Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 72
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
Am J Physiol Heart Circ Physiol ; 315(6): H1759-H1764, 2018 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-30265150

RESUMO

H2S is an endogenous gasotransmitter that increases cerebral blood flow. In the cerebral vascular endothelium, H2S is produced by cystathionine δ-lyase (CSE). Endothelin-1 (ET-1) has constrictor and dilator influences on the cerebral circulation. The mechanism of the vasodilation caused by ET-1 may involve endothelium-derived factors. We hypothesize that ET-1-elicited dilation of pial arterioles requires an elevation of H2S production in the cerebral vascular endothelium. We investigated the effects of ET-1 on CSE-catalyzed brain H2S production and pial arteriolar diameter using cranial windows in newborn pigs in vivo. H2S was measured in periarachnoid cerebrospinal fluid. ET-1 (10-12-10-8 M) caused an elevation of H2S that was reduced by the CSE inhibitors propargylglycine (PPG) and ß-cyano-l-alanine (BCA). Low doses of ET-1 (10-12-10-11 M) produced vasodilation of pial arterioles that was blocked PPG and BCA, suggesting the importance of H2S influences. The vasodilator effects of H2S may require activation of smooth muscle cell membrane ATP-sensitive K+ (KATP) channels and large-conductance Ca2+-activated K+ (BK) channels. The KATP inhibitor glibenclamide and the BK inhibitor paxilline blocked CSE/H2S-dependent dilation of pial arterioles to ET-1. In contrast, the vasoconstrictor response of pial arterioles to 10-8 M ET-1 was not modulated by PPG, BCA, glibenclamide, or paxilline and, therefore, was independent of CSE/H2S influences. Pial arteriolar constriction response to higher levels of ET-1 was independent of CSE/H2S and KATP/BKCa channel activation. These data suggest that H2S is an endothelium-derived factor that mediates the vasodilator effects of ET-1 in the cerebral circulation via a mechanism that involves activation of KATP and BK channels in vascular smooth muscle. NEW & NOTEWORTHY Disorders of the cerebral circulation in newborn infants may lead to lifelong neurological disabilities. We report that vasoactive peptide endothelin-1 exhibits vasodilator properties in the neonatal cerebral circulation by stimulating production of H2S, an endothelium-derived messenger with vasodilator properties. The ability of endothelin-1 to stimulate brain production of H2S may counteract the reduction in cerebral blood flow and prevent the cerebral vascular dysfunction caused by stroke, asphyxia, cerebral hypoxia, ischemia, and vasospasm.


Assuntos
Encéfalo/irrigação sanguínea , Circulação Cerebrovascular , Endotelina-1/farmacologia , Sulfitos/líquido cefalorraquidiano , Vasodilatação , Alanina/análogos & derivados , Alanina/farmacologia , Alcinos/farmacologia , Animais , Arteríolas/efeitos dos fármacos , Arteríolas/metabolismo , Arteríolas/fisiologia , Endotélio Vascular/metabolismo , Inibidores Enzimáticos/farmacologia , Glicina/análogos & derivados , Glicina/farmacologia , Canais KATP/metabolismo , Canais de Potássio Ativados por Cálcio de Condutância Alta/metabolismo , Músculo Liso Vascular/metabolismo , Bloqueadores dos Canais de Potássio/farmacologia , Suínos
2.
Am J Physiol Heart Circ Physiol ; 315(4): H978-H988, 2018 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-30028198

RESUMO

Neonatal asphyxia leads to cerebrovascular disease and neurological complications via a mechanism that may involve oxidative stress. Carbon monoxide (CO) is an antioxidant messenger produced via a heme oxygenase (HO)-catalyzed reaction. Cortical astrocytes are the major cells in the brain that express constitutive HO-2 isoform. We tested the hypothesis that CO, produced by astrocytes, has cerebroprotective properties during neonatal asphyxia. We developed a survival model of prolonged asphyxia in newborn pigs that combines insults of severe hypoxia, hypercapnia, and acidosis while avoiding extreme hypotension and cerebral blood flow reduction. During the 60-min asphyxia, CO production by brain and astrocytes was continuously elevated. Excessive formation of reactive oxygen species during asphyxia/reventilation was potentiated by the HO inhibitor tin protoporphyrin, suggesting that endogenous CO has antioxidant effects. Cerebral vascular outcomes tested 24 and 48 h after asphyxia demonstrated the sustained impairment of cerebral vascular responses to astrocyte- and endothelium-specific vasodilators. Postasphyxia cerebral vascular dysfunction was aggravated in newborn pigs pretreated with tin protoporphyrin to inhibit brain HO/CO. The CO donor CO-releasing molecule-A1 (CORM-A1) reduced brain oxidative stress during asphyxia/reventilation and prevented postasphyxia cerebrovascular dysfunction. The antioxidant and antiapoptotic effects of HO/CO and CORM-A1 were confirmed in primary cultures of astrocytes from the neonatal pig brain exposed to glutamate excitotoxicity. Overall, prolonged neonatal asphyxia leads to neurovascular injury via an oxidative stress-mediated mechanism that is counteracted by an astrocyte-based constitutive antioxidant HO/CO system. We propose that gaseous CO or CO donors can be used as novel approaches for prevention of neonatal brain injury caused by prolonged asphyxia. NEW & NOTEWORTHY Asphyxia in newborn infants may lead to lifelong neurological disabilities. Using the model of prolonged asphyxia in newborn piglets, we propose novel antioxidant therapy based on systemic administration of low doses of a carbon monoxide donor that prevent loss of cerebral blood flow regulation and may improve the neurological outcome of asphyxia.


Assuntos
Arteríolas/efeitos dos fármacos , Asfixia Neonatal/tratamento farmacológico , Astrócitos/efeitos dos fármacos , Boranos/farmacologia , Dióxido de Carbono/metabolismo , Carbonatos/farmacologia , Circulação Cerebrovascular/efeitos dos fármacos , Transtornos Cerebrovasculares/prevenção & controle , Fármacos Neuroprotetores/farmacologia , Pia-Máter/irrigação sanguínea , Animais , Animais Recém-Nascidos , Antioxidantes/farmacologia , Apoptose/efeitos dos fármacos , Arteríolas/metabolismo , Arteríolas/fisiopatologia , Asfixia Neonatal/complicações , Asfixia Neonatal/metabolismo , Asfixia Neonatal/fisiopatologia , Astrócitos/metabolismo , Astrócitos/patologia , Velocidade do Fluxo Sanguíneo , Células Cultivadas , Transtornos Cerebrovasculares/etiologia , Transtornos Cerebrovasculares/metabolismo , Transtornos Cerebrovasculares/fisiopatologia , Modelos Animais de Doenças , Feminino , Heme Oxigenase (Desciclizante)/metabolismo , Heme Oxigenase-1/metabolismo , Masculino , Estresse Oxidativo/efeitos dos fármacos , Espécies Reativas de Oxigênio/metabolismo , Sus scrofa , Fatores de Tempo , Vasodilatação/efeitos dos fármacos
3.
Pediatr Res ; 84(2): 290-295, 2018 08.
Artigo em Inglês | MEDLINE | ID: mdl-29907849

RESUMO

BACKGROUND: Hypercapnia causes cerebral vasodilation and increased cerebral blood flow (CBF). During prolonged hypercapnia it is unknown whether cerebral vasodilation persists and whether cerebrovascular function is preserved. We investigated the effects of prolonged severe hypercapnia on pial arteriolar diameters (PAD) and cerebrovascular reactivity to vasodilators and vasoconstrictors. METHODS: Piglets were anesthetized, intubated and ventilated. Closed cranial windows were implanted to measure PAD. Changes in PAD were documented during hypercapnia (PaCO2 75-80 mm Hg). Cerebrovascular reactivity was documented during normocapnia and at 30, 60, and 120 min of hypercapnia. RESULTS: Cerebral vasodilation to hypercapnia was sustained over 120 min. Cerebrovascular responses to vasodilators and vasoconstrictors were preserved during hypercapnia. During hypercapnia, vasodilatory responses to second vasodilators were similar to normocapnia, while exposure to vasoconstrictors caused significant vasoconstriction. CONCLUSIONS: Prolonged severe hypercapnia causes sustained vasodilation of pial arteriolar diameters indicative of hyperperfusion. During hypercapnia, cerebral vascular responses to vasodilators and vasoconstrictors were preserved, suggesting that cerebral vascular function remained intact. Of note, cerebral vessels during hypercapnia were capable of further dilation when exposed to additional cerebral vasodilators and, significant vasoconstriction when exposed to vasoconstrictors. Extrapolating these findings to infants, we suggest that severe hypercapnia should be avoided, because it could cause/increase cerebrovascular injury.


Assuntos
Arteríolas/fisiopatologia , Circulação Cerebrovascular , Hipercapnia/fisiopatologia , Vasodilatação/efeitos dos fármacos , Ácido 15-Hidroxi-11 alfa,9 alfa-(epoximetano)prosta-5,13-dienoico/farmacologia , Animais , Animais Recém-Nascidos , Biomarcadores , Dióxido de Carbono/sangue , Modelos Animais de Doenças , Endotelinas/farmacologia , Feminino , Ácido Glutâmico/farmacologia , Isoproterenol/farmacologia , Masculino , Nitroprussiato/farmacologia , Pia-Máter/irrigação sanguínea , Suínos , Vasoconstrição/efeitos dos fármacos , Vasoconstritores/farmacologia , Vasodilatadores/farmacologia
4.
Pediatr Res ; 82(5): 881-887, 2017 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-28665933

RESUMO

BackgroundThe potential contribution of sex-related variables to cerebrovascular functions in neonates remains elusive. Newborn piglets provide a translationally relevant model for studying the effects of seizures in the neonatal brain. The present study investigated whether sex differences contribute to cerebrovascular functions in healthy and epileptic newborn pigs.MethodsEpileptic seizures were induced in female and male newborn pigs by bicuculline. An antioxidant drug, the carbon monoxide-releasing molecule CORM-A1, was administered enterally before or during seizures. The responses of pial arterioles to endothelium-, astrocyte-, and vascular smooth muscle-dependent vasodilators were tested in intact and 48-h postictal piglets using the cranial window technique.ResultsIn intact newborn pigs, we did not observe any sex-related differences in cerebrovascular functions. In the postictal male and female newborn pigs, a marked reduction in responses of pial arterioles to endothelium- and astrocyte-dependent vasodilators was detected. CORM-A1, administered before or during seizures, greatly improved the outcome of seizures on cerebrovascular functions in both male and female piglets.ConclusionWe found no evidence of sex-related differences in cerebral vasodilator functions in control and epileptic newborn pigs. In both male and female newborns, epileptic seizures lead to prolonged cerebral vascular dysfunction that is effectively prevented by CORM-A1 therapy.


Assuntos
Antioxidantes/farmacologia , Boranos/farmacologia , Carbonatos/farmacologia , Artérias Cerebrais/efeitos dos fármacos , Artérias Cerebrais/fisiopatologia , Transtornos Cerebrovasculares/prevenção & controle , Pia-Máter/irrigação sanguínea , Convulsões/tratamento farmacológico , Vasodilatação/efeitos dos fármacos , Animais , Animais Recém-Nascidos , Astrócitos/efeitos dos fármacos , Astrócitos/metabolismo , Bicuculina , Artérias Cerebrais/metabolismo , Transtornos Cerebrovasculares/etiologia , Transtornos Cerebrovasculares/fisiopatologia , Modelos Animais de Doenças , Células Endoteliais/efeitos dos fármacos , Células Endoteliais/metabolismo , Feminino , Humanos , Masculino , Convulsões/induzido quimicamente , Convulsões/metabolismo , Convulsões/fisiopatologia , Fatores Sexuais , Sus scrofa , Fatores de Tempo , Vasodilatadores/farmacologia
5.
Am J Physiol Heart Circ Physiol ; 311(5): H1202-H1213, 2016 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-27591217

RESUMO

Epileptic seizures in neonates cause cerebrovascular injury and impairment of cerebral blood flow (CBF) regulation. In the bicuculline model of seizures in newborn pigs, we tested the hypothesis that selective head cooling prevents deleterious effects of seizures on cerebral vascular functions. Preventive or therapeutic ictal head cooling was achieved by placing two head ice packs during the preictal and/or ictal states, respectively, for the ∼2-h period of seizures. Head cooling lowered the brain and core temperatures to 25.6 ± 0.3 and 33.5 ± 0.1°C, respectively. Head cooling had no anticonvulsant effects, as it did not affect the bicuculline-evoked electroencephalogram parameters, including amplitude, duration, spectral power, and spike frequency distribution. Acute and long-term cerebral vascular effects of seizures in the normothermic and head-cooled groups were tested during the immediate (2-4 h) and delayed (48 h) postictal periods. Seizure-induced cerebral vascular injury during the immediate postictal period was detected as terminal deoxynucleotidyl transferase-mediated dUTP nick-end labeling-positive staining of cerebral arterioles and a surge of brain-derived circulating endothelial cells in peripheral blood in the normothermic group, but not in the head-cooled groups. During the delayed postictal period, endothelium-dependent cerebral vasodilator responses were greatly reduced in the normothermic group, indicating impaired CBF regulation. Preventive or therapeutic ictal head cooling mitigated the endothelial injury and greatly reduced loss of postictal cerebral vasodilator functions. Overall, head cooling during seizures is a clinically relevant approach to protecting the neonatal brain by preventing cerebrovascular injury and the loss of the endothelium-dependent control of CBF without reducing epileptiform activity.


Assuntos
Arteríolas/fisiopatologia , Artérias Cerebrais/fisiopatologia , Circulação Cerebrovascular , Transtornos Cerebrovasculares/fisiopatologia , Cabeça , Hipotermia Induzida/métodos , Convulsões/fisiopatologia , Vasodilatação/fisiologia , Animais , Animais Recém-Nascidos , Bicuculina/toxicidade , Transtornos Cerebrovasculares/etiologia , Transtornos Cerebrovasculares/prevenção & controle , Convulsivantes/toxicidade , Eletroencefalografia , Células Endoteliais/citologia , Feminino , Marcação In Situ das Extremidades Cortadas , Masculino , Convulsões/induzido quimicamente , Convulsões/complicações , Suínos
6.
J Pharmacol Exp Ther ; 356(1): 106-15, 2016 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-26555891

RESUMO

Despite preventive education, the combined consumption of alcohol and caffeine (particularly from "energy drinks") continues to rise. Physiologic perturbations by separate intake of ethanol and caffeine have been widely documented. However, the biologic actions of the alcohol-caffeine combination and their underlying subcellular mechanisms have been scarcely studied. Using intravital microscopy on a closed-cranial window and isolated, pressurized vessels, we investigated the in vivo and in vitro action of ethanol-caffeine mixtures on cerebral arteries from rats and mice, widely recognized models to address cerebrovascular pathophysiology and pharmacology. Caffeine at concentrations found in human circulation after ingestion of one to two cups of coffee (10 µM) antagonized the endothelium-independent constriction of cerebral arteries evoked by ethanol concentrations found in blood during moderate-heavy alcohol intoxication (40-70 mM). Caffeine antagonism against alcohol was similar whether evaluated in vivo or in vitro, suggesting independence of systemic factors and drug metabolism, but required a functional endothelium. Moreover, caffeine protection against alcohol increased nitric oxide (NO•) levels over those found in the presence of ethanol alone, disappeared upon blocking NO• synthase, and could not be detected in pressurized cerebral arteries from endothelial nitric-oxide synthase knockout (eNOS(-/-)) mice. Finally, incubation of de-endothelialized cerebral arteries with the NO• donor sodium nitroprusside (10 µM) fully restored the protective effect of caffeine. This study demonstrates for the first time that caffeine antagonizes ethanol-induced cerebral artery constriction and identifies endothelial NO• as the critical caffeine effector on smooth muscle targets. Conceivably, situations that perturb endothelial function and/or NO• availability will critically alter caffeine antagonism of alcohol-induced cerebrovascular constriction without significantly disrupting endothelium-independent, alcohol-induced cerebral artery constriction itself.


Assuntos
Cafeína/farmacologia , Depressores do Sistema Nervoso Central/farmacologia , Estimulantes do Sistema Nervoso Central/farmacologia , Artérias Cerebrais/efeitos dos fármacos , Endotélio Vascular/fisiologia , Etanol/antagonistas & inibidores , Etanol/farmacologia , Óxido Nítrico/fisiologia , Vasoconstrição/efeitos dos fármacos , Animais , Cafeína/sangue , Depressores do Sistema Nervoso Central/sangue , Endotélio Vascular/efeitos dos fármacos , Endotélio Vascular/enzimologia , Etanol/sangue , Masculino , Camundongos , Camundongos Knockout , Músculo Liso Vascular/efeitos dos fármacos , Doadores de Óxido Nítrico/farmacologia , Óxido Nítrico Sintase Tipo III/genética , Nitroprussiato/farmacologia , Ratos , Ratos Sprague-Dawley
7.
Physiol Rep ; 3(3)2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-25804265

RESUMO

Neonatal seizures have been associated with cerebrovascular endothelial injury and neurological disabilities. In a piglet model, the long-term loss of endothelial regulation of cerebral blood flow coincides with the surge of brain-derived circulating endothelial cells (BCECs) in blood. We hypothesized that BCECs could serve as a noninvasive biomarker of cerebrovascular injury in neonates with seizures. In a prospective pilot feasibility study, we enrolled newborn infants with confirmed diagnoses of perinatal asphyxia and intraventricular hemorrhage (IVH); both are commonly associated with seizures. Infants without clinical evidence of cerebrovascular injuries were representative of the control group. BCECs were detected in the CD45-negative fraction of peripheral blood mononuclear cells by coexpression of CD31 (common endothelial antigen) and GLUT1 (blood-brain barrier antigen) via automated flow cytometry method. In Infants with asphyxia (n = 12) and those with IVH grade III/IV (n = 5), the BCEC levels were 9.9 ± 0.9% and 19.0 ± 2.0%, respectively. These levels were significantly higher than the control group (n = 27), 0.9 ± 0.2%, P < 0.001. BCECs in infants with cerebrovascular insults with documented clinical seizures (n = 10; 16.8 ± 1.3%) were significantly higher than infants with cerebrovascular insults with subclinical or no seizures (n = 7; 9.5 ± 1.2%); P < 0.001. BCEC levels decreased with seizure control. BCECs levels were elevated in infants with seizures caused by severe IVH and perinatal asphyxia. We suggest that monitoring BCEC levels in peripheral blood can potentially offer a biological marker that reflects cerebrovascular insult and recovery. Further studies with a larger number of patients are required to support these findings.

8.
J Cereb Blood Flow Metab ; 35(2): 193-9, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-25370858

RESUMO

Cerebral blood flow dysregulation caused by oxidative stress contributes to adverse neurologic outcome of seizures. A carbon monoxide (CO) donor CORM-A1 has antioxidant and cytoprotective properties. We investigated whether enteral supplements of CORM-A1 can improve cerebrovascular outcome of bicuculline-induced seizures in newborn piglets. CORM-A1 (2 mg/kg) was given to piglets via an oral gastric tube 10 minutes before or 20 minutes after seizure onset. Enteral CORM-A1 elevated CO in periarachnoid cerebrospinal fluid and produced a dilation of pial arterioles. Postictal cerebral vascular responses to endothelium-, astrocyte-, and vascular smooth muscle-dependent vasodilators were tested 48 hours after seizures by intravital microscopy. The postictal responses of pial arterioles to bradykinin, glutamate, the AMPA receptor agonist quisqualic acid, ADP, and heme were greatly reduced, suggesting that seizures cause injury to endothelial and astrocyte components of the neurovascular unit. In contrast, in the two groups of piglets receiving enteral CORM-A1, the postictal cerebral vascular responsiveness to these dilators was improved. Overall, enteral supplements of CORM-A1 before or during seizures offer a novel effective therapeutic option to deliver cytoprotective mediator CO to the brain, reduce injury to endothelial and astrocyte components of cerebral blood flow regulation and to improve the cerebrovascular outcome of neonatal seizures.


Assuntos
Boranos/farmacologia , Monóxido de Carbono , Carbonatos/farmacologia , Circulação Cerebrovascular/efeitos dos fármacos , Transtornos Cerebrovasculares/tratamento farmacológico , Suplementos Nutricionais , Convulsões/tratamento farmacológico , Difosfato de Adenosina/farmacologia , Animais , Arteríolas/metabolismo , Arteríolas/patologia , Bradicinina/farmacologia , Transtornos Cerebrovasculares/etiologia , Transtornos Cerebrovasculares/metabolismo , Transtornos Cerebrovasculares/patologia , Agonistas de Aminoácidos Excitatórios/farmacologia , Feminino , Ácido Glutâmico/farmacologia , Heme/farmacologia , Masculino , Ácido Quisquálico/farmacologia , Convulsões/complicações , Convulsões/metabolismo , Convulsões/patologia , Suínos , Fatores de Tempo , Vasodilatadores/farmacologia
9.
Physiol Rep ; 2(8)2014 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-25168876

RESUMO

Mechanisms by which Pco2 controls cerebral vascular tone remain uncertain. We hypothesize that potassium channel activation contributes to the neonatal cerebrovascular dilation in response to increases in Paco2. To test this hypothesis, experiments were performed on newborn pigs with surgically implanted, closed cranial windows. Hypercapnia was induced by ventilation with elevated Pco2 gas in the absence and presence of the KATP channel inhibitor, glibenclamide and/or the KCa channel inhibitor, paxillin. Dilations to pinacidil, a selective KATP channel activator, without and with glibenclamide, were used to evaluate the efficacy of KATP channel inhibition. Dilations to NS1619, a selective KCa channel activator, without and with paxillin, were used to evaluate the efficacy of KCa channel inhibition. Cerebrovascular responses to the KATP and KCa channel activators, pinacidil and NS1619, respectively, cAMP-dependent dilator, isoproterenol, and cGMP-dependent dilator, sodium nitroprusside (SNP), were used to evaluate the selectivity of glibenclamide and paxillin. Glibenclamide blocked dilation to pinacidil, but did not inhibit dilations to NS1619, isoproterenol, or SNP. Glibenclamide prior to hypercapnia decreased mean pial arteriole dilation ~60%. Glibenclamide treatment during hypercapnia constricted arterioles ~35%. The level of hypercapnia, Paco2 between 50 and 75 mmHg, did not appear to be involved in efficacy of glibenclamide in blocking dilation to Paco2. Similarly to glibenclamide and KATP channel inhibition, paxillin blocked dilation to the KCa channel agonist, NS1619, and attenuated, but did not block, arteriolar dilation to hypercapnia. Treatment with both glibenclamide and paxillin abolished dilation to hypercapnia. Therefore, either glibenclamide or paxillin that block dilation to their channel agonists, pinacidil or NS1619, respectively, only partially inhibit dilation to hypercapnia. Block of both KATP and KCa channels completely prevent dilation hypercapnia. These data suggest hypercapnia activates both KATP and KCa channels leading to cerebral arteriolar dilation in newborn pigs.

10.
Alcohol Clin Exp Res ; 38(5): 1216-26, 2014 May.
Artigo em Inglês | MEDLINE | ID: mdl-24588122

RESUMO

BACKGROUND: Binge drinking represents the major form of excessive alcohol (ethanol [EtOH]) consumption in the United States. Episodic (such as binge) drinking results in blood alcohol levels (BAL) of 18 to 80 mM and leads to alcohol-induced cerebral artery constriction (AICAC). AICAC was shown to arise from EtOH-induced inhibition of large-conductance, calcium/voltage-gated potassium (BK) channels in the vascular smooth muscle. Factors that modulate BK channel-mediated AICAC remain largely unknown. METHODS: Male Sprague Dawley rats were placed on high-cholesterol (2% of cholesterol) diet for 18 to 23 weeks. Their littermates were placed on control iso-caloric diet. AICAC was evaluated both in vivo and in vitro, by means of pial arteriole diameter monitoring through a closed cranial window and diameter measurements of isolated, pressurized cerebral arteries. Cholesterol level in the cerebral artery tissue was manipulated by methyl-ß-cyclodextrin to reverse dietary-induced accumulation of cholesterol. BK channel surface presence on the plasma membrane of cerebral artery myocytes was evaluated by immunofluorescence staining. BK channel function in pressurized cerebral artery was assessed using selective BK channel blocker paxilline. RESULTS: Within 5 minutes of 50 mM EtOH injection into carotid artery in vivo, arteriole diameter decreased by 20% in control group. Pial arteriole constriction was significantly reduced in rats on high-cholesterol diet, resulting in only 10% reduction in diameter. BAL in both groups, however, remained the same. Significant reduction in AICAC in group on high-cholesterol diet compared to control was also observed after middle cerebral artery dissection and in vitro pressurization at 60 mmHg, this reduction remaining after endothelium removal. Cholesterol level in de-endothelialized cerebral arteries was significantly increased in rats on high-cholesterol diet. Removal of excessive cholesterol content restored AICAC to the level observed in cerebral arteries of rats on normal diet. Immunofluorescence staining of BK channel-forming and accessory, smooth muscle-specific ß1 subunit in freshly isolated cerebral artery myocyte showed that high-cholesterol diet did not down-regulate surface presence of BK protein. However, paxilline-induced cerebral artery constriction was diminished in arteries from rats on high-cholesterol diet. CONCLUSIONS: Our data indicate that dietary cholesterol protects against AICAC. This protection is caused by cholesterol buildup in the arterial tissue and diminished function (but not surface presence) of EtOH target-BK channel.


Assuntos
Artérias Cerebrais/efeitos dos fármacos , Colesterol na Dieta/farmacologia , Etanol/farmacologia , Vasoconstrição/efeitos dos fármacos , Animais , HDL-Colesterol/sangue , LDL-Colesterol/sangue , Canais de Potássio Ativados por Cálcio de Condutância Alta/efeitos dos fármacos , Masculino , Ratos Sprague-Dawley
11.
Am J Physiol Cell Physiol ; 304(11): C1105-15, 2013 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-23576575

RESUMO

In cerebral microvascular endothelial cells (CMVEC) of newborn pigs, glutamate at excitotoxic concentrations (mM) causes apoptosis mediated by reactive oxygen species (ROS). Carbon monoxide (CO) produced by CMVEC or delivered by a CO-releasing molecule, CORM-A1, has antioxidant properties. We tested the hypothesis that CORM-A1 prevents cerebrovascular endothelial barrier dysfunction caused by glutamate excitotoxicity. First, we identified the glutamate receptors (GluRs) and enzymatic sources of ROS involved in the mechanism of endothelial apoptosis. In glutamate-exposed CMVEC, ROS formation and apoptosis were blocked by rotenone, 2-thenoyltrifluoroacetone (TTFA), and antimycin, indicating that mitochondrial complexes I, II, and III are the major sources of oxidative stress. Agonists of ionotropic GluRs (iGluRs) N-methyl-D-aspartate (NMDA), cis-ACPD, AMPA, and kainate increased ROS production and apoptosis, whereas iGluR antagonists exhibited antiapoptotic properties, suggesting that iGluRs mediate glutamate-induced endothelial apoptosis. The functional consequences of endothelial injury were tested in the model of blood-brain barrier (BBB) composed of CMVEC monolayer on semipermeable membranes. Glutamate and iGluR agonists reduced transendothelial electrical resistance and increased endothelial paracellular permeability to 3-kDa dextran. CORM-A1 exhibited potent antioxidant and antiapoptotic properties in CMVEC and completely prevented BBB dysfunction caused by glutamate and iGluR agonists. Overall, the endothelial component of the BBB is a cellular target for excitotoxic glutamate that, via a mechanism involving a iGluR-mediated activation of mitochondrial ROS production and apoptosis, leads to BBB opening that may be prevented by the antioxidant and antiapoptotic actions of CORMs. Antioxidant CORMs therapy may help preserve BBB functional integrity in neonatal cerebrovascular disease.


Assuntos
Apoptose/fisiologia , Barreira Hematoencefálica/metabolismo , Boranos/metabolismo , Carbonatos/metabolismo , Células Endoteliais/metabolismo , Estresse Oxidativo/fisiologia , Receptores Ionotrópicos de Glutamato/metabolismo , Animais , Barreira Hematoencefálica/patologia , Encéfalo/metabolismo , Encéfalo/patologia , Fragmentação do DNA , Células Endoteliais/patologia , Aminoácidos Excitatórios/metabolismo , Aminoácidos Excitatórios/toxicidade , Feminino , Ácido Glutâmico/metabolismo , Ácido Glutâmico/toxicidade , Masculino , Espécies Reativas de Oxigênio/metabolismo , Suínos
12.
Mol Pharmacol ; 83(5): 1030-44, 2013 May.
Artigo em Inglês | MEDLINE | ID: mdl-23455312

RESUMO

The Ca(2+)/voltage-gated K(+) large conductance (BK) channel ß1 subunit is particularly abundant in vascular smooth muscle. By determining their phenotype, BK ß1 allows the BK channels to reduce myogenic tone, facilitating vasodilation. The endogenous steroid lithocholic acid (LCA) dilates cerebral arteries via BK channel activation, which requires recognition by a BK ß1 site that includes Thr169. Whether exogenous nonsteroidal agents can access this site to selectively activate ß1-containing BK channels and evoke vasodilation remain unknown. We performed a chemical structure database similarity search using LCA as a template, along with a two-step reaction to generate sodium 3-hydroxyolean-12-en-30-oate (HENA). HENA activated the BK (cbv1 + ß1) channels cloned from rat cerebral artery myocytes with a potency (EC50 = 53 µM) similar to and an efficacy (×2.5 potentiation) significantly greater than that of LCA. This HENA action was replicated on native channels in rat cerebral artery myocytes. HENA failed to activate the channels made of cbv1 + ß2, ß3, ß4, or ß1T169A, indicating that this drug selectively targets ß1-containing BK channels via the BK ß1 steroid-sensing site. HENA (3-45 µM) dilated the rat and C57BL/6 mouse pressurized cerebral arteries. Consistent with the electrophysiologic results, this effect was larger than that of LCA. HENA failed to dilate the arteries from the KCNMB1 knockout mouse, underscoring BK ß1's role in HENA action. Finally, carotid artery-infusion of HENA (45 µM) dilated the pial cerebral arterioles via selective BK-channel targeting. In conclusion, we have identified for the first time a nonsteroidal agent that selectively activates ß1-containing BK channels by targeting the steroid-sensing site in BK ß1, rendering vasodilation.


Assuntos
Artérias Cerebrais/efeitos dos fármacos , Colanos/farmacologia , Subunidades beta do Canal de Potássio Ativado por Cálcio de Condutância Alta/metabolismo , Esteroides/farmacologia , Vasodilatação/efeitos dos fármacos , Animais , Artérias Cerebrais/metabolismo , Colanos/química , Feminino , Subunidades beta do Canal de Potássio Ativado por Cálcio de Condutância Alta/química , Ácido Litocólico/farmacologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Monócitos/efeitos dos fármacos , Monócitos/metabolismo , Ratos , Ratos Sprague-Dawley , Esteroides/química , Xenopus laevis
13.
Am J Physiol Heart Circ Physiol ; 302(11): H2257-66, 2012 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-22467311

RESUMO

In newborn pigs, vasodilation of pial arterioles in response to glutamate is mediated via carbon monoxide (CO), a gaseous messenger endogenously produced from heme degradation by a heme oxygenase (HO)-catalyzed reaction. We addressed the hypothesis that ionotropic glutamate receptors (iGluRs), including N-methyl-D-aspartic acid (NMDA)- and 2-amino-3-(5-methyl-3-oxo-1,2-oxazol-4-yl) propanoic acid (AMPA)/kainate-type receptors, expressed in cortical astrocytes mediate glutamate-induced astrocyte HO activation that leads to cerebral vasodilation. Acute vasoactive effects of topical iGluR agonists were determined by intravital microscopy using closed cranial windows in anesthetized newborn pigs. iGluR agonists, including NMDA, (±)1-aminocyclopentane-cis-1,3-dicarboxylic acid (cis-ACPD), AMPA, and kainate, produced pial arteriolar dilation. Topical L-2-aminoadipic acid, a gliotoxin that selectively disrupts glia limitans, reduced vasodilation caused by iGluR agonists, but not by hypercapnia, bradykinin, or sodium nitroprusside. In freshly isolated and cultured cortical astrocytes constitutively expressing HO-2, iGluR agonists NMDA, cis-ACPD, AMPA, and kainate rapidly increased CO production two- to threefold. Astrocytes overexpressing inducible HO-1 had high baseline CO but were less sensitive to glutamate stimulation of CO production when compared with HO-2-expressing astrocytes. Glutamate-induced astrocyte HO-2-mediated CO production was inhibited by either the NMDA receptor antagonist (R)-3C4HPG or the AMPA/kainate receptor antagonist DNQX. Accordingly, either antagonist abolished pial arteriolar dilation in response to glutamate, NMDA, and AMPA, indicating functional interaction among various subtypes of astrocytic iGluRs in response to glutamate stimulation. Overall, these data indicate that the astrocyte component of the neurovascular unit is responsible for the vasodilation response of pial arterioles to topically applied glutamate via iGluRs that are functionally linked to activation of constitutive HO in newborn piglets.


Assuntos
Arteríolas/efeitos dos fármacos , Astrócitos/metabolismo , Monóxido de Carbono/metabolismo , Circulação Cerebrovascular/fisiologia , Glutamatos/farmacologia , Receptores de Glutamato/metabolismo , Vasodilatação/efeitos dos fármacos , Animais , Animais Recém-Nascidos , Arteríolas/fisiologia , Astrócitos/citologia , Astrócitos/efeitos dos fármacos , Células Cultivadas , Feminino , Heme Oxigenase (Desciclizante)/metabolismo , Técnicas In Vitro , Ácido Caínico/farmacologia , Masculino , Modelos Animais , N-Metilaspartato/farmacologia , Receptores de Glutamato/efeitos dos fármacos , Suínos , Vasodilatação/fisiologia , Ácido alfa-Amino-3-hidroxi-5-metil-4-isoxazol Propiônico/farmacologia
14.
J Physiol ; 590(11): 2709-20, 2012 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-22508960

RESUMO

Hydrogen sulfide (H2S) is a gaseous vasodilator produced by endothelial cells. Mechanisms by which H2S induces vasodilatation are unclear. We tested the hypothesis that H2S dilates cerebral arterioles by modulating local and global intracellular Ca²âº signals in smooth muscle cells. High-speed confocal imaging revealed that Na2S, an H2S donor, increased Ca²âº spark frequency ∼1.43-fold and decreased global intracellular Ca²âº concentration ([Ca²âº]i) by ∼37 nM in smooth muscle cells of intact piglet cerebral arterioles. In contrast, H2S did not alter Ca²âº wave frequency. In voltage-clamped (-40 mV) cells, H2S increased the frequency of iberiotoxin-sensitive, Ca²âº spark-induced transient Ca²âº-activated K⁺ (KCa) currents ∼1.83-fold, but did not alter the amplitude of these events. H2S did not alter the activity of single KCa channels recorded in the absence of Ca²âº sparks in arteriole smooth muscle cells. H2S increased SR Ca²âº load ([Ca²âº]SR), measured as caffeine (10 and 20mM)-induced [Ca²âº]i transients, ∼1.5-fold. H2S hyperpolarized (by ∼18 mV) and dilated pressurized (40 mmHg) cerebral arterioles. Iberiotoxin, a KCa channel blocker, reduced H2S-induced hyperpolarization by ∼51%. Iberiotoxin and ryanodine, a ryanodine receptor channel inhibitor, reduced H2S-induced vasodilatation by ∼38 and ∼37%, respectively. In summary, our data indicate that H2S elevates [Ca²âº]SR, leading to Ca²âº spark activation in cerebral arteriole smooth muscle cells. The subsequent elevation in transient KCa current frequency leads to membrane hyperpolarization, a reduction in global [Ca²âº]i and vasodilatation.


Assuntos
Encéfalo/irrigação sanguínea , Cálcio/fisiologia , Sulfeto de Hidrogênio/farmacologia , Miócitos de Músculo Liso/efeitos dos fármacos , Vasodilatação/efeitos dos fármacos , Animais , Arteríolas/efeitos dos fármacos , Arteríolas/fisiologia , Encéfalo/efeitos dos fármacos , Encéfalo/fisiologia , Técnicas In Vitro , Potenciais da Membrana/efeitos dos fármacos , Miócitos de Músculo Liso/fisiologia , Canais de Potássio Cálcio-Ativados/fisiologia , Canal de Liberação de Cálcio do Receptor de Rianodina/fisiologia , Retículo Sarcoplasmático/fisiologia , Suínos
15.
J Cereb Blood Flow Metab ; 32(6): 1024-34, 2012 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-22354150

RESUMO

Postictal cerebrovascular dysfunction is an adverse effect of seizures in newborn piglets. The brain heme oxygenase (HO) provides protection against cerebrovascular dysfunction. We investigated the contribution of reactive oxygen species (ROS) to seizure-induced vascular damage and the mechanism of HO vasoprotection. In a bicuculline model of seizures, we addressed the hypotheses: (1) seizures increase brain ROS; (2) ROS contribute to cerebral vascular dysfunction; (3) ROS initiate a vasoprotective mechanisms by activating endogenous HO; and (4) HO products have antioxidant properties. As assessed by dihydroethidium oxidation (ox-DHE), seizures increased ROS in cerebral vessels and cortical astrocytes; ox-DHE elevation was prevented by tiron and apocynin. An HO inhibitor, tin protoporphyrin, potentiated, whereas an HO-1 inducer, cobalt protoporphyrin, blocked seizure-induced increase in DHE oxidation. Heme oxygenase products carbon monoxide (CO) (CORM-A1) and bilirubin attenuated ox-DHE elevation during seizures. Antioxidants tiron and bilirubin prevented the loss of postictal cerebrovascular dilations to bradykinin, glutamate, and sodium nitroprusside. Tiron and apocynin abrogated activation of the brain HO during seizures. Overall, these data suggest that long-term adverse cerebrovascular effects of seizures are attributed to oxidative stress. On the other hand, seizure-induced ROS are required for activation of the endogenous antioxidant HO/CO/bilirubin system that alleviates oxidative stress-induced loss of postictal cerebrovascular function in piglets.


