopenthylxanthine, a selective A(1)R antagonist. These results indicate that endogenous adenosine in the TMN suppresses the histaminergic system via A(1)R to promote NREM sleep.


Assuntos
Adenosina/administração & dosagem , Região Hipotalâmica Lateral/efeitos dos fármacos , Receptor A1 de Adenosina/fisiologia , Receptores Histamínicos H1/fisiologia , Fases do Sono/efeitos dos fármacos , Agonistas do Receptor A1 de Adenosina , Inibidores de Adenosina Desaminase , Animais , Coformicina/farmacologia , Inibidores Enzimáticos/farmacologia , Histamina/metabolismo , Região Hipotalâmica Lateral/fisiologia , Masculino , Camundongos , Camundongos Knockout , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Ratos , Ratos Sprague-Dawley , Receptor A1 de Adenosina/genética , Fases do Sono/fisiologia
15.
Proc Natl Acad Sci U S A ; 105(39): 15118-23, 2008 Sep 30.
Artigo em Inglês | MEDLINE | ID: mdl-18809912

RESUMO

We examined how the endogenous anticonvulsant adenosine might influence gamma-aminobutyric acid type A (GABA(A)) receptor stability and which adenosine receptors (ARs) were involved. Upon repetitive activation (GABA 500 microM), GABA(A) receptors, microtransplanted into Xenopus oocytes from neurosurgically resected epileptic human nervous tissues, exhibited an obvious GABA(A)-current (I(GABA)) run-down, which was consistently and significantly reduced by treatment with the nonselective adenosine receptor antagonist CGS15943 (100 nM) or with adenosine deaminase (ADA) (1 units/ml), that inactivates adenosine. It was also found that selective antagonists of A2B (MRS1706, 10 nM) or A3 (MRS1334, 30 nM) receptors reduced I(GABA) run-down, whereas treatment with the specific A1 receptor antagonist DPCPX (10 nM) was ineffective. The selective A2A receptor antagonist SCH58261 (10 nM) reduced or potentiated I(GABA) run-down in approximately 40% and approximately 20% of tested oocytes, respectively. The ADA-resistant, AR agonist 2-chloroadenosine (2-CA) (10 microM) potentiated I(GABA) run-down but only in approximately 20% of tested oocytes. CGS15943 administration again decreased I(GABA) run-down in patch-clamped neurons from either human or rat neocortex slices. I(GABA) run-down in pyramidal neurons was equivalent in A1 receptor-deficient and wt neurons but much larger in neurons from A2A receptor-deficient mice, indicating that, in mouse cortex, GABA(A)-receptor stability is tonically influenced by A2A but not by A1 receptors. I(GABA) run-down from wt mice was not affected by 2-CA, suggesting maximal ARs activity by endogenous adenosine. Our findings strongly suggest that cortical A2-A3 receptors alter the stability of GABA(A) receptors, which could offer therapeutic opportunities.


Assuntos
Adenosina/metabolismo , Anticonvulsivantes/metabolismo , Epilepsia/metabolismo , Antagonistas de Receptores Purinérgicos P1 , Receptores de GABA-A/metabolismo , Adenosina/farmacologia , Adenosina Desaminase/farmacologia , Adulto , Animais , Anticonvulsivantes/farmacologia , Córtex Cerebral/efeitos dos fármacos , Córtex Cerebral/metabolismo , Feminino , Agonistas de Receptores de GABA-A , Humanos , Masculino , Neurônios/metabolismo , Oócitos , Tratos Piramidais/metabolismo , Pirimidinas/farmacologia , Quinazolinas/farmacologia , Ratos , Triazóis/farmacologia , Xantinas/farmacologia , Xenopus , Ácido gama-Aminobutírico/metabolismo , Ácido gama-Aminobutírico/farmacologia
16.
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
18.
Ambio ; 39 Suppl 1: 31-5, 2010.
Artigo em Inglês | MEDLINE | ID: mdl-20873684

RESUMO

There is a need to reduce the amount of fossil energy used for transport, both because of the easily available fossil fuel is becoming sparser and because of climate concerns. In this article, the concept of "peak oil" is briefly presented. Second, a practical approach to reduction of fossil fuel use for transport elaborated by two British commissions is presented. A key feature is the introduction of electric cars. This raises the third issue covered in this article: namely, how battery technology is going to meet the increasing needs posed by the transport sector.


Assuntos
Fontes Geradoras de Energia , Meios de Transporte , Conservação dos Recursos Naturais/métodos , Combustíveis Fósseis
19.
Parkinsonism Relat Disord ; 80 Suppl 1: S3-S6, 2020 11.
Artigo em Inglês | MEDLINE | ID: mdl-33349578

RESUMO

The quest for a non-dopaminergic approach to treating Parkinson's disease (PD) has been quietly progressing over the past several decades, and is now finding its momentum. Here, in what is more a memoir than a comprehensive review, we discuss work carried out over the past 50 years to show that adenosine acts as a critical signaling molecule via actions against a specific family of receptors. Importantly for PD, adenosine A2A receptors have a selective localization to the basal ganglia and specifically to the indirect output pathway, offering a targeted, non-dopaminergic opportunity to modulate basal ganglia output.


Assuntos
Gânglios da Base/metabolismo , Encéfalo/metabolismo , Levodopa/metabolismo , Doença de Parkinson/metabolismo , Receptores Purinérgicos P1/metabolismo , Animais , Encéfalo/patologia , Humanos , Doença de Parkinson/terapia , Receptores Purinérgicos P1/efeitos dos fármacos
20.
Physiol Genomics ; 37(3): 199-210, 2009 May 13.
Artigo em Inglês | MEDLINE | ID: mdl-19258493

RESUMO

Caffeine is the most widely consumed psychoactive substance and has complex pharmacological actions in brain. In this study, we employed a novel drug target validation strategy to uncover the multiple molecular targets of caffeine using combined A(2A) receptor (A(2A)R) knockouts (KO) and microarray profiling. Caffeine (10 mg/kg) elicited a distinct profile of striatal gene expression in WT mice compared with that by A(2A)R gene deletion or by administering caffeine into A(2A)R KO mice. Thus, A(2A)Rs are required but not sufficient to elicit the striatal gene expression by caffeine (10 mg/kg). Caffeine (50 mg/kg) induced complex expression patterns with three distinct sets of striatal genes: 1) one subset overlapped with those elicited by genetic deletion of A(2A)Rs; 2) the second subset elicited by caffeine in WT as well as A(2A)R KO mice; and 3) the third subset elicited by caffeine only in A(2A)R KO mice. Furthermore, striatal gene sets elicited by the phosphodiesterase (PDE) inhibitor rolipram and the GABA(A) receptor antagonist bicucullin, overlapped with the distinct subsets of striatal genes elicited by caffeine (50 mg/kg) administered to A(2A)R KO mice. Finally, Gene Set Enrichment Analysis reveals that adipocyte differentiation/insulin signaling is highly enriched in the striatal gene sets elicited by both low and high doses of caffeine. The identification of these distinct striatal gene populations and their corresponding multiple molecular targets, including A(2A)R, non-A(2A)R (possibly A(1)Rs and pathways associated with PDE and GABA(A)R) and their interactions, and the cellular pathways affected by low and high doses of caffeine, provides molecular insights into the acute pharmacological effects of caffeine in the brain.


Assuntos
Cafeína/farmacologia , Perfilação da Expressão Gênica , Análise de Sequência com Séries de Oligonucleotídeos/métodos , Receptor A2A de Adenosina/fisiologia , Animais , Bicuculina/farmacologia , Estimulantes do Sistema Nervoso Central/farmacologia , Análise por Conglomerados , Relação Dose-Resposta a Droga , Feminino , Antagonistas GABAérgicos/farmacologia , Regulação da Expressão Gênica/efeitos dos fármacos , Masculino , Camundongos , Camundongos Knockout , Neostriado/efeitos dos fármacos , Neostriado/metabolismo , Inibidores de Fosfodiesterase/farmacologia , Receptor A2A de Adenosina/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Rolipram/farmacologia
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa