Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
1.
J Cereb Blood Flow Metab ; 42(10): 1920-1932, 2022 10.
Artigo em Inglês | MEDLINE | ID: mdl-35673977

RESUMO

Cerebrovascular effects of ß-site amyloid precursor protein-cleaving enzyme 1 (BACE1) inactivation have not been systematically studied. In the present study we employed cultured human brain microvascular endothelial cells (BMECs), BACE1-knockout (BACE1-/-) mice and conditional (tamoxifen-induced) endothelium-specific BACE1-knockout (eBACE1-/-) mice to determine effect of BACE1 inhibition on expression and function of endothelial nitric oxide synthase (eNOS). Deletion of BACE1 caused upregulation of eNOS and glypican-1 (GPC1) in human BMECs treated with BACE1-siRNA, and cerebral microvessels of male BACE1-/- mice and male eBACE1-/- mice. In addition, BACE1siRNA treatment increased NO production in human BMECs. These effects appeared to be independent of amyloid ß-peptide production. Furthermore, adenoviral-mediated overexpression of BACE1 in human BMECs down-regulated GPC1 and eNOS. Treatment of human BMECs with GPC1siRNA suppressed mRNA and protein levels of eNOS. In basilar arteries of male eBACE1-/- mice, endothelium-dependent relaxations to acetylcholine and endothelium-independent relaxations to NO donor, DEA-NONOate, were not affected, consistent with unchanged expression of eNOS and phosphorylation of eNOS at Ser1177 in large cerebral arteries. In aggregate, our findings suggest that under physiological conditions, inactivation of endothelial BACE1 increases expression of eNOS in cerebral microvessels but not in large brain arteries. This effect appears to be mediated by increased GPC1 expression.


Assuntos
Precursor de Proteína beta-Amiloide , Óxido Nítrico Sintase Tipo III , Acetilcolina/metabolismo , Secretases da Proteína Precursora do Amiloide , Peptídeos beta-Amiloides/metabolismo , Precursor de Proteína beta-Amiloide/metabolismo , Animais , Ácido Aspártico Endopeptidases/genética , Células Endoteliais/metabolismo , Endotélio Vascular/metabolismo , Glipicanas/metabolismo , Humanos , Masculino , Camundongos , Óxido Nítrico/metabolismo , Óxido Nítrico Sintase Tipo III/metabolismo , RNA Mensageiro/metabolismo , RNA Interferente Pequeno/metabolismo , Tamoxifeno
2.
J Cereb Blood Flow Metab ; 37(1): 106-122, 2017 01.
Artigo em Inglês | MEDLINE | ID: mdl-26661245

RESUMO

We tested hypothesis that activation of the prostacyclin (PGI2) receptor (IP receptor) signaling pathway in cerebral microvessels plays an important role in the metabolism of amyloid precursor protein (APP). In human brain microvascular endothelial cells activation of IP receptor with the stable analogue of PGI2, iloprost, stimulated expression of amyloid precursor protein and a disintegrin and metalloprotease 10 (ADAM10), resulting in an increased production of the neuroprotective and anticoagulant molecule, soluble APPα (sAPPα). Selective agonist of IP receptor, cicaprost, and adenylyl cyclase activator, forskolin, also enhanced expression of amyloid precursor protein and ADAM10. Notably, in cerebral microvessels of IP receptor knockout mice, protein levels of APP and ADAM10 were reduced. In addition, iloprost increased protein levels of peroxisome proliferator-activated receptor δ (PPARδ) in human brain microvascular endothelial cells. PPARδ-siRNA abolished iloprost-augmented protein expression of ADAM10. In contrast, GW501516 (a selective agonist of PPARδ) upregulated ADAM10 and increased production of sAPPα. Genetic deletion of endothelial PPARδ (ePPARδ-/-) in mice significantly reduced cerebral microvascular expression of ADAM10 and production of sAPPα. In vivo treatment with GW501516 increased sAPPα content in hippocampus of wild type mice but not in hippocampus of ePPARδ-/- mice. Our findings identified previously unrecognized role of IP-PPARδ signal transduction pathway in the production of sAPPα in cerebral microvasculature.


Assuntos
Secretases da Proteína Precursora do Amiloide/biossíntese , Encéfalo/irrigação sanguínea , Endotélio Vascular/metabolismo , Epoprostenol/fisiologia , Microvasos/metabolismo , Animais , Epoprostenol/metabolismo , Hipocampo/metabolismo , Humanos , Camundongos , Camundongos Knockout , PPAR gama/metabolismo , Receptores de Epoprostenol/metabolismo , Transdução de Sinais/fisiologia , Solubilidade
3.
PLoS One ; 10(12): e0144062, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26629821

RESUMO

The hippocampus is one of the earliest and most affected regions in Alzheimer's disease (AD), followed by the cortex while the cerebellum is largely spared. Importantly, endothelial dysfunction is a common feature of cerebral blood vessels in AD. In this study, we sought to determine if regional heterogeneity of cerebral microvessels might help explain the susceptibility of the hippocampus and cortex as compared to the cerebellum. We isolated microvessels from wild type mice from the cerebellum, cortex, and hippocampus to characterize their vascular phenotype. Superoxide anion was significantly higher in microvessels isolated from the cortex and hippocampus as compared to the cerebellum. Importantly, protein levels of NADPH oxidase (NOX)-2 and NOX-4 were significantly higher in the cortical and hippocampal microvessels as compared to microvessels from the cerebellum. In addition, expression of manganese superoxide dismutase protein was significantly lower in microvessels from the cortex and hippocampus as compared to cerebellum while other antioxidant enzymes were unchanged. There was no difference in eNOS protein expression between the microvessels of the three brain regions; however, bioavailability of tetrahydrobiopterin (BH4), an essential cofactor for eNOS activity, was significantly reduced in microvessels from the hippocampus and cortex as compared to the cerebellum. Higher levels of superoxide and reduced tetrahydrobiopterin bioavailability may help explain the vulnerability of the hippocampus and cortical microvessels to oxidative stress and development of endothelial dysfunction.


Assuntos
Cerebelo/patologia , Córtex Cerebral/patologia , Hipocampo/patologia , Microvasos/patologia , Estresse Oxidativo/fisiologia , Doença de Alzheimer/metabolismo , Doença de Alzheimer/fisiopatologia , Animais , Antioxidantes/metabolismo , Disponibilidade Biológica , Biopterinas/análogos & derivados , Biopterinas/metabolismo , Cerebelo/metabolismo , Córtex Cerebral/metabolismo , Hipocampo/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Microvasos/metabolismo , Óxido Nítrico Sintase Tipo III/metabolismo , Superóxido Dismutase/metabolismo , Superóxidos/metabolismo
4.
Brain Res ; 1625: 198-205, 2015 Nov 02.
Artigo em Inglês | MEDLINE | ID: mdl-26343845

RESUMO

Tetrahydrobiopterin (BH4) is a critical determinant of nitric oxide (NO) production by nitric oxide synthase (NOS) in the vascular endothelium and its biosynthesis is regulated by the enzymatic activity of GTP-cyclohydrolase I (GTPCH I). The present study was designed to determine the effects of endothelium-targeted overexpression of GTPCH I (eGCH-Tg) on murine cerebral vascular function. Endothelium targeted over-expression of GTPCH I was associated with a significant increase in levels of BH4, as well as its oxidized product, 7,8-dihydrobiopterin (7,8-BH2) in cerebral microvessels. Importantly, ratio of BH4 to 7,8-BH2, indicative of BH4 available for eNOS activation, was significantly increased in eGCH-Tg mice. However, expression of endothelial NOS, levels of nitrate/nitrite--indicative of NO production--remained unchanged between cerebral microvessels of wild-type and eGCH-Tg mice. Furthermore, increased BH4 biosynthesis neither affected production of superoxide anion nor expression of antioxidant proteins. Moreover, endothelium-specific GTPCH I overexpression did not alter intracellular levels of cGMP, reflective of NO signaling in cerebral microvessels. The obtained results suggest that, despite a significant increase in BH4 bioavailability, generation of endothelial NO in cerebral microvessels remained unchanged in eGCH-Tg mice. We conclude that under physiological conditions the levels of BH4 in the cerebral microvessels are optimal for activation of endothelial NOS and NO/cGMP signaling.


Assuntos
Endotélio Vascular/metabolismo , GTP Cicloidrolase/metabolismo , Microvasos/citologia , Microvasos/metabolismo , Análise de Variância , Animais , Biopterinas/análogos & derivados , Biopterinas/metabolismo , Catecolaminas/sangue , Artérias Cerebrais/citologia , GMP Cíclico/metabolismo , GTP Cicloidrolase/genética , Regulação da Expressão Gênica/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Óxido Nítrico/metabolismo , Óxido Nítrico Sintase/metabolismo , Óxido Nítrico Sintase Tipo III/metabolismo , Superóxidos/metabolismo
5.
J Neurochem ; 134(6): 1129-38, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-26111938

RESUMO

In this study, we tested the hypothesis that reduced bioavailability of tetrahydrobiopterin (BH4) is a major mechanism responsible for pathogenesis of endothelial dysfunction in cerebral microvessels of transgenic mice expressing the Swedish double mutation of human amyloid precursor protein (APP) (Tg2576 mice). Endothelial nitric oxide synthase (eNOS) protein expression was significantly increased in cerebral vasculature of Tg2576 mice. In contrast, bioavailability of BH4 was significantly reduced (p < 0.05). Moreover, superoxide anion production was increased in cerebral microvessels of Tg2576 mice (p < 0.05). Incubation with NOS inhibitor, Nω-nitro-L-arginine methyl ester, decreased superoxide anion indicating that uncoupled eNOS is most likely the source of superoxide anion. Increasing BH4 bioavailability either exogenously by BH4 supplementation or endogenously by treatment with the selective peroxisome proliferator-activated receptor--delta activator GW501516 (2 mg/kg/day, 14 days) attenuated eNOS uncoupling and decreased superoxide anion production in cerebral microvessels of Tg2576 mice (p < 0.05). Treatment with GW501516 restored the biological activity of endothelial nitric oxide in cerebral microvessels of Tg2576 mice, as indicated by the increased nitrite/nitrate content and 3,5-cyclic guanosine monophosphate levels (p < 0.05). Our studies indicate that sub-optimal BH4 bioavailability in cerebral vasculature is an important contributor to oxidant stress and endothelial dysfunction in Tg2576 mouse model of Alzheimer's disease. Existing evidence suggests that Aß peptides-induced up-regulation of expression and activity of NADPH oxidase causes increased production of superoxide anion (.O2(-)). .O2(-) can also be converted to hydrogen peroxide (H2O2) by enzymatic activity of superoxide dismutase (SOD) or spontaneous dismutation. Elevation of .O2(-) and H2O2 might cause oxidation of tetrahydrobiopterin (BH4) to dihydrobiopterin (BH2) and subsequent uncoupling of endothelial nitric oxide synthase (eNOS) (a) thus reducing levels of nitric oxide (NO) and 3',5'-cyclic guanosine monophosphate (cGMP). Supplementation of BH4 or activation of PPARδ prevents detrimental effects of eNOS uncoupling by restoring bioavailability of BH4 and scavenging of .O2(-), respectively (b). Activation of PPARδ also increases expression of catalase thereby inactivating H2O2. Generation of H2O2 by uncoupled eNOS in cerebral microvessels of Tg2576 mice is hypothetical.


Assuntos
Doença de Alzheimer/metabolismo , Precursor de Proteína beta-Amiloide/metabolismo , Biopterinas/análogos & derivados , Óxido Nítrico Sintase Tipo III/metabolismo , Estresse Oxidativo/fisiologia , Animais , Biopterinas/metabolismo , Western Blotting , Encéfalo/metabolismo , Modelos Animais de Doenças , Endotélio Vascular/metabolismo , Feminino , Humanos , Camundongos , Camundongos Transgênicos , Microvasos
6.
J Neurochem ; 131(4): 521-9, 2014 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-25041251

RESUMO

This study was designed to determine whether treatment with erythropoietin (EPO) could protect cerebral microvasculature against the pathological consequences of endothelial nitric oxide (NO) synthase uncoupling. Wild-type and GTP cyclohydrolase I (GTPCH-I)-deficient hph1 mice were administered EPO (1000 U/kg/day, s.c., 3 days). Cerebral microvessels of hph1 mice demonstrated reduced tetrahydrobiopterin (BH4) bioavailability, increased production of superoxide anions and impaired endothelial NO signaling. Treatment of hph1 mice with EPO attenuated the levels of 7,8-dihydrobiopterin, the oxidized product of BH4, and significantly increased the ratio of BH4 to 7,8-dihydrobiopterin. Moreover, EPO decreased the levels of superoxide anions and increased NO bioavailability in cerebral microvessels of hph1 mice. Attenuated oxidation of BH4 and inhibition of endothelial NO synthase uncoupling were explained by the increased expression of antioxidant proteins, manganese superoxide dismutase, and catalase. The protective effects of EPO observed in cerebral microvessels of hph1 mice were also observed in GTPCH-I siRNA-treated human brain microvascular endothelial cells exposed to EPO (1 U/mL or 10 U/mL; 3 days). Our results suggest that EPO might protect the neurovascular unit against oxidative stress by restoring bioavailability of BH4 and endothelial NO in the cerebral microvascular endothelium. We demonstrate that treatment with erythropoietin (EPO) could protect cerebral microvasculature against the pathological consequences of endothelial nitric oxide (NO) synthase uncoupling. Our results suggest that EPO might protect the neurovascular unit against oxidative stress by restoring bioavailability of tetrahydrobiopterin (BH4) and endothelial nitric oxide.


Assuntos
Biopterinas/análogos & derivados , Eritropoetina/metabolismo , Microvasos/efeitos dos fármacos , Óxido Nítrico Sintase Tipo III/metabolismo , Estresse Oxidativo/efeitos dos fármacos , Animais , Biopterinas/metabolismo , Células Cultivadas , Células Endoteliais/efeitos dos fármacos , Células Endoteliais/metabolismo , Humanos , Técnicas In Vitro , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Microvasos/citologia , Mutação/genética , Mutação/imunologia , Óxido Nítrico/metabolismo , Complexo Repressor Polycomb 1/genética , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/metabolismo
7.
Am J Physiol Heart Circ Physiol ; 306(7): H1001-10, 2014 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-24486511

RESUMO

Peroxisome proliferator-activated receptor (PPAR)-δ is a nuclear hormone receptor that is mainly involved in lipid metabolism. Recent studies have suggested that PPAR-δ agonists exert vascular protective effects. The present study was designed to characterize vascular function in mice with genetic inactivation of PPAR-δ in the endothelium. Mice with vascular endothelial cell-specific deletion of the PPAR-δ gene (ePPARδ(-/-) mice) were generated using loxP/Cre technology. ePPARδ(-/-) mice were normotensive and did not display any sign of metabolic syndrome. Endothelium-dependent relaxations to ACh and endothelium-independent relaxations to the nitric oxide (NO) donor diethylammonium (Z)-1-(N,N-diethylamino)diazen-1-ium-1,2-diolate were both significantly impaired in the aorta and carotid arteries of ePPARδ(-/-) mice (P < 0.05). In ePPARδ(-/-) mouse aortas, phosphorylation of endothelial NO synthase at Ser(1177) was significantly decreased (P < 0.05). However, basal levels of cGMP were unexpectedly increased (P < 0.05). Enzymatic activity of GTP-cyclohydrolase I and tetrahydrobiopterin levels were also enhanced in ePPARδ(-/-) mice (P < 0.05). Most notably, endothelium-specific deletion of the PPAR-δ gene significantly decreased protein expressions of catalase and glutathione peroxidase 1 and resulted in increased levels of H2O2 in the aorta (P < 0.05). In contrast, superoxide anion production was unaltered. Moreover, treatment with catalase prevented the endothelial dysfunction and elevation of cGMP detected in aortas of ePPARδ(-/-) mice. The findings suggest that increased levels of cGMP caused by H2O2 impair vasodilator reactivity to endogenous and exogenous NO. We speculate that chronic elevation of H2O2 predisposes PPAR-δ-deficient arteries to oxidative stress and vascular dysfunction.


Assuntos
Aorta/metabolismo , Artéria Carótida Primitiva/metabolismo , Endotélio Vascular/metabolismo , PPAR gama/deficiência , Vasodilatação , Animais , Antioxidantes/metabolismo , Aorta/efeitos dos fármacos , Aorta/fisiopatologia , Biopterinas/análogos & derivados , Biopterinas/metabolismo , Artéria Carótida Primitiva/efeitos dos fármacos , Artéria Carótida Primitiva/fisiopatologia , Catalase/metabolismo , GMP Cíclico/metabolismo , Relação Dose-Resposta a Droga , Endotélio Vascular/efeitos dos fármacos , Endotélio Vascular/fisiopatologia , GTP Cicloidrolase/metabolismo , Glutationa Peroxidase/metabolismo , Peróxido de Hidrogênio/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Óxido Nítrico/metabolismo , Óxido Nítrico Sintase Tipo III/metabolismo , Estresse Oxidativo , PPAR gama/genética , Fosforilação , Superóxidos/metabolismo , Vasodilatação/efeitos dos fármacos , Vasodilatadores/farmacologia , Glutationa Peroxidase GPX1
8.
Brain Res ; 1483: 89-95, 2012 Nov 05.
Artigo em Inglês | MEDLINE | ID: mdl-22982594

RESUMO

Peroxisome proliferator-activated receptor delta (PPARδ) is ubiquitously expressed in the vasculature, including cerebral circulation. The role of PPARδ in metabolism of tetrahydrobiopterin (BH4) has not been studied in the cerebral microvasculature. In the present study, the effects of PPARδ agonist GW501516 on uncoupling of endothelial nitric oxide synthase (eNOS) were determined in cerebral microvessels of BH4-deficient hph-1 mice. Wild-type (B6CBA) and hph-1 mice were orally gavaged with a selective PPARδ activator, GW501516 (2 mg/kg/day) for 14 days, and thereafter, cerebral microvessels were isolated and studied. Treatment of hph-1 mice with GW501516 significantly reduced oxidation of BH4 and increased the ratio of BH4 to 7,8-BH2 (P<0.05, n=6-9). Attenuation of L-NAME-inhibitable superoxide anion levels by GW501516 demonstrated that activation of PPARδ might prevent uncoupling of endothelial nitric oxide synthase (eNOS, P<0.05, n=6-9). Western blotting studies demonstrated that GW501516 selectively increased the endothelial expressions of CuZn superoxide dismutase (P<0.05, n=6-9) and catalase (P<0.05, n=6-8). PPARδ activation increased the total nitrite and nitrate (NO2+NO3) content in cerebral microvessels (P<0.05, n=6). Obtained results suggest that in vivo activation of PPARδ prevents eNOS uncoupling, restores bioavailability of NO and may help preserve endothelial function in the BH4-deficient cerebral circulation.


Assuntos
Microvasos/efeitos dos fármacos , Microvasos/enzimologia , Óxido Nítrico Sintase Tipo III/metabolismo , Complexo Repressor Polycomb 1/genética , Tiazóis/farmacologia , Análise de Variância , Animais , Biopterinas/análogos & derivados , Biopterinas/metabolismo , Pressão Sanguínea/efeitos dos fármacos , Córtex Cerebral/anatomia & histologia , Inibidores Enzimáticos/farmacologia , GTP Cicloidrolase/metabolismo , Regulação Enzimológica da Expressão Gênica/efeitos dos fármacos , Técnicas In Vitro , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Transgênicos , Microvasos/citologia , Mutação/genética , NG-Nitroarginina Metil Éster/farmacologia , PPAR delta/antagonistas & inibidores , Superóxidos/metabolismo
9.
J Neurochem ; 122(6): 1211-8, 2012 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-22784235

RESUMO

In this study, we used the GTP cyclohydrolase I-deficient mice, i.e., hyperphenylalaninemic (hph-1) mice, to test the hypothesis that the loss of tetrahydrobiopterin (BH(4)) in cerebral microvessels causes endothelial nitric oxide synthase (eNOS) uncoupling, resulting in increased superoxide anion production and inhibition of endothelial nitric oxide signaling. Both homozygous mutant (hph-1(-/-)) and heterozygous mutant (hph-1(+/-) mice) demonstrated reduction in GTP cyclohydrolase I activity and reduced bioavailability of BH(4). In the cerebral microvessels of hph-1(+/-) and hph-1(-/-) mice, increased superoxide anion production was inhibited by supplementation of BH(4) or NOS inhibitor- L- N(G) -nitro arginine-methyl ester, indicative of eNOS uncoupling. Expression of 3-nitrotyrosine was significantly increased, whereas NO production and cGMP levels were significantly reduced. Expressions of antioxidant enzymes namely copper and zinc superoxide dismutase, manganese superoxide dismutase, and catalase were not affected by uncoupling of eNOS. Reduced levels of BH(4), increased superoxide anion production, as well as inhibition of NO signaling were not different between the microvessels of male and female mice. The results of our study are the first to demonstrate that, regardless of gender, reduced BH(4) bioavailability causes eNOS uncoupling, increases superoxide anion production, inhibits eNOS/cGMP signaling, and imposes significant oxidative stress in the cerebral microvasculature.


Assuntos
Encéfalo/irrigação sanguínea , Microvasos/enzimologia , Óxido Nítrico Sintase Tipo III , Óxido Nítrico/antagonistas & inibidores , Óxido Nítrico/fisiologia , Fenilalanina Hidroxilase/deficiência , Transdução de Sinais , Superóxidos/metabolismo , Animais , Encéfalo/enzimologia , GMP Cíclico/antagonistas & inibidores , GMP Cíclico/fisiologia , Feminino , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Mutantes , Óxido Nítrico Sintase Tipo III/antagonistas & inibidores , Óxido Nítrico Sintase Tipo III/metabolismo , Óxido Nítrico Sintase Tipo III/fisiologia , Estresse Oxidativo/fisiologia , Transdução de Sinais/fisiologia
10.
Adv Pharmacol ; 60: 257-85, 2010.
Artigo em Inglês | MEDLINE | ID: mdl-21081221

RESUMO

Erythropoietin (EPO) is a therapeutic product of recombinant DNA technology and it has been in clinical use as stimulator of erythropoiesis over the last two decades. Identification of EPO and its receptor (EPOR) in the cardiovascular system expanded understanding of physiological and pathophysiological role of EPO. In experimental models of cardiovascular and cerebrovascular disorders, EPO exerts protection either by preventing apoptosis of cardiac myocytes, smooth muscle cells, and endothelial cells, or by increasing endothelial production of nitric oxide. In addition, EPO stimulates mobilization of progenitor cells from bone marrow thereby accelerating repair of injured endothelium and neovascularization. A novel signal transduction pathway involving EPOR--ß-common heteroreceptor is postulated to enhance EPO-mediated tissue protection. A better understanding of the role of ß-common receptor signaling as well as development of novel analogs of EPO with enhanced nonhematopoietic protective effects may expand clinical application of EPO in prevention and treatment of cardiovascular and cerebrovascular disorders.


Assuntos
Sistema Cardiovascular/efeitos dos fármacos , Sistema Cardiovascular/metabolismo , Eritropoetina/metabolismo , Eritropoetina/farmacologia , Animais , Doenças Cardiovasculares/tratamento farmacológico , Eritropoetina/uso terapêutico , Humanos
11.
Stroke ; 41(2): 350-6, 2010 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-20019327

RESUMO

BACKGROUND AND PURPOSE: The role of brain-derived neurotrophic factor (BDNF) and its receptor, tropomyosin receptor kinase B, in control of cerebral circulation is poorly understood. The present study was designed to investigate the cerebral vascular effects of BDNF in vivo. METHODS: Replication incompetent adenovirus encoding either rat BDNF (AdBDNF) or green fluorescent protein was injected intracisternally into rabbits. Forty-eight hours later, animals were euthanized. Plasma and cerebrospinal fluid levels of BDNF were measured by enzyme-linked immunosorbent assay, vasomotor function of isolated basilar arteries was studied in organ chambers, protein expression in the basilar arteries was studied by Western blotting, prostanoid levels were measured by enzyme-linked immunosorbent assay, and cyclic adenosine 3',5'-monophosphate levels were measured by radioimmunoassay. RESULTS: The levels of BDNF in the cerebrospinal fluid were significantly elevated in AdBDNF-treated rabbits as compared with adenovirus encoding green fluorescent protein-treated rabbits (37+/-5 ng/mL versus 0.006+/-0.003 ng/mL, respectively; P<0.05; n=14). Western blotting studies revealed that in basilar arteries, AdBDNF increased protein expression of prostacyclin synthase, whereas expression of endothelial nitric oxide synthase and phosphorylated (Ser 1177) endothelial nitric oxide synthase remained unchanged. During incubation with arachidonic acid (1 micromol/L), PGI(2) production and levels of cyclic adenosine 3',5'-monophosphate were significantly elevated only in AdBDNF-treated rabbit basilar arteries (P<0.05, n=6). Relaxations to acetylcholine (10(-9) to 10(-5) mol/L) and arachidonic acid (10(-9) to 10(-5) mol/L) were significantly potentiated in basilar arteries from rabbits injected with AdBDNF. Potentiation of relaxations to acetylcholine in AdBDNF-treated basilar arteries was inhibited by the nonselective cyclooxygenase inhibitor, indomethacin (10(-5) mol/L, P<0.05, n=6) and constitutive phospholipase A(2) inhibitor, AACOCF3 (2x10(-5) mol/L, P<0.05, n=5). CONCLUSIONS: Our results demonstrate that in cerebral arteries, BDNF-induced activation of tropomyosin receptor kinase B receptor signaling in vivo promotes prostacyclin biosynthesis. These findings provide novel mechanistic insight into the vascular protective effect of BDNF in cerebral circulation.


Assuntos
Fator Neurotrófico Derivado do Encéfalo/líquido cefalorraquidiano , Artérias Cerebrais/metabolismo , Circulação Cerebrovascular/fisiologia , Epoprostenol/biossíntese , Receptor trkB/metabolismo , Vasodilatação/fisiologia , Adenoviridae/genética , Animais , Ácido Araquidônico/farmacologia , Artéria Basilar/metabolismo , Isquemia Encefálica/metabolismo , Isquemia Encefálica/fisiopatologia , Fator Neurotrófico Derivado do Encéfalo/sangue , Fator Neurotrófico Derivado do Encéfalo/genética , Proteínas do Líquido Cefalorraquidiano/análise , Proteínas do Líquido Cefalorraquidiano/metabolismo , AMP Cíclico/metabolismo , Inibidores de Ciclo-Oxigenase/farmacologia , Sistema Enzimático do Citocromo P-450/metabolismo , Modelos Animais de Doenças , Vetores Genéticos , Oxirredutases Intramoleculares/metabolismo , Masculino , Músculo Liso Vascular/metabolismo , Coelhos , Transfecção , Regulação para Cima/genética
12.
Peptides ; 29(8): 1451-5, 2008 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-18448202

RESUMO

The present study aimed to define the ability of erythropoietin (EPO) to mobilize hematopoietic stem cells (c-kit(+)/sca-1(+)/lin-1(-); KSL-cells) and hematopoietic progenitor cells (CD34(+) cells), including vascular endothelial growth factor receptor 2 expressing hematopoietic progenitor cells (CD34(+)/Flk-1(+) cells). We also sought to determine the role of endothelial nitric oxide synthase (eNOS) in EPO-induced mobilization. Wild type (WT) and eNOS(-/-) mice were injected bi-weekly with recombinant erythropoietin (EPO, 1000U/kg, s.c.) for 14 days. EPO increased the number of KSL, CD34(+), CD34(+)/Flk-1(+) cells in circulating blood of wild type mice. These effects of EPO were abolished in eNOS(-/-) mice. Our results demonstrate that, EPO stimulates mobilization of hematopoietic stem and progenitor cells. This effect of EPO is critically dependent on activation of eNOS.


Assuntos
Eritropoetina/fisiologia , Mobilização de Células-Tronco Hematopoéticas , Óxido Nítrico Sintase Tipo III/metabolismo , Animais , Ativação Enzimática/fisiologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout
13.
Eur J Pharmacol ; 572(2-3): 189-96, 2007 Oct 31.
Artigo em Inglês | MEDLINE | ID: mdl-17640632

RESUMO

The ability of agmatine, formed from L-arginine by the enzyme arginine decarboxylase (ADC), to modulate vasomotor function in rat aorta was investigated in the present study. Agmatine-mediated modulation of vasomotor tone was studied in organ chambers, protein expression quantified by Western blot analysis and cyclic guanosine 5'-monophosphate (cGMP) levels measured by radioimmunoassay. Agmatine (10(-10) to 10(-3) M) produced concentration-dependent relaxations (82+/-5%) in phenylephrine-contracted endothelium intact rat aorta. Relaxations to agmatine were diminished on denudation of endothelium and nitric oxide synthase (NOS) inhibition by L-Nomega-nitro arginine or soluble guanylate cyclase inhibition by 1H-[1,2,4]oxadiazolo[4,3-a]quinoxalin-1-one (P<0.001) abolished agmatine-mediated relaxations, while relaxations were insensitive to inducible NOS inhibition by 1400W. Agmatine-treated aorta demonstrated increased protein expression of phosphorylated S473-Akt and phosphorylated S1177-endothelial nitric oxide synthase (eNOS), and elevated the levels of cyclic GMP (P<0.01). Agmatine-mediated potentiation of relaxations and elevation of cGMP levels was sensitive to phosphatidylinositol 3'-kinase inhibitor, wortmannin. Relaxations to agmatine were also affected by pre-treatment with tetraethylammonium (P<0.01) or apamin (P<0.05), and were not affected by charybdotoxin. Relaxations to agmatine were partially affected by pre-treatment of aortic rings with barium chloride (P<0.05), and glybenclamide (P<0.05). Results obtained suggest that agmatine activates protein kinase B/Akt to phosphorylate eNOS and elevate cyclic GMP levels to produce vasodilatation of aorta. Agmatine-mediated relaxations in rat aorta seems to be mediated mainly by endothelial NO-mediated activation of small conductance Ca2+-activated K+ channels, and partly by ATP-sensitive and inward rectifying K+ channels.


Assuntos
Agmatina/metabolismo , Endotélio Vascular/fisiologia , Óxido Nítrico Sintase Tipo III/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Agmatina/farmacologia , Animais , Aorta Torácica/fisiologia , GMP Cíclico/metabolismo , Ativação Enzimática , Técnicas In Vitro , Canais KATP/fisiologia , Canais de Potássio Ativados por Cálcio de Condutância Alta/fisiologia , Masculino , Relaxamento Muscular , Músculo Liso Vascular/fisiologia , Fosforilação , Canais de Potássio/fisiologia , Ratos , Ratos Sprague-Dawley , Canais de Potássio Ativados por Cálcio de Condutância Baixa/fisiologia
14.
Circ Res ; 100(9): 1379-88, 2007 May 11.
Artigo em Inglês | MEDLINE | ID: mdl-17395873

RESUMO

In the present study we hypothesized that endothelial progenitor cells (EPCs) enhance production of vasoprotective substances in cerebral arteries. Isolated mononuclear cells from rabbit peripheral blood were cultured in endothelial growth medium (EGM-2) for 7 days to yield EPCs. Rabbit basilar arteries were exposed to autologous EPCs ( approximately 5x10(5) cells) in vitro or in vivo. Twenty-four hours after intracisternal delivery of autologous EPCs, basilar arteries were isolated and expression of vasoregulatory proteins, production of prostacyclin (PGI(2)), and cAMP were determined. Arteries transplanted with EPCs demonstrated increased protein expression of cyclooxygenase-2 and PGI(2) in adventitia, media, and endothelium. Furthermore, production of PGI(2) and arterial content of cAMP, second messenger for PGI(2), were significantly augmented after transplantation of EPCs. In contrast, production of thromboxane A(2) was significantly reduced, whereas production of prostaglandin E(2) remained unchanged. The increased production of PGI(2) and arterial content of cAMP were inhibited only by a selective cyclooxygenase-2 inhibitor, NS-398. In vitro or in vivo treatment of basilar artery with conditioned media from EPCs also caused increase in cyclooxygenase-2 and PGI(2) synthase protein expression associated with elevation of cAMP. Our results suggest that in cerebral arteries, paracrine effect of EPCs promotes vasoprotection by increasing PGI(2) production and intracellular concentration of cAMP. This effect appears to be mediated by activation of arachidonic acid metabolism via stimulation of cyclooxygenase-2/PGI(2) synthase pathway.


Assuntos
Artéria Basilar/metabolismo , Ciclo-Oxigenase 2/biossíntese , Células Endoteliais/citologia , Epoprostenol/biossíntese , Células-Tronco/fisiologia , Animais , Células Cultivadas , AMP Cíclico/fisiologia , Ativação Enzimática , Histamina/farmacologia , Masculino , Nitrobenzenos/farmacologia , Coelhos , Transplante de Células-Tronco , Sulfonamidas/farmacologia
15.
Acta Pharmacol Sin ; 27(11): 1389-94, 2006 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-17049112

RESUMO

Cerebral vasospasm after subarachnoid hemorrhage (SAH) is a major clinical problem causing cerebral ischemia and infarction. The pathogenesis of vasospasm is related to a number of pathological processes including endothelial damage and alterations in vasomotor function leading to narrowing of arterial diameter and a subsequent decrease in cerebral blood flow. Discovery of the tissue protective effects of erythropoietin (EPO) stimulated the search for therapeutic application of EPO for the prevention and treatment of cerebrovascular disease. Recent studies have identified the role of EPO in vascular protection mediated by the preservation of endothelial cell integrity and stimulation of angiogenesis. In this review, we discuss the EPO-induced activation of endothelial nitric oxide (NO) synthase and its contribution to the prevention of cerebral vasospasm.


Assuntos
Artérias Cerebrais/enzimologia , Eritropoetina/farmacologia , Eritropoetina/fisiologia , Óxido Nítrico Sintase Tipo III/fisiologia , Vasoespasmo Intracraniano/metabolismo , Animais , Humanos , Janus Quinase 2/metabolismo , Fosforilação , Ligação Proteica , Receptores da Eritropoetina/metabolismo , Proteínas Recombinantes
16.
Am J Physiol Heart Circ Physiol ; 291(2): H781-6, 2006 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-16565320

RESUMO

The discovery of tissue protective effects of erythropoietin has stimulated significant interest in erythropoietin (Epo) as a novel therapeutic approach to vascular protection. The present study was designed to determine the cerebral vascular effects of recombinant Epo in vivo. Recombinant adenoviral vectors (10(9) plaque-forming units/animal) encoding genes for human erythropoietin (AdEpo) and beta-galactosidase (AdLacZ) were injected into the cisterna magna of rabbits. After 48 h, basilar arteries were harvested for analysis of vasomotor function, Western blotting, and measurement of cGMP levels. Gene transfer of AdEpo increased the expressions of recombinant Epo and its receptor in the basilar arteries. Arteries exposed to recombinant Epo demonstrated attenuation of contractile responses to histamine (10(-9) to 10(-5) mol/l) (P < 0.05, n = 5). Endothelium-dependent relaxations to acetylcholine (10(-9) to 10(-5) mol/l) were significantly augmented (P < 0.05, n = 5), whereas endothelium-independent relaxations to a nitric oxide (NO) donor 2-(N,N-diethylamino)diazenolate-2-oxide sodium salt remained unchanged in AdEpo-transduced basilar arteries. Transduction with AdEpo increased the protein expression of endothelial NO synthase (eNOS) and phosphorylated the S1177 form of the enzyme. Basal levels of cGMP were significantly elevated in arteries transduced with AdEpo consistent with increased NO production. Our studies suggest that in cerebral circulation, Epo enhances endothelium-dependent vasodilatation mediated by NO. This effect could play an important role in the vascular protective effect of Epo.


Assuntos
Artérias Cerebrais/enzimologia , Eritropoetina/farmacologia , Óxido Nítrico Sintase Tipo III/metabolismo , Adenoviridae/genética , Animais , Western Blotting , Artérias Cerebrais/efeitos dos fármacos , AMP Cíclico/metabolismo , GMP Cíclico/metabolismo , Endotélio Vascular/fisiologia , Eritropoetina/genética , Técnicas de Transferência de Genes , Vetores Genéticos , Histamina/metabolismo , Humanos , Técnicas In Vitro , Óperon Lac/genética , Masculino , Doadores de Óxido Nítrico , Coelhos , Proteínas Recombinantes , Estimulação Química , Transdução Genética , Vasodilatação/efeitos dos fármacos , Vasodilatação/fisiologia , beta-Galactosidase/genética
17.
Stroke ; 36(12): 2731-7, 2005 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-16269632

RESUMO

BACKGROUND AND PURPOSE: In the present study, the effect of subarachnoid hemorrhage (SAH) on the phosphorylation of endothelial NO synthase (eNOS) and the ability of recombinant erythropoietin (Epo) to augment this vasodilator mechanism in the spastic arteries were studied. METHODS: Recombinant adenoviral vectors (10(9) plaque-forming units per animal) encoding genes for human Epo (AdEpo), and beta-galactosidase were injected immediately after injection of autologous arterial blood into the cisterna magna (day 0) of rabbits. Cerebral angiography was performed on day 0 and day 2, and basilar arteries were harvested for Western blots, measurement of cGMP levels, and analysis of vasomotor functions. RESULTS: Injection of autologous arterial blood into cisterna magna resulted in significant vasospasm of the basilar arteries. Despite the narrowing of arterial diameter and reduced expression of eNOS, expressions of phosphorylated protein kinase B (Akt) and phosphorylated eNOS were significantly increased in spastic arteries. Gene transfer of AdEpo reversed the vasospasm. AdEpo-transduced basilar arteries demonstrated significant augmentation of the endothelium-dependent relaxations to acetylcholine, whereas the relaxations to an NO donor, 2-(N,N-diethylamino)diazenolate-2-oxide sodium salt, were not affected. Transduction with AdEpo further increased the expression of phosphorylated Akt and eNOS and elevated basal levels of cGMP in the spastic arteries. CONCLUSIONS: Phosphorylation of eNOS appears to be an adaptive mechanism activated during development of vasospasm. The vascular protective effect of Epo against cerebral vasospasm induced by SAH may be mediated in part by phosphorylation of Akt/eNOS.


Assuntos
Endotélio Vascular/metabolismo , Eritropoetina/farmacologia , Óxido Nítrico Sintase/metabolismo , Hemorragia Subaracnóidea/complicações , Vasoespasmo Intracraniano/tratamento farmacológico , Vasoespasmo Intracraniano/metabolismo , Animais , Angiografia Cerebral , Técnicas de Transferência de Genes , Masculino , Fosforilação , Coelhos , Proteínas Recombinantes , Regulação para Cima , Vasoespasmo Intracraniano/diagnóstico por imagem , Vasoespasmo Intracraniano/etiologia
18.
Stroke ; 36(1): 158-60, 2005 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-15569867

RESUMO

BACKGROUND AND PURPOSE: Mutation of serine 1179 to aspartate on the endothelial NO synthase (eNOS) increases NO production in the absence of stimulation by agonists. The present study was designed to determine the effect of recombinant S1179DeNOS gene expression on the vasomotor function of human pial arteries. METHODS: Pial arteries were isolated from 28 patients undergoing temporal lobectomy for intractable seizures. Adenoviral vectors (10(10) pfu/mL) encoding beta-galactosidase (AdCMVLacZ) or S1179DeNOS (AdCMVS1179DeNOS) were used for ex vivo gene transfer, and vasomotor function was evaluated in control and transduced arteries. RESULTS: Contractions to cumulative additions of U46619 were not affected by expression of LacZ or S1179DeNOS. Endothelium-dependent relaxations to bradykinin or endothelium-independent relaxations to Diethylaminodiazen-1-ium-1,2-dioate were significantly reduced in arteries expressing S1179DeNOS. A superoxide dismutase mimetic, manganese (III) tetrakis (4-benzoic acid) porphyrin chloride, failed to improve the reduced relaxations to bradykinin. The levels of cGMP were significantly elevated in arteries expressing S1179DeNOS. CONCLUSIONS: Our results support the concept that high local production of NO in pial arterial wall causes adaptive reduction of vasodilator reactivity to NO.


Assuntos
Artérias Cerebrais/enzimologia , Óxido Nítrico Sintase/genética , Óxido Nítrico Sintase/metabolismo , Adulto , Ácido Aspártico/genética , Artérias Cerebrais/fisiopatologia , GMP Cíclico/biossíntese , Endotélio Vascular/metabolismo , Endotélio Vascular/fisiopatologia , Epilepsia/enzimologia , Epilepsia/fisiopatologia , Feminino , Expressão Gênica , Humanos , Masculino , Óxido Nítrico Sintase Tipo III , Mutação Puntual , Proteínas Recombinantes/metabolismo , Serina/genética , Vasodilatação
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA