Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Arterioscler Thromb Vasc Biol ; 42(7): e217-e227, 2022 07.
Artigo em Inglês | MEDLINE | ID: mdl-35652755

RESUMO

BACKGROUND: An activated, proinflammatory endothelium is a key feature in the development of complications of obesity and type 2 diabetes and can be caused by insulin resistance in endothelial cells. METHODS: We analyzed primary human endothelial cells by RNA sequencing to discover novel insulin-regulated genes and used endothelial cell culture and animal models to characterize signaling through CXCR4 (C-X-C motif chemokine receptor 4) in endothelial cells. RESULTS: CXCR4 was one of the genes most potently regulated by insulin, and this was mediated by PI3K (phosphatidylinositol 3-kinase), likely through FoxO1, which bound to the CXCR4 promoter. CXCR4 mRNA in CD31+ cells was 77% higher in mice with diet-induced obesity compared with lean controls and 37% higher in db/db mice than db/+ controls, consistent with upregulation of CXCR4 in endothelial cell insulin resistance. SDF-1 (stromal cell-derived factor-1)-the ligand for CXCR4-increased leukocyte adhesion to cultured endothelial cells. This effect was lost after deletion of CXCR4 by gene editing while 80% of the increase was prevented by treatment of endothelial cells with insulin. In vivo microscopy of mesenteric venules showed an increase in leukocyte rolling after intravenous injection of SDF-1, but most of this response was prevented in transgenic mice with endothelial overexpression of IRS-1 (insulin receptor substrate-1). CONCLUSIONS: Endothelial cell insulin signaling limits leukocyte/endothelial cell interaction induced by SDF-1 through downregulation of CXCR4. Improving insulin signaling in endothelial cells or inhibiting endothelial CXCR4 may reduce immune cell recruitment to the vascular wall or tissue parenchyma in insulin resistance and thereby help prevent several vascular complications.


Assuntos
Diabetes Mellitus Tipo 2 , Resistência à Insulina , Receptores CXCR4/metabolismo , Animais , Quimiocina CXCL12/genética , Quimiocina CXCL12/metabolismo , Diabetes Mellitus Tipo 2/genética , Diabetes Mellitus Tipo 2/metabolismo , Células Endoteliais/metabolismo , Endotélio/metabolismo , Insulina , Leucócitos/metabolismo , Camundongos , Obesidade/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Receptores CXCR4/genética
2.
Sci Rep ; 11(1): 5983, 2021 03 16.
Artigo em Inglês | MEDLINE | ID: mdl-33727615

RESUMO

The potentially fatal cardiovascular effects of hypoglycaemia are not well understood and large animal models of the counter-regulatory responses and cardiovascular consequences of insulin-induced hypoglycaemia are needed to understand the mechanisms in humans. The aim of this study was to develop a human-like minipig model of hypoglycaemia including healthy and diabetic pigs to investigate endocrine, electrocardiographic and platelet effects. Hypoglycaemia was induced using a hyperinsulinaemic, hypoglycaemic clamp and an insulin bolus protocol. Plasma glucose, glucagon, C-peptide, insulin, epinephrine and platelet aggregation responses were measured before, during and after hypoglycaemia. Continuous electrocardiographic recordings were obtained. Hypoglycaemia at a plasma glucose concentration of 0.8-1.0 mM in the clamp induced 25-fold increase in epinephrine and sixfold and threefold increase in glucagon for healthy and diabetic pigs, respectively. The hypoglycaemic clamp induced QTc-interval prolongation and increase in cardiac arrhythmias. In the bolus approach, the non-diabetic group reached plasma glucose target of 1.5 mM and QTc-interval was prolonged after insulin injection, but before glucose nadir. The diabetic group did not reach hypoglycaemic target, but still demonstrated QTc-interval prolongation. These results demonstrate effects of hyperinsulinaemic hypoglycaemia closely resembling human physiology, indicating the minipig as a translational animal model of counter-regulatory endocrine and myocardial effects of hypoglycaemia.


Assuntos
Hiperinsulinismo Congênito/complicações , Hiperinsulinismo Congênito/veterinária , Cardiopatias/diagnóstico , Cardiopatias/etiologia , Doenças dos Suínos/sangue , Animais , Biomarcadores/sangue , Glicemia , Plaquetas/metabolismo , Plaquetas/ultraestrutura , Gerenciamento Clínico , Modelos Animais de Doenças , Suscetibilidade a Doenças , Eletrocardiografia , Sistema Endócrino/metabolismo , Agregação Plaquetária , Testes de Função Plaquetária , Suínos , Doenças dos Suínos/diagnóstico , Porco Miniatura , Avaliação de Sintomas
3.
J Endocrinol ; 248(3): 289-301, 2021 03.
Artigo em Inglês | MEDLINE | ID: mdl-33449915

RESUMO

The glucagon-like peptide-1 receptor (GLP1R) is expressed in the renal vasculature and known to be downregulated under hypertensive conditions in rats and humans. However, little is known about the regulation in other types of renal pathology involving vascular changes. This study investigates the expression of the GLP1R in renal vasculature after glomerular injury in the nephrotoxic nephritis mouse model, high cholesterol, and atherosclerosis in the Ldlr-/- mouse on Western diet, and ex vivo injury in an organ culture model. The immunohistochemical signal of the GLP1R was significantly decreased in arteries from mice with nephrotoxic nephritis after 42 days compared to 7 days and saline control (P < 0.05). Histological evaluation of kidneys from Ldlr-/- mice on Western diet showed a decreased GLP1R specific immunohistochemical signal (P < 0.05). The dilatory response to liraglutide was decreased in Western diet fed Ldlr-/- mice compared to C57Bl/6J controls (P < 0.05). Organ culture significantly decreased the immunohistochemical signal of the GLP1R (P <0.05) and the expression of Glp1r mRNA (P < 0.005) compared to fresh. Organ cultured vessels showed vascular smooth muscle cell remodelling as Acta2 expression was decreased (P < 0.005) and Ednrb was increased (P < 0.05). In conclusion, nephrotoxic nephritis and hypercholesterolaemia led to decreased GLP1R specific immunohistochemical signal. Ex vivo vascular injury in the organ culture model leads to a decrease in expression of GLP1R expressionand contractile VSMC specific markers and increase in expression of dedifferentiation markers suggestive of an inverse relationship between phenotypic switch of the VSMC and the expression of the GLP1R; however, the causal relationship remains elusive.


Assuntos
Receptor do Peptídeo Semelhante ao Glucagon 1/metabolismo , Artéria Renal/metabolismo , Doenças Vasculares/metabolismo , Animais , Feminino , Camundongos , Nefrite/metabolismo , Técnicas de Cultura de Órgãos
4.
Basic Clin Pharmacol Toxicol ; 128(1): 103-114, 2021 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-32896073

RESUMO

Recent clinical intervention studies have shown that the GLP1 analogue liraglutide lowers cardiovascular risk, but the underlying mechanism has not yet been fully elucidated. This study investigated the effects of liraglutide on endothelial function in the Ldlr-/- mouse model. Mice (n = 12/group) were fed Western diet (WD) or chow for 12 weeks followed by 4 weeks of treatment with liraglutide (1 mg/kg/day) or vehicle subcutaneously. Weight loss, blood lipid content, plaque burden, vasomotor function of the aorta and gene expression pattern in aorta and brachiocephalic artery were monitored. Liraglutide treatment significantly induced weight loss (P < .0001), decreased blood triglycerides (P < .0001) and total cholesterol (P < .0001) in WD-fed mice but did not decrease plaque burden. Liraglutide also improved endothelium-mediated dilation of the distal thoracis aorta (P = .0067), but it did not affect phenylephrine or sodium nitroprusside responses. Fluidigm analyses of 96 genes showed significantly altered expression of seven genes related to inflammation, vascular smooth muscle cells and extracellular matrix composition in liraglutide-treated animals. We conclude that treatment with liraglutide decreased endothelial dysfunction and that this could be linked to decreased inflammation or regulation of vascular remodelling.


Assuntos
Anti-Inflamatórios/farmacologia , Aorta Torácica/efeitos dos fármacos , Aterosclerose/tratamento farmacológico , Células Endoteliais/efeitos dos fármacos , Endotélio Vascular/efeitos dos fármacos , Inflamação/prevenção & controle , Liraglutida/farmacologia , Remodelação Vascular/efeitos dos fármacos , Animais , Aorta Torácica/metabolismo , Aorta Torácica/patologia , Aorta Torácica/fisiopatologia , Aterosclerose/metabolismo , Aterosclerose/patologia , Aterosclerose/fisiopatologia , Modelos Animais de Doenças , Células Endoteliais/metabolismo , Células Endoteliais/patologia , Endotélio Vascular/metabolismo , Endotélio Vascular/patologia , Endotélio Vascular/fisiopatologia , Regulação da Expressão Gênica , Inflamação/metabolismo , Inflamação/patologia , Inflamação/fisiopatologia , Masculino , Camundongos Knockout , Placa Aterosclerótica , Receptores de LDL/genética , Receptores de LDL/metabolismo , Transdução de Sinais , Vasodilatação/efeitos dos fármacos
5.
J Cereb Blood Flow Metab ; 36(7): 1195-201, 2016 07.
Artigo em Inglês | MEDLINE | ID: mdl-27052838

RESUMO

Transient global cerebral ischemia is often followed by delayed disturbances of cerebral blood flow, contributing to neuronal injury. The pathophysiological processes underlying such disturbances are incompletely understood. Here, using an established model of transient global cerebral ischemia, we identify dramatically impaired neurovascular coupling following ischemia. This impairment results from the loss of functional inward rectifier potassium (KIR) channels in the smooth muscle of parenchymal arterioles. Therapeutic strategies aimed at protecting or restoring cerebrovascular KIR channel function may therefore improve outcomes following ischemia.


Assuntos
Arteríolas/metabolismo , Ataque Isquêmico Transitório/fisiopatologia , Músculo Liso Vascular/metabolismo , Acoplamento Neurovascular/fisiologia , Tecido Parenquimatoso/irrigação sanguínea , Canais de Potássio Corretores do Fluxo de Internalização/metabolismo , Animais , Arteríolas/fisiopatologia , Circulação Cerebrovascular/fisiologia , Endotélio Vascular , Ataque Isquêmico Transitório/metabolismo , Masculino , Músculo Liso Vascular/fisiopatologia , Ratos Sprague-Dawley
6.
BMC Genomics ; 16: 119, 2015 Feb 22.
Artigo em Inglês | MEDLINE | ID: mdl-25766280

RESUMO

BACKGROUND: microRNAs (miRNAs) are important regulators of translation and have been implicated in the pathogenesis of a number of cardiovascular diseases, including stroke, and suggested as possible prognostic biomarkers. Our aim was to identify miRNAs that are differentially regulated in cerebral arteries after subarachnoid hemorrhage (SAH), using a rat injection model of SAH and a qPCR-based screen of 728 rat miRNAs. Additionally, serum was analyzed for a possible spill-over to the circulation of regulated miRNAs from the vessel walls. RESULTS: We identified 482 different miRNAs expressed in cerebral arteries post-SAH. Two miRNAs, miR-30a and miR-143, were significantly upregulated in cerebral arteries after SAH when compared to sham-operated animals. However, none of these exhibited significantly altered serum levels after SAH versus post-sham surgery. The most robust upregulation was seen for miR-143, which has several predicted targets and is a strong regulator of vascular morphology. We hypothesize that miR-30a and miR-143 may play a role in the vascular wall changes seen after SAH. CONCLUSIONS: We report that miR-30a and miR-143 in the cerebral arteries show significant changes over time after SAH, but do not differ from sham-operated rats at 24 h post-SAH. Although this finding suggests interesting novel possible mechanisms involved in post-SAH cerebrovascular changes, the lack of regulation of these miRNAs in serum excludes their use as blood-borne biomarkers for cerebrovascular changes following SAH.


Assuntos
Artérias Cerebrais/metabolismo , MicroRNAs/sangue , Hemorragia Subaracnóidea/genética , Animais , Artérias Cerebrais/patologia , Artérias Cerebrais/cirurgia , Modelos Animais de Doenças , Regulação da Expressão Gênica , Humanos , Ratos , Hemorragia Subaracnóidea/sangue , Hemorragia Subaracnóidea/patologia
7.
J Neuroinflammation ; 11: 207, 2014 Dec 10.
Artigo em Inglês | MEDLINE | ID: mdl-25498987

RESUMO

BACKGROUND: It has been suggested that transcriptional upregulation of cerebral artery contractile endothelin (ETB) and 5-hydroxytryptamine (5-HT1B) receptors play an important role in the development of late cerebral ischemia and increased vasoconstriction after subarachnoid hemorrhage (SAH). We tested the hypothesis that inhibition of calcium calmodulin-dependent protein kinase II (CaMKII) may reduce cerebral vasoconstriction mediated by endothelin and serotonin receptors and improve neurological outcome after experimental SAH. METHODS: SAH was induced in adult rats by injection of 250 µL autologous blood into the basal cisterns. The CaMKII activity in cerebral vessels was studied by Western blot and immunohistochemistry. The vasomotor responses of middle cerebral and basilar arteries were measured in a sensitive myograph system. The functional outcome was examined by the rotating pole test 2 and 3 days after SAH. RESULTS: SAH induced a rapid early increase in phosphorylated CaMKII protein at 1 h that was attenuated by cisternal administration of the CaMKII inhibitor KN93 (0.501 µg/kg) 45 min prior and immediately after SAH as evaluated by Western blot. Application of KN93 at 1 h and every 12 h post-SAH significantly reduced vascular CaMKII immunoreactivity at 72 h. In addition, contractile responses of cerebral arteries to endothelin-1 (ET-1) and 5-hydroxycarboxamide (5-CT) were increased at this time-point. KN93 treatment significantly attenuated the contraction induced by ET-1 and 5-CT. Importantly, treatment with the CaMKII inhibitor prevented SAH-induced deficits in neurological function, as evaluated by the rotating pole test, and similar sensorimotor scores were seen in sham-operated animals. CONCLUSIONS: The present study has shown that SAH is associated with increased contractile responses to ET-1 and 5-CT in cerebral arteries and enhanced early activation of CaMKII. Treatment with the CaMKII inhibitor KN93 attenuated the contractile responses and prevented impaired sensorimotor function after SAH.


Assuntos
Proteína Quinase Tipo 2 Dependente de Cálcio-Calmodulina/antagonistas & inibidores , Endotelinas/metabolismo , Receptores de Serotonina/metabolismo , Hemorragia Subaracnóidea/complicações , Vasoconstrição/efeitos dos fármacos , Animais , Western Blotting , Artérias Cerebrais/efeitos dos fármacos , Artérias Cerebrais/metabolismo , Circulação Cerebrovascular/efeitos dos fármacos , Modelos Animais de Doenças , Inibidores Enzimáticos/farmacologia , Imuno-Histoquímica , Masculino , Ratos , Ratos Sprague-Dawley , Hemorragia Subaracnóidea/metabolismo , Hemorragia Subaracnóidea/fisiopatologia
8.
PLoS One ; 9(3): e92417, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24642693

RESUMO

BACKGROUND: Global cerebral ischemia following cardiac arrest is associated with increased cerebral vasoconstriction and decreased cerebral blood flow, contributing to delayed neuronal cell death and neurological detriments in affected patients. We hypothesize that upregulation of contractile ETB and 5-HT1B receptors, previously demonstrated in cerebral arteries after experimental global ischemia, are a key mechanism behind insufficient perfusion of the post-ischemic brain, proposing blockade of this receptor upregulation as a novel target for prevention of cerebral hypoperfusion and delayed neuronal cell death after global cerebral ischemia. The aim was to characterize the time-course of receptor upregulation and associated neuronal damage after global ischemia and investigate whether treatment with the MEK1/2 inhibitor U0126 can prevent cerebrovascular receptor upregulation and thereby improve functional outcome after global cerebral ischemia. Incomplete global cerebral ischemia was induced in Wistar rats and the time-course of enhanced contractile responses and the effect of U0126 in cerebral arteries were studied by wire myography and the neuronal cell death by TUNEL. The expression of ETB and 5-HT1B receptors was determined by immunofluorescence. RESULTS: Enhanced vasoconstriction peaked in fore- and midbrain arteries 3 days after ischemia. Neuronal cell death appeared initially in the hippocampus 3 days after ischemia and gradually increased until 7 days post-ischemia. Treatment with U0126 normalised cerebrovascular ETB and 5-HT1B receptor expression and contractile function, reduced hippocampal cell death and improved survival rate compared to vehicle treated animals. CONCLUSIONS: Excessive cerebrovascular expression of contractile ETB and 5-HT1B receptors is a delayed response to global cerebral ischemia peaking 3 days after the insult, which likely contributes to the development of delayed neuronal damage. The enhanced cerebrovascular contractility can be prevented by treatment with the MEK1/2 inhibitor U0126, diminishes neuronal damage and improves survival rate, suggesting MEK1/2 inhibition as a novel strategy for early treatment of neurological consequences following global cerebral ischemia.


Assuntos
Isquemia Encefálica/tratamento farmacológico , Butadienos/farmacologia , Hipóxia Encefálica/prevenção & controle , Nitrilas/farmacologia , Receptor de Endotelina B/metabolismo , Receptor 5-HT1B de Serotonina/metabolismo , Animais , Isquemia Encefálica/patologia , Butadienos/uso terapêutico , Circulação Cerebrovascular/efeitos dos fármacos , Avaliação Pré-Clínica de Medicamentos , MAP Quinase Quinase 1/antagonistas & inibidores , MAP Quinase Quinase 1/metabolismo , MAP Quinase Quinase 2/antagonistas & inibidores , MAP Quinase Quinase 2/metabolismo , Nitrilas/uso terapêutico , Ratos , Ratos Wistar , Receptor de Endotelina B/genética , Receptor 5-HT1B de Serotonina/genética , Resultado do Tratamento , Regulação para Cima/efeitos dos fármacos , Vasoconstrição/efeitos dos fármacos
9.
J Cereb Blood Flow Metab ; 33(8): 1259-69, 2013 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-23715060

RESUMO

After subarachnoid hemorrhage (SAH), pathologic changes in cerebral arteries contribute to delayed cerebral ischemia and poor outcome. We hypothesize such changes are triggered by early intracellular signals, targeting of which may prevent SAH-induced vasculopathy. We performed an unbiased quantitative analysis of early SAH-induced phosphorylations in cerebral arteries and evaluated identified signaling components as targets for prevention of delayed vasculopathy and ischemia. Labeled phosphopeptides from rat cerebral arteries were quantified by high-resolution tandem mass spectrometry. Selected SAH-induced phosphorylations were validated by immunoblotting and monitored over a 24-hour time course post SAH. Moreover, inhibition of key phosphoproteins was performed. Major SAH-induced phosphorylations were observed on focal adhesion complexes, extracellular regulated kinase 1/2 (ERK1/2), calcium calmodulin-dependent kinase II, signal transducer and activator of transcription (STAT3) and c-Jun, the latter two downstream of ERK1/2. Inhibition of ERK1/2 6-hour post SAH prevented increases in cerebrovascular constrictor receptors, matrix metalloprotease-9, wall thickness, and improved neurologic outcome. STAT3 inhibition partially mimicked these effects. The study shows that quantitative mass spectrometry is a strong approach to study in vivo vascular signaling. Moreover, it shows that targeting of ERK1/2 prevents delayed pathologic changes in cerebral arteries and improves outcome, and identifies SAH-induced signaling components downstream and upstream of ERK1/2.


Assuntos
Artérias Cerebrais/fisiopatologia , Fosfoproteínas/genética , Proteoma/genética , Transdução de Sinais/fisiologia , Hemorragia Subaracnóidea/fisiopatologia , Animais , Western Blotting , Proteína Quinase Tipo 2 Dependente de Cálcio-Calmodulina/metabolismo , Cromatografia Líquida , Imuno-Histoquímica , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Sistema de Sinalização das MAP Quinases/fisiologia , Masculino , Metaloproteinase 9 da Matriz/genética , Metaloproteinase 9 da Matriz/metabolismo , Neuropeptídeos/metabolismo , Fosfoproteínas/fisiologia , Fosforilação , Proteoma/fisiologia , Desempenho Psicomotor/fisiologia , Ratos , Ratos Sprague-Dawley , Fator de Transcrição STAT3/genética , Fator de Transcrição STAT3/fisiologia , Espectrometria de Massas por Ionização por Electrospray
10.
BMC Neurosci ; 14: 34, 2013 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-23496889

RESUMO

BACKGROUND: Upregulation of vasoconstrictor receptors in cerebral arteries, including endothelin B (ETB) and 5-hydroxytryptamine 1B (5-HT(1B)) receptors, has been suggested to contribute to delayed cerebral ischemia, a feared complication after subarachnoid hemorrhage (SAH). This receptor upregulation has been shown to be mediated by intracellular signalling via the mitogen activated protein kinase kinase (MEK1/2)--extracellular regulated kinase 1/2 (ERK1/2) pathway. However, it is not known what event(s) that trigger MEK-ERK1/2 activation and vasoconstrictor receptor upregulation after SAH.We hypothesise that the drop in cerebral blood flow (CBF) and wall tension experienced by cerebral arteries in acute SAH is a key triggering event. We here investigate the importance of the duration of this acute CBF drop in a rat SAH model in which a fixed amount of blood is injected into the prechiasmatic cistern either at a high rate resulting in a short acute CBF drop or at a slower rate resulting in a prolonged acute CBF drop. RESULTS: We demonstrate that the duration of the acute CBF drop is determining for a) degree of early ERK1/2 activation in cerebral arteries, b) delayed upregulation of vasoconstrictor receptors in cerebral arteries and c) delayed CBF reduction, neurological deficits and mortality. Moreover, treatment with an inhibitor of MEK-ERK1/2 signalling during an early time window from 6 to 24 h after SAH was sufficient to completely prevent delayed vasoconstrictor receptor upregulation and improve neurological outcome several days after the SAH. CONCLUSIONS: Our findings suggest a series of events where 1) the acute CBF drop triggers early MEK-ERK1/2 activation, which 2) triggers the transcriptional upregulation of vasoconstrictor receptors in cerebral arteries during the following days, where 3) the resulting enhanced cerebrovascular contractility contribute to delayed cerebral ischemia.


Assuntos
Isquemia Encefálica/etiologia , Isquemia Encefálica/metabolismo , Receptor de Endotelina B/metabolismo , Receptor 5-HT1B de Serotonina/metabolismo , Hemorragia Subaracnóidea/complicações , Regulação para Cima/fisiologia , Análise de Variância , Animais , Antipirina/análogos & derivados , Área Sob a Curva , Pressão Sanguínea/fisiologia , Isquemia Encefálica/mortalidade , Butadienos/farmacologia , Isótopos de Carbono , Artérias Cerebrais/metabolismo , Circulação Cerebrovascular/fisiologia , Modelos Animais de Doenças , Inibidores Enzimáticos/farmacologia , Fluxometria por Laser-Doppler , Sistema de Sinalização das MAP Quinases/fisiologia , Masculino , Atividade Motora/fisiologia , Doenças do Sistema Nervoso/etiologia , Doenças do Sistema Nervoso/prevenção & controle , Nitrilas/farmacologia , Ratos , Ratos Sprague-Dawley , Receptor de Endotelina B/genética , Receptor 5-HT1B de Serotonina/genética , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/fisiologia , Regulação para Cima/efeitos dos fármacos
11.
J Neuroinflammation ; 9: 274, 2012 Dec 21.
Artigo em Inglês | MEDLINE | ID: mdl-23259581

RESUMO

BACKGROUND: Subarachnoid hemorrhage (SAH) is associated with high morbidity and mortality. It is suggested that the associated inflammation is mediated through activation of the mitogen-activated protein kinase (MAPK) pathway which plays a crucial role in the pathogenesis of delayed cerebral ischemia after SAH. The aim of this study was first to investigate the timecourse of altered expression of proinflammatory cytokines and matrix metalloproteinase in the cerebral arteries walls following SAH. Secondly, we investigated whether administration of a specific mitogen-activated protein kinase kinase (MEK)1/2 inhibitor, U0126, given at 6 h after SAH prevents activation of the MEK/extracellular signal-regulated kinase 1/2 pathway and the upregulation of cerebrovascular inflammatory mediators and improves neurological function. METHODS: SAH was induced in rats by injection of 250 µl of autologous blood into basal cisterns. U0126 was given intracisternally using two treatment regimens: (A) treatments at 6, 12, 24 and 36 h after SAH and experiments terminated at 48 h after SAH, or (B) treatments at 6, 12, and 24 h after SAH and terminated at 72 h after SAH. Cerebral arteries were harvested and interleukin (IL)-6, IL-1ß, tumor necrosis factor α (TNF)α, matrix metalloproteinase (MMP)-9 and phosphorylated ERK1/2 (pERK1/2) levels investigated by immunohistochemistry. Early activation of pERK1/2 was measured by western blot. Functional neurological outcome after SAH was also analyzed. RESULTS: Expression levels of IL-1ß, IL-6, MMP-9 and pERK1/2 proteins were elevated over time with an early increase at around 6 h and a late peak at 48 to 72 h post-SAH in cerebral arteries. Enhanced expression of TNFα in cerebral arteries started at 24 h and increased until 96 h. In addition, SAH induced sensorimotor and spontaneous behavior deficits in the animals. Treatment with U0126 starting at 6 h after SAH prevented activation of MEK-ERK1/2 signaling. Further, U0126 significantly decreased the upregulation of inflammation proteins at 48 and 72 h following SAH and improved neurological function. We found no differences between treatment regimens A and B. CONCLUSIONS: These results show that SAH induces early activation of the MEK-ERK1/2 pathway in cerebral artery walls, which is associated with upregulation of proinflammatory cytokines and MMP-9. Inhibition of the MEK-ERK1/2 pathway by U0126 starting at 6 h post-SAH prevented upregulation of cytokines and MMP-9 in cerebral vessels, and improved neurological outcome.


Assuntos
Artérias Cerebrais/enzimologia , Citocinas/metabolismo , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Transdução de Sinais/fisiologia , Hemorragia Subaracnóidea/patologia , Animais , Astrócitos/efeitos dos fármacos , Astrócitos/enzimologia , Transfusão de Sangue Autóloga/efeitos adversos , Butadienos/uso terapêutico , Artérias Cerebrais/patologia , Citocinas/genética , Modelos Animais de Doenças , Inibidores Enzimáticos/uso terapêutico , Comportamento Exploratório/efeitos dos fármacos , Proteína Glial Fibrilar Ácida/metabolismo , Masculino , Metaloproteinase 9 da Matriz/genética , Metaloproteinase 9 da Matriz/metabolismo , Atividade Motora/efeitos dos fármacos , Doenças do Sistema Nervoso/tratamento farmacológico , Doenças do Sistema Nervoso/etiologia , Nitrilas/uso terapêutico , Ratos , Ratos Sprague-Dawley , Transdução de Sinais/efeitos dos fármacos , Estatísticas não Paramétricas , Hemorragia Subaracnóidea/complicações , Hemorragia Subaracnóidea/tratamento farmacológico , Hemorragia Subaracnóidea/metabolismo , Fatores de Tempo , Fator de Necrose Tumoral alfa/metabolismo
12.
PLoS One ; 7(7): e41852, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22848635

RESUMO

BACKGROUND: Global ischemic stroke is one of the most prominent consequences of cardiac arrest, since the diminished blood flow to the brain results in cell damage and sometimes permanently impaired neurological function. The post-arrest period is often characterised by cerebral hypoperfusion due to subacute hemodynamic disturbances, the pathophysiology of which are poorly understood. In two other types of stroke, focal ischemic stroke and subarachnoid hemorrhage, it has earlier been demonstrated that the expression of certain vasoconstrictor receptors is increased in cerebral arteries several days after the insult, a phenomenon that leads to increased contraction of cerebral arteries, reduced perfusion of the affected area and worsened ischemic damage. Based on these findings, the aim of the present study was to investigate if transient global cerebral ischemia is associated with upregulation of vasoconstrictive endothelin and 5-hydroxytryptamine receptors in cerebral arteries. Experimental transient forebrain ischemia of varying durations was induced in male wistar rats, followed by reperfusion for 48 hours. Neurological function was assessed daily by three different tests and cerebrovascular expression and contractile function of endothelin and 5-hydroxytryptamine receptors were evaluated by wire myography, immunohistochemistry and western blotting. RESULTS: Transient forebrain ischemia induced neurological deficits as well as functional upregulation of vasoconstrictive ET(B) and 5-HT(1B) receptors in cerebral arteries supplying mid- and forebrain regions. No receptor upregulation was seen in arteries supplying the hindbrain. Immunohistochemical stainings and western blotting demonstrated expressional upregulation of these receptor subtypes in the mid- and forebrain arteries and confirmed that the receptors were located in the smooth muscle layer of the cerebral arteries. CONCLUSIONS: This study reveals a new pathophysiological aspect of global ischemic stroke, namely expressional upregulation of vasoconstrictor receptors in cerebral arteries two days after the insult, which might contribute to cerebral hypoperfusion and delayed neuronal damage after cardiac arrest.


Assuntos
Regulação da Expressão Gênica , Ataque Isquêmico Transitório/metabolismo , Prosencéfalo/irrigação sanguínea , Prosencéfalo/metabolismo , Receptor de Endotelina B/metabolismo , Receptor 5-HT1B de Serotonina/metabolismo , Animais , Artérias Carótidas/efeitos dos fármacos , Artérias Carótidas/metabolismo , Artérias Carótidas/fisiopatologia , Endotelina-1/farmacologia , Regulação da Expressão Gênica/efeitos dos fármacos , Ataque Isquêmico Transitório/fisiopatologia , Masculino , Prosencéfalo/efeitos dos fármacos , Ratos , Ratos Wistar , Serotonina/análogos & derivados , Serotonina/farmacologia , Vasoconstrição/efeitos dos fármacos , Venenos de Víboras/farmacologia
13.
Exp Brain Res ; 219(4): 507-20, 2012 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-22585122

RESUMO

Cerebral arteries subjected to different types of experimental stroke upregulate their expression of certain G-protein-coupled vasoconstrictor receptors, a phenomenon that worsens the ischemic brain damage. Upregulation of contractile endothelin B (ET(B)) and 5-hydroxytryptamine 1B (5-HT(1B)) receptors has been demonstrated after subarachnoid hemorrhage and global ischemic stroke, but the situation is less clear after focal ischemic stroke. Changes in smooth muscle calcium handling have been implicated in different vascular diseases but have not hitherto been investigated in cerebral arteries after stroke. Here, we evaluate changes of ET(B) and 5-HT(1B) receptors, intracellular calcium levels, and calcium channel expression in rat middle cerebral artery (MCA) after focal cerebral ischemia and in vitro organ culture, a proposed model of vasoconstrictor receptor changes after stroke. Rats were subjected to 2 h MCA occlusion followed by reperfusion for 1 or 24 h. Alternatively, MCAs from naïve rats were cultured for 1 or 24 h. ET(B) and 5-HT(1B) receptor-mediated contractions were evaluated by wire myography. Receptor and channel expressions were measured by real-time PCR and immunohistochemistry. Intracellular calcium was measured by FURA-2. Expression and contractile functions of ET(B) and 5-HT(1B) receptors were strongly upregulated and slightly downregulated, respectively, 24 h after experimental stroke or organ culture. ET(B) receptor-mediated contraction was mediated by calcium from intracellular and extracellular sources, whereas 5-HT(1B) receptor-mediated contraction was solely dependent on extracellular calcium. Organ culture and stroke increased basal intracellular calcium levels in MCA smooth muscle cells and decreased the expression of inositol triphosphate receptor and transient receptor potential canonical calcium channels, but not voltage-operated calcium channels.


Assuntos
Cálcio/metabolismo , Artérias Cerebrais/metabolismo , Receptor de Endotelina B/biossíntese , Receptor 5-HT1B de Serotonina/biossíntese , Acidente Vascular Cerebral/metabolismo , Vasoconstrição/fisiologia , Animais , Artérias Cerebrais/efeitos dos fármacos , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Líquido Intracelular/efeitos dos fármacos , Líquido Intracelular/metabolismo , Masculino , Músculo Liso Vascular/efeitos dos fármacos , Músculo Liso Vascular/metabolismo , Técnicas de Cultura de Órgãos , Ratos , Ratos Wistar , Vasoconstrição/efeitos dos fármacos , Venenos de Víboras/farmacologia
14.
J Cereb Blood Flow Metab ; 31(7): 1554-71, 2011 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-21559027

RESUMO

Cerebral ischemia remains a major cause of morbidity and mortality with little advancement in subacute treatment options. This review aims to cover and discuss novel insight obtained during the last decade into plastic changes in the vasoconstrictor receptor profiles of cerebral arteries and microvessels that takes place after different types of stroke. Receptors like the endothelin type B, angiotensin type 1, and 5-hydroxytryptamine type 1B/1D receptors are upregulated in the smooth muscle layer of cerebral arteries after different types of ischemic stroke as well as after subarachnoid hemorrhage, yielding rather dramatic changes in the contractility of the vessels. Some of the signal transduction processes mediating this receptor upregulation have been elucidated. In particular the extracellular regulated kinase 1/2 pathway, which is activated early in the process, has proven to be a promising therapeutic target for prevention of vasoconstrictor receptor upregulation after stroke. Together, those findings provide new perspectives on the pathophysiology of ischemic stroke and point toward a novel way of reducing vasoconstriction, neuronal cell death, and thus neurologic deficits after stroke.


Assuntos
Encéfalo/irrigação sanguínea , Artérias Cerebrais/metabolismo , Transdução de Sinais , Acidente Vascular Cerebral/metabolismo , Vasoconstrição , Animais , Encéfalo/efeitos dos fármacos , Encéfalo/metabolismo , Encéfalo/fisiopatologia , Artérias Cerebrais/efeitos dos fármacos , Artérias Cerebrais/fisiopatologia , Humanos , Transdução de Sinais/efeitos dos fármacos , Acidente Vascular Cerebral/tratamento farmacológico , Acidente Vascular Cerebral/fisiopatologia , Vasoconstrição/efeitos dos fármacos
15.
J Neurosurg ; 114(4): 1143-53, 2011 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-20597604

RESUMO

OBJECT: Delayed cerebral ischemia after subarachnoid hemorrhage (SAH) remains a major cause of death and disability. It has been hypothesized that cerebrovascular upregulation of vasoconstrictor receptors is a key step in the development of delayed cerebral ischemia. Upregulation of endothelin-B (ET(B)) and 5-hydroxytryptamine 1B (5-HT(1B)) receptors has been demonstrated in cerebral artery smooth muscles in the delayed ischemic phase after experimental SAH, and intracellular signaling via the mitogen-activated protein kinase kinase (MEK)-extracellular signal-regulated kinase 1/2 pathway has been shown to be involved in this upregulation. The aim in the present study was to determine whether treatment with the MEK1/2 inhibitor U0126 can prevent cerebrovascular receptor upregulation and improve functional outcome after experimental SAH in rats. METHODS: Subarachnoid hemorrhage was induced in male Sprague-Dawley rats by the injection of 250 µl of autologous blood into the basal cisterns. Either U0126 or vehicle was intracisternally administered at 6, 12, 24, and 36 hours after SAH. Smooth muscle ET(B) and 5-HT(1B) receptor upregulation was studied in isolated cerebral artery segments through immunohistochemical and myographic studies of contractile responses to receptor-specific agonists. Gross sensorimotor function in the rats after SAH was assessed using a rotating pole test. RESULTS: Contractile concentration-response curves for middle cerebral artery (MCA) and basilar artery (BA) segments to endothelin-1 (ET-1) and 5-carboxamidotryptamine (5-CT) were shifted leftward for SAH-induced compared with shamoperated rats due to enhanced contractile responses to individual doses of the agonists (for example, contractile responses of the BA to 3 × 10(-10) M of ET-1 and 3 × 10(-7) M of 5-CT were 9.98 ± 5.01% and 16.75 ± 3.62% of the maximal contractile capacity, respectively, in sham-operated rats and 62.78 ± 9.9% and 45.44 ± 10.62%, respectively, in SAH-induced rats). In vivo treatment with 0.19 µg/kg U0126 normalized responses in the SAH-induced rats to levels in the sham-operated rats. Protein expression of ET(B) and 5-HT(1B) receptors in cerebrovascular smooth muscles from SAH-induced rats was increased to 175 ± 33.17% and 167.7 ± 24.74%, respectively, of the levels in sham-operated rats. Endothelin-B and 5-HT(1B) expression levels in U0126-treated SAH-induced rats were at the levels in sham-operated rats (101.9 ± 13.38% and 91.44 ± 16.75%, respectively). In a rotating pole test used to assess gross sensorimotor function on the 2nd day after surgery, sham-operated rats achieved an average score of 5.37 ± 0.23, SAH-induced rats scored 3.35 ± 0.67, and SAH-induced U0126-treated rats scored 5.00 ± 0.4. CONCLUSIONS: The authors demonstrated that experimental SAH induces upregulation of ET(B) and 5-HT(1B) receptors in cerebrovascular smooth muscles and that treatment with the MEK1/2 inhibitor U0126 abolishes this receptor upregulation. They also demonstrated that experimental SAH results in sensorimotor deficits as assessed by a rotating pole test. These deficits were alleviated by U0126 treatment, suggesting that cerebrovascular receptor upregulation is critical for the functional outcome of delayed cerebral ischemia. The authors suggest that inhibition of MEK1/2 may be a promising new SAH treatment strategy.


Assuntos
Butadienos/uso terapêutico , Antagonistas do Receptor de Endotelina B , MAP Quinase Quinase 1/antagonistas & inibidores , MAP Quinase Quinase 2/antagonistas & inibidores , Nitrilas/uso terapêutico , Inibidores de Proteínas Quinases/uso terapêutico , Receptor 5-HT1B de Serotonina/efeitos dos fármacos , Antagonistas da Serotonina/farmacologia , Hemorragia Subaracnóidea/tratamento farmacológico , Animais , Comportamento Animal/efeitos dos fármacos , Artérias Cerebrais/patologia , Imuno-Histoquímica , Masculino , Contração Muscular/efeitos dos fármacos , Músculo Liso Vascular/efeitos dos fármacos , Doenças do Sistema Nervoso/etiologia , Doenças do Sistema Nervoso/psicologia , Equilíbrio Postural/efeitos dos fármacos , Ratos , Ratos Sprague-Dawley , Serotonina/análogos & derivados , Serotonina/farmacologia , Agonistas do Receptor de Serotonina/farmacologia , Hemorragia Subaracnóidea/enzimologia , Hemorragia Subaracnóidea/metabolismo , Regulação para Cima/efeitos dos fármacos , Vasoconstrição/efeitos dos fármacos
16.
Cell Oncol ; 32(4): 259-74, 2010 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-20364069

RESUMO

The epidermal growth factor family of receptor tyrosine kinases (ErbBs) plays essential roles in tumorigenesis and cancer disease progression, and therefore has become an attractive target for structure-based drug design. ErbB receptors are activated by ligand-induced homo- and heterodimerization. Structural studies have revealed that ErbB receptor dimers are stabilized by receptor-receptor interactions, primarily mediated by a region in the second extracellular domain, termed the "dimerization arm". The present study is the first biological characterization of a peptide, termed Inherbin3, which constitutes part of the dimerization arm of ErbB3. Inherbin3 binds to the extracellular domains of all four ErbB receptors, with the lowest peptide binding affinity for ErbB4. Inherbin3 functions as an antagonist of epidermal growth factor (EGF)-ErbB1 signaling. We show that Inherbin3 inhibits EGF-induced ErbB1 phosphorylation, cell growth, and migration in two human tumor cell lines, A549 and HN5, expressing moderate and high ErbB1 levels, respectively. Furthermore, we show that Inherbin3 inhibits tumor growth in vivo and induces apoptosis in a tumor xenograft model employing the human non-small cell lung cancer cell line A549. The Inherbin3 peptide may be a useful tool for investigating the mechanisms of ErbB receptor homo- and heterodimerization. Moreover, the here described biological effects of Inherbin3 suggest that peptide-based targeting of ErbB receptor dimerization is a promising anti-cancer therapeutic strategy.


Assuntos
Carcinoma Pulmonar de Células não Pequenas/tratamento farmacológico , Receptores ErbB/antagonistas & inibidores , Neoplasias Pulmonares/tratamento farmacológico , Fragmentos de Peptídeos/farmacologia , Mucosa Respiratória/efeitos dos fármacos , Animais , Apoptose/efeitos dos fármacos , Carcinoma Pulmonar de Células não Pequenas/metabolismo , Carcinoma Pulmonar de Células não Pequenas/patologia , Processos de Crescimento Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Movimento Celular/efeitos dos fármacos , Receptores ErbB/biossíntese , Receptores ErbB/genética , Feminino , Humanos , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/patologia , Camundongos , Camundongos SCID , Transplante de Neoplasias , Fragmentos de Peptídeos/química , Agregação de Receptores/efeitos dos fármacos , Receptor ErbB-3/química , Mucosa Respiratória/metabolismo , Mucosa Respiratória/patologia , Carga Tumoral/efeitos dos fármacos
20.
J Neurosci Res ; 86(4): 727-43, 2008 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-17975827

RESUMO

The neural cell adhesion molecule (NCAM) plays a crucial role in neuronal development, synaptic plasticity, and regeneration. NCAM works as "smart glue" that not only mediates cell-cell adhesion but also induces activation of a complex network of intracellular signaling cascades on homophilic or heterophilic binding. Stimulation of NCAM by homophilic interactions induces neuronal differentiation through activation of a number of signaling molecules, including the fibroblast growth factor receptor, non-receptor kinases Fyn and focal adhesion kinase, growth-associated protein-43, the mitogen-activated protein kinase pathway, intracellular Ca(2+), and protein kinases A, C, and G. This review presents and discusses the current knowledge in the area of NCAM signaling with a focus on the events involved in NCAM-mediated neurite outgrowth.


Assuntos
Moléculas de Adesão de Célula Nervosa/fisiologia , Neuritos/metabolismo , Transdução de Sinais/fisiologia , Animais , Humanos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...