Assuntos
Antioxidantes/metabolismo , Bilirrubina/metabolismo , Monóxido de Carbono/metabolismo , Circulação Cerebrovascular , Heme Oxigenase (Desciclizante)/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Convulsões/metabolismo , Animais , Astrócitos/metabolismo , Bradicinina/metabolismo , Inibidores Enzimáticos/farmacologia , Ácido Glutâmico/metabolismo , Metaloporfirinas/farmacologia , Estresse Oxidativo , Protoporfirinas/farmacologia , Espécies Reativas de Oxigênio/metabolismo , Convulsões/fisiopatologia , Suínos
16.
Am J Physiol Heart Circ Physiol ; 301(6): H2482-7, 2011 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-21984542

RESUMO

Arachidonic acid (AA) and prostaglandin (PG) E(2) stimulate carbon monoxide (CO) production, and AA metabolism is known to be associated with the generation of reactive oxygen species (ROS). This study was conducted to address the hypothesis that CO and/or ROS mediate cerebrovascular dilation in newborn pigs. Experiments were performed on anesthetized newborn pigs with closed cranial windows. Different concentrations of AA (10(-8)-10(-6) M), PGE(2) (10(-8)-10(-6) M), iloprost (10(-8)-10(-6) M), and their vehicle (artificial cerebrospinal fluid) were given. Piglets with PGE(2) and iloprost received indomethacin (5 mg/kg iv) to inhibit cyclooxygenase. AA, PGE(2), and iloprost caused concentration-dependent increases in pial arteriolar diameter. The effects of both AA and PGE(2) in producing cerebral vascular dilation and associated CO production were blocked by the heme oxygenase inhibitor chromium mesoporphyrin (2 × 10(-5) M), but not by the prostacyclin analog, iloprost. ROS inhibitor tempol (SOD mimetic) (1 × 10(-5) M) and the H(2)O(2) scavenger catalase (1,000 U/ml) also do not block these vasodilator effects of AA and PGE(2). Heme-L-lysinate-induced cerebrovascular dilation and CO production was blocked by chromium mesoporphyrin. Hypoxanthine plus xanthine oxidase, a combination that is known to generate ROS, caused pial arteriolar dilation and CO production that was inhibited by tempol and catalase. These data suggest that AA- and PGE(2)-induced cerebral vascular dilation is mediated by CO, independent of ROS.


Assuntos
Ácido Araquidônico/metabolismo , Monóxido de Carbono/metabolismo , Dinoprostona/metabolismo , Pia-Máter/irrigação sanguínea , Espécies Reativas de Oxigênio/metabolismo , Vasodilatação , Administração Tópica , Animais , Animais Recém-Nascidos , Antioxidantes/administração & dosagem , Arteríolas/efeitos dos fármacos , Arteríolas/metabolismo , Inibidores de Ciclo-Oxigenase/administração & dosagem , Relação Dose-Resposta a Droga , Inibidores Enzimáticos/administração & dosagem , Heme Oxigenase (Desciclizante)/antagonistas & inibidores , Heme Oxigenase (Desciclizante)/metabolismo , Transdução de Sinais , Suínos , Vasodilatação/efeitos dos fármacos , Vasodilatadores/administração & dosagem , Xantina Oxidase/metabolismo
17.
Am J Physiol Heart Circ Physiol ; 301(2): H428-33, 2011 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-21572018

RESUMO

Glutamate-stimulated, astrocyte-derived carbon monoxide (CO) causes cerebral arteriole dilation by activating smooth muscle cell large-conductance Ca(2+)-activated K(+) channels. Here, we examined the hypothesis that glutamate activates heme oxygenase (HO)-2 and CO production via the intracellular Ca(2+) concentration ([Ca(2+)](i))/Ca(2+)-calmodulin signaling pathway in newborn pig astrocytes. The major findings are: 1) glutamate stimulated Ca(2+) transients and increased steady-state [Ca(2+)](i) in cerebral cortical astrocytes in primary culture, 2) in astrocytes permeabilized with ionomycin, elevation of [Ca(2+)](i) concentration-dependently increased CO production, 3) glutamate did not affect CO production at any [Ca(2+)](i) when the [Ca(2+)](i) was held constant, 4) thapsigargin, a sarco/endoplasmic reticulum Ca(2+)-ATPase blocker, decreased basal CO production and blocked glutamate-induced increases in CO, and 5) calmidazolium, a calmodulin inhibitor, blocked CO production induced by glutamate and by [Ca(2+)](i) elevation. Taken together, our data are consistent with the hypothesis that glutamate elevates [Ca(2+)](i) in astrocytes, leading to Ca(2+)- and calmodulin-dependent HO-2 activation, and CO production.


Assuntos
Astrócitos/metabolismo , Sinalização do Cálcio , Monóxido de Carbono/metabolismo , Ácido Glutâmico/metabolismo , Análise de Variância , Animais , Animais Recém-Nascidos , Astrócitos/efeitos dos fármacos , Sinalização do Cálcio/efeitos dos fármacos , Calmodulina/antagonistas & inibidores , Calmodulina/metabolismo , Células Cultivadas , Relação Dose-Resposta a Droga , Ativação Enzimática , Inibidores Enzimáticos/farmacologia , Heme Oxigenase (Desciclizante)/metabolismo , Imidazóis/farmacologia , Ionomicina/farmacologia , Ionóforos/farmacologia , ATPases Transportadoras de Cálcio do Retículo Sarcoplasmático/antagonistas & inibidores , ATPases Transportadoras de Cálcio do Retículo Sarcoplasmático/metabolismo , Suínos , Tapsigargina/farmacologia , Fatores de Tempo , Regulação para Cima
18.
Am J Physiol Heart Circ Physiol ; 301(1): H1-H11, 2011 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-21498777

RESUMO

Carbon monoxide (CO) is produced by heme oxygenase (HO)-catalyzed heme degradation to CO, iron, and biliverdin. HO has two active isoforms, HO-1 (inducible) and HO-2 (constitutive). HO-2, but not HO-1, is highly expressed in endothelial and smooth muscle cells and in adjacent astrocytes in the brain. HO-1 is expressed basally only in the spleen and liver but can be induced to a varying extent in most tissues. Elevating heme, protein phosphorylation, Ca(2+) influx, and Ca(2+)/calmodulin-dependent processes increase HO-2 activity. CO dilates cerebral arterioles and may constrict or dilate skeletal muscle and renal arterioles. Selected vasodilatory stimuli, including seizures, glutamatergic stimulation, hypoxia, hypotension, and ADP, increase CO, and the inhibition of HO attenuates the dilation to these stimuli. Astrocytic HO-2-derived CO causes glutamatergic dilation of pial arterioles. CO dilates by activating smooth muscle cell large-conductance Ca(2+)-activated K(+) (BK(Ca)) channels. CO binds to BK(Ca) channel-bound heme, leading to an increase in Ca(2+) sparks-to-BK(Ca) channel coupling. Also, CO may bind directly to the BK(Ca) channel at several locations. Endothelial nitric oxide and prostacyclin interact with HO/CO in circulatory regulation. In cerebral arterioles in vivo, in contrast to dilation to acute CO, a prolonged exposure of cerebral arterioles to elevated CO produces progressive constriction by inhibiting nitric oxide synthase. The HO/CO system is highly protective to the vasculature. CO suppresses apoptosis and inhibits components of endogenous oxidant-generating pathways. Bilirubin is a potent reactive oxygen species scavenger. Still many questions remain about the physiology and biochemistry of HO/CO in the circulatory system and about the function and dysfunction of this gaseous mediator system.


Assuntos
Vasos Sanguíneos/fisiologia , Monóxido de Carbono/fisiologia , Fenômenos Fisiológicos Cardiovasculares , Animais , Vasos Sanguíneos/metabolismo , Monóxido de Carbono/metabolismo , Circulação Cerebrovascular/fisiologia , Regulação Enzimológica da Expressão Gênica/fisiologia , Heme Oxigenase (Desciclizante)/biossíntese , Heme Oxigenase (Desciclizante)/genética , Heme Oxigenase-1/biossíntese , Heme Oxigenase-1/genética , Humanos , Vasodilatação/fisiologia
19.
Am J Physiol Heart Circ Physiol ; 300(6): H2088-95, 2011 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-21421823

RESUMO

Hydrogen sulfide (H(2)S) is a gaseous signaling molecule that appears to contribute to the regulation of vascular tone and blood pressure. Multiple potential mechanisms of vascular regulation by H(2)S exist. Here, we tested the hypothesis that piglet cerebral arteriole smooth muscle cells generate ATP-sensitive K(+) (K(ATP)) currents and that H(2)S induces vasodilation by activating K(ATP) currents. Gas chromatography/mass spectrometry data demonstrated that after placing Na(2)S, an H(2)S donor, in solution, it rapidly (1 min) converts to H(2)S. Patch-clamp electrophysiology indicated that pinacidil (a K(ATP) channel activator), Na(2)S, and NaHS (another H(2)S donor) activated K(+) currents at physiological steady-state voltage (-50 mV) in isolated cerebral arteriole smooth muscle cells. Glibenclamide, a selective K(ATP) channel inhibitor, fully reversed pinacidil-induced K(+) currents and partially reversed (∼58%) H(2)S-induced K(+) currents. Western blot analysis indicated that piglet arterioles expressed inwardly rectifying K(+) 6.1 (K(ir)6.1) channel and sulfonylurea receptor 2B (SUR2B) K(ATP) channel subunits. Pinacidil dilated pressurized (40 mmHg) piglet arterioles, and glibenclamide fully reversed this effect. Na(2)S also induced reversible and repeatable vasodilation with an EC(50) of ∼30 µM, and this effect was partially reversed (∼55%) by glibenclamide. Vasoregulation by H(2)S was also studied in pressurized resistance-size cerebral arteries of mice with a genetic deletion in the gene encoding SUR2 (SUR2 null). Pinacidil- and H(2)S-induced vasodilations were smaller in arterioles of SUR2 null mice than in wild-type controls. These data indicate that smooth muscle cell K(ATP) currents control newborn cerebral arteriole contractility and that H(2)S dilates cerebral arterioles by activating smooth muscle cell K(ATP) channels containing SUR2 subunits.


Assuntos
Membrana Celular/metabolismo , Artérias Cerebrais/efeitos dos fármacos , Sulfeto de Hidrogênio/farmacologia , Canais KATP/metabolismo , Músculo Liso Vascular/metabolismo , Vasodilatação/efeitos dos fármacos , Transportadores de Cassetes de Ligação de ATP/genética , Transportadores de Cassetes de Ligação de ATP/metabolismo , Animais , Arteríolas/efeitos dos fármacos , Arteríolas/fisiologia , Artérias Cerebrais/fisiologia , Feminino , Glibureto/farmacologia , Canais KATP/efeitos dos fármacos , Masculino , Camundongos , Camundongos Knockout , Modelos Animais , Técnicas de Patch-Clamp , Pinacidil/farmacologia , Canais de Potássio Corretores do Fluxo de Internalização/genética , Canais de Potássio Corretores do Fluxo de Internalização/metabolismo , Receptores de Droga/genética , Receptores de Droga/metabolismo , Receptores de Sulfonilureias , Suínos , Vasodilatação/fisiologia , Vasodilatadores/farmacologia
20.
Am J Physiol Heart Circ Physiol ; 300(2): H440-7, 2011 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-21131483

RESUMO

Hydrogen sulfide (H2S) is a gaseous signaling molecule that appears to be involved in numerous biological processes, including regulation of blood pressure and vascular tone. The present study is designed to address the hypothesis that H2S is a functionally significant, endogenous dilator in the newborn cerebrovascular circulation. In vivo experiments were conducted using newborn pigs with surgically implanted, closed, cranial windows. Topical application of H2S concentration-dependently (10(-6) to 2×10(-4) M) dilated pial arterioles. This dilation was blocked by glibenclamide (10(-6) M). L-cysteine, the substrate of the H2S-producing enzymes cystathionine γ-lyase (CSE) and cystathionine ß-synthase (CBS), also dilated pial arterioles. The dilation to L-cysteine was blocked by the CSE inhibitor d,l-propargylglycine (PPG, 10 mM) but was unaffected by the CBS inhibitor amino-oxyacetate (AOA, 1 mM). Western blots detected CSE, but not CBS, in cerebral microvessels, whereas CBS is detected in brain parenchyma. Immunohistological CSE expression is predominantly vascular while CBS is expressed mainly in neurons and astrocytes. L-cysteine (5 mM) increased H2S concentration in cerebrospinal fluid (CSF), measured by GC-MS, from 561±205 to 2,783±818 nM before but not during treatment with PPG (1,030±70 to 622±78 nM). Dilation to hypercapnia was inhibited by PPG but not AOA. Hypercapnia increased CSF H2S concentration from 763±243 to 4,337±1789 nM before but not during PPG treatment (357±178 vs. 425±217 nM). These data show that H2S is a dilator of the newborn cerebral circulation and that endogenous CSE can produce sufficient H2S to decrease vascular tone. H2S appears to be a physiologically significant dilator in the cerebral circulation.


Assuntos
Animais Recém-Nascidos/fisiologia , Capilares/efeitos dos fármacos , Circulação Cerebrovascular/efeitos dos fármacos , Sulfeto de Hidrogênio/metabolismo , Sulfeto de Hidrogênio/farmacologia , Músculo Liso Vascular/efeitos dos fármacos , Animais , Arteríolas/efeitos dos fármacos , Arteríolas/fisiologia , Química Encefálica/fisiologia , Cisteína/farmacologia , Glibureto/farmacologia , Hipercapnia/fisiopatologia , Hipóxia/fisiopatologia , Imuno-Histoquímica , Isoproterenol/farmacologia , Canais KATP/antagonistas & inibidores , Tono Muscular/efeitos dos fármacos , Nitroprussiato/farmacologia , Suínos , Vasodilatação/efeitos dos fármacos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA