Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 63
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Prostaglandins Other Lipid Mediat ; 173: 106850, 2024 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-38735559

RESUMO

Midlife metabolic syndrome (MetS) is associated with cognitive impairment in late life. The mechanism of delayed MetS-related cognitive dysfunction (MetSCD) is not clear, but it has been linked to systemic inflammation and chronic cerebral microangiopathy. Currently there is no treatment for late life MetSCD other than early risk factor modification. We investigated the effect of soluble epoxide hydrolase (sEH) inhibitor 4-[[trans-4-[[(tricyclo[3.3.1.13,7]dec-1-ylamino)carbonyl]amino]cyclohexyl]oxy]-benzoic acid (t-AUCB) on cognitive performance, cerebral blood flow (CBF), and central and peripheral inflammation in the high-fat diet (HFD) model of MetS in mice. At 6 weeks of age, male mice were randomly assigned to receive either HFD or standard chow (STD) for 6 months. Mice received either t-AUCB or vehicle for 4 weeks. Cognitive performance was evaluated, followed by CBF measurement using magnetic resonance imaging (MRI). At the end of the study, blood was collected for measurement of eicosanoids and inflammatory cytokines. The brains were then analyzed by immunohistochemistry for glial activation markers. The HFD caused a significant impairment in novel object recognition. Treatment with t-AUCB increased plasma levels of 14,15-EET, prevented this cognitive impairment and modified hippocampal glial activation and plasma cytokine levels, without affecting CBF in mice on HFD. In conclusion, sEH inhibition for four weeks prevents cognitive deficits in mice on chronic HFD by modulating inflammatory processes without affecting CBF.


Assuntos
Disfunção Cognitiva , Modelos Animais de Doenças , Epóxido Hidrolases , Inflamação , Síndrome Metabólica , Animais , Masculino , Camundongos , Benzoatos/farmacologia , Benzoatos/uso terapêutico , Circulação Cerebrovascular/efeitos dos fármacos , Disfunção Cognitiva/tratamento farmacológico , Disfunção Cognitiva/etiologia , Disfunção Cognitiva/patologia , Disfunção Cognitiva/metabolismo , Dieta Hiperlipídica/efeitos adversos , Inibidores Enzimáticos/farmacologia , Inibidores Enzimáticos/uso terapêutico , Epóxido Hidrolases/antagonistas & inibidores , Epóxido Hidrolases/metabolismo , Inflamação/tratamento farmacológico , Inflamação/patologia , Síndrome Metabólica/tratamento farmacológico , Síndrome Metabólica/complicações , Síndrome Metabólica/patologia , Camundongos Endogâmicos C57BL
2.
Stroke ; 54(2): 364-373, 2023 02.
Artigo em Inglês | MEDLINE | ID: mdl-36689578

RESUMO

BACKGROUND: Type 2 diabetes (DM2) exacerbates stroke injury, reduces efficacy of endovascular therapy, and worsens long-term functional outcome. Sex differences exist in stroke incidence, response to therapy, poststroke microvascular dysfunction, and functional recovery. In this study, we tested the hypotheses that poor outcome after stroke in the setting of DM2 is linked to impaired microvascular tissue reperfusion and that male and female DM2 mice exhibit different microvascular reperfusion response after transient middle cerebral artery occlusion (MCAO). METHODS: Transient MCAO was induced for 60 minutes using an intraluminal filament in young adult DM2 and nondiabetic control male and female mice. Capillary flux in deep cortical layers was assessed using optical coherence tomography-based optical microangiography (OMAG), and associated regional brain infarct size was evaluated by hematoxylin and eosin staining. RESULTS: Compared to baseline, MCAO reduced absolute capillary red blood cell flux by 84% at 24 hours post-MCAO in male DM2 (P<0.001) but not male control mice. When normalized to pre-MCAO baseline, red blood cell flux 24 hours after stroke was 64% lower in male DM2 mice than male nondiabetic controls (P<0.01). In females, MCAO decreased capillary flux by 48% at 24 hours post-MCAO compared with baseline in DM2 (P<0.05) but not in control mice. Red blood cell flux of female DM2 mice did not differ from that of nondiabetic controls either before or 24 hours after MCAO. Furthermore, normalized capillary flux 24 hours after MCAO failed to differ between female DM2 mice and nondiabetic controls. Concomitantly, male but not female DM2 mice experienced 25% larger infarct in caudate-putamen versus respective nondiabetic controls (P<0.05). CONCLUSIONS: DM2 impairs capillary perfusion and exacerbates ischemic deep brain injury in male but not female young adult mice. Premenopausal females appear to be protected against DM2-related capillary dysfunction and brain injury.


Assuntos
Lesões Encefálicas , Diabetes Mellitus Experimental , Diabetes Mellitus Tipo 2 , Traumatismo por Reperfusão , Acidente Vascular Cerebral , Ratos , Camundongos , Feminino , Animais , Masculino , Infarto da Artéria Cerebral Média , Ratos Wistar , Caracteres Sexuais , Reperfusão , Modelos Animais de Doenças , Artéria Cerebral Média
3.
Am J Physiol Cell Physiol ; 322(5): C1011-C1021, 2022 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-35385329

RESUMO

Arachidonic acid metabolites epoxyeicosatrienoates (EETs) and hydroxyeicosatetraenoates (HETEs) are important regulators of myocardial blood flow and coronary vascular resistance (CVR), but their mechanisms of action are not fully understood. We applied a chemoproteomics strategy using a clickable photoaffinity probe to identify G protein-coupled receptor 39 (GPR39) as a microvascular smooth muscle cell (mVSMC) receptor selective for two endogenous eicosanoids, 15-HETE and 14,15-EET, which act on the receptor to oppose each other's activity. The former increases mVSMC intracellular calcium via GPR39 and augments coronary microvascular resistance, and the latter inhibits these actions. Furthermore, we find that the efficacy of both ligands is potentiated by zinc acting as an allosteric modulator. Measurements of coronary perfusion pressure (CPP) in GPR39-null hearts using the Langendorff preparation indicate the receptor senses these eicosanoids to regulate microvascular tone. These results implicate GPR39 as an eicosanoid receptor and key regulator of myocardial tissue perfusion. Our findings will have a major impact on understanding the roles of eicosanoids in cardiovascular physiology and disease and provide an opportunity for the development of novel GPR39-targeting therapies for cardiovascular disease.


Assuntos
Sistema Enzimático do Citocromo P-450 , Eicosanoides , Ácido Araquidônico/metabolismo , Vasos Coronários/metabolismo , Sistema Enzimático do Citocromo P-450/metabolismo , Eicosanoides/análise , Eicosanoides/metabolismo , Eicosanoides/farmacologia , Resistência Vascular
4.
Behav Pharmacol ; 33(2&3): 63-89, 2022 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-33136615

RESUMO

Cannabis is one of the most frequently used psychoactive substances in the world. The most common route of administration for cannabis and cannabinoid constituents such as Δ-9-tetrahydrocannabinol (THC) and cannabidiol (CBD) is via smoking or vapor inhalation. Preclinical vapor models have been developed, although the vaporization devices and delivery methods vary widely across laboratories. This review examines the emerging field of preclinical vapor models with a focus on cannabinoid exposure in order to (1) summarize vapor exposure parameters and other methodological details across studies; (2) discuss the pharmacological and behavioral effects produced by exposure to vaporized cannabinoids; and (3) compare behavioral effects of cannabinoid vapor administration with those of other routes of administration. This review will serve as a guide for past and current vapor delivery methods in animals, synergize findings across studies, and propose future directions for this area of research.


Assuntos
Canabidiol , Canabinoides , Cannabis , Alucinógenos , Animais , Animais de Laboratório , Canabidiol/farmacologia , Agonistas de Receptores de Canabinoides , Canabinoides/farmacologia , Dronabinol/farmacologia
5.
Int J Mol Sci ; 23(20)2022 Oct 12.
Artigo em Inglês | MEDLINE | ID: mdl-36293020

RESUMO

STAT3 plays a protective role against ischemic brain injury; however, it is not clear which brain cell type mediates this effect, and by which mechanism. We tested the hypothesis that endothelial STAT3 contributes to protection from cerebral ischemia, by preserving cerebrovascular endothelial function and blood-brain barrier (BBB) integrity. The objective of this study was to determine the role of STAT3 in cerebrovascular endothelial cell (EC) survival and function, and its role in tissue outcome after cerebral ischemia. We found that in primary mouse brain microvascular ECs, STAT3 was constitutively active, and its phosphorylation was reduced by oxygen-glucose deprivation (OGD), recovering after re-oxygenation. STAT3 inhibition, using two mechanistically different pharmacological inhibitors, increased EC injury after OGD. The sub-lethal inhibition of STAT3 caused endothelial dysfunction, demonstrated by reduced nitric oxide release in response to acetylcholine and reduced barrier function of the endothelial monolayer. Finally, mice with reduced endothelial STAT3 (Tie2-Cre; STAT3flox/wt) sustained larger brain infarcts after middle cerebral artery occlusion (MCAO) compared to wild-type (WT) littermates. We conclude that STAT3 is vital to maintaining cerebrovascular integrity, playing a role in EC survival and function, and protection against cerebral ischemia. Endothelial STAT3 may serve as a potential target in preventing endothelial dysfunction after stroke.


Assuntos
Lesões Encefálicas , Isquemia Encefálica , Animais , Camundongos , Óxido Nítrico/metabolismo , Acetilcolina/metabolismo , Isquemia Encefálica/metabolismo , Barreira Hematoencefálica/metabolismo , Infarto da Artéria Cerebral Média/metabolismo , Glucose/metabolismo , Oxigênio/metabolismo , Lesões Encefálicas/metabolismo
6.
Calcif Tissue Int ; 108(6): 808-818, 2021 06.
Artigo em Inglês | MEDLINE | ID: mdl-33517470

RESUMO

Ionizing radiation, from both space and radiation therapy, is known to affect bone health. While there have been studies investigating changes in bone density and microstructure from radiation exposure, the effects of radiation on material properties are unknown. The current study addresses this gap by assessing bone material property changes in rats exposed to helium-4 radiation through spherical micro-indentation. Rats were exposed to a single dose of 0, 5, and 25 cGy whole body helium-4 radiation. Animals were euthanized at 7, 30, 90, or 180-days after exposure. Spherical micro-indentation was performed on axial cross sections of the femur cortical bone to determine instantaneous and relaxed shear moduli. At 90-days after exposure, the 25 cGy exposure caused a significant decline in shear modulus compared to control and 5 cGy groups. The instantaneous modulus decreased 33% and the relaxed modulus decreased 32% as compared to the sham group. This decline was followed by a recovery of both moduli, which was observed by 180-days after exposure; at 180 days, the moduli were no longer statistically different from those at 7 or 30 days. The observed decrease at 90 days, followed by recovery to baseline levels, can be attributed to the biological mechanisms involved in bone formation that were affected by radiation, bone turnover, and systemic changes in hormones due to radiation exposure. Continued assessment of the mechanisms that drive such a response in material properties may enable identification of pathways for therapeutic countermeasures against radiation exposure.


Assuntos
Osso e Ossos , Hélio , Animais , Densidade Óssea , Osso Cortical , Fêmur , Ratos
7.
Int J Mol Sci ; 22(11)2021 May 21.
Artigo em Inglês | MEDLINE | ID: mdl-34063817

RESUMO

Soluble epoxide hydrolase (sEH) is abundant in the brain, is upregulated in type 2 diabetes mellitus (DM2), and is possible mediator of ischemic injury via the breakdown of neuroprotective epoxyeicosatrienoic acids (EETs). Prophylactic, pre-ischemic sEH blockade with 4-[[trans-4-[[(tricyclo[3.3.1.13,7]dec-1-ylamino)carbonyl]amino]cyclohexyl]oxy]-benzoic acid (tAUCB) reduces stroke-induced infarct in normal and diabetic mice, with larger neuroprotection in DM2. The present study tested whether benefit occurs in normal and DM2 mice if tAUCB is administered after stroke onset. We performed 60 min middle cerebral artery occlusion in young adult male C57BL mice divided into four groups: normal or DM2, with t-AUCB 2 mg/kg or vehicle 30 min before reperfusion. Endpoints were (1) cerebral blood flow (CBF) by laser Doppler, and (2) brain infarct at 24 h. In nondiabetic mice, t-AUCB reduced infarct size by 30% compared to vehicle-treated mice in the cortex (31.4 ± 4 vs. 43.8 ± 3 (SEM)%, respectively) and 26% in the whole hemisphere (26.3 ± 3 vs. 35.2 ± 2%, both p < 0.05). In contrast, in DM2 mice, tAUCB failed to ameliorate either cortical or hemispheric injury. No differences were seen in CBF. We conclude that tAUCB administered after ischemic stroke onset exerts brain protection in nondiabetic but not DM2 mice, that the neuroprotection appears independent of changes in gross CBF, and that DM2-induced hyperglycemia abolishes t-AUCB-mediated neuroprotection after stroke onset.


Assuntos
Diabetes Mellitus Experimental/metabolismo , Diabetes Mellitus Tipo 2/metabolismo , Epóxido Hidrolases/antagonistas & inibidores , Substâncias Protetoras/farmacologia , Acidente Vascular Cerebral/metabolismo , Animais , Benzoatos/farmacologia , Encéfalo/efeitos dos fármacos , Encéfalo/metabolismo , Circulação Cerebrovascular/efeitos dos fármacos , Infarto da Artéria Cerebral Média/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Acidente Vascular Cerebral/tratamento farmacológico , Ureia/análogos & derivados , Ureia/farmacologia
8.
Am J Physiol Heart Circ Physiol ; 318(1): H189-H202, 2020 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-31834840

RESUMO

We hypothesized that ranolazine-induced adenosine release is responsible for its beneficial effects in ischemic heart disease. Sixteen open-chest anesthetized dogs with noncritical coronary stenosis were studied at rest, during dobutamine stress, and during dobutamine stress with ranolazine. Six additional dogs without stenosis were studied only at rest. Regional myocardial function and perfusion were assessed. Coronary venous blood was drawn. Murine endothelial cells and cardiomyocytes were incubated with ranolazine and adenosine metabolic enzyme inhibitors, and adenosine levels were measured. Cardiomyocytes were also exposed to dobutamine and dobutamine with ranolazine. Modeling was employed to determine whether ranolazine can bind to an enzyme that alters adenosine stores. Ranolazine was associated with increased adenosine levels in the absence (21.7 ± 3.0 vs. 9.4 ± 2.1 ng/mL, P < 0.05) and presence of ischemia (43.1 ± 13.2 vs. 23.4 ± 5.3 ng/mL, P < 0.05). Left ventricular end-systolic wall stress decreased (49.85 ± 4.68 vs. 57.42 ± 3.73 dyn/cm2, P < 0.05) and endocardial-to-epicardial myocardial blood flow ratio tended to normalize (0.89 ± 0.08 vs. 0.76 ± 0.10, P = nonsignificant). Adenosine levels increased in cardiac endothelial cells and cardiomyocytes when incubated with ranolazine that was reversed when cytosolic-5'-nucleotidase (cN-II) was inhibited. Point mutation of cN-II aborted an increase in its specific activity by ranolazine. Similarly, adenosine levels did not increase when cardiomyocytes were incubated with dobutamine. Modeling demonstrated plausible binding of ranolazine to cN-II with a docking energy of -11.7 kcal/mol. We conclude that the anti-adrenergic and cardioprotective effects of ranolazine-induced increase in tissue adenosine levels, likely mediated by increasing cN-II activity, may contribute to its beneficial effects in ischemic heart disease.NEW & NOTEWORTHY Ranolazine is a drug used for treatment of angina pectoris in patients with ischemic heart disease. We discovered a novel mechanism by which this drug may exhibit its beneficial effects. It increases coronary venous levels of adenosine both at rest and during dobutamine-induced myocardial ischemia. Ranolazine also increases adenosine levels in endothelial cells and cardiomyocytes in vitro, by principally increasing activity of the enzyme cytosolic-5'-nucleotidase. Adenosine has well-known myocardial protective and anti-adrenergic properties that may explain, in part, ranolazine's beneficial effect in ischemic heart disease.


Assuntos
Adenosina/metabolismo , Fármacos Cardiovasculares/farmacologia , Estenose Coronária/tratamento farmacológico , Miócitos Cardíacos/efeitos dos fármacos , Ranolazina/farmacologia , 5'-Nucleotidase/química , 5'-Nucleotidase/metabolismo , Animais , Sítios de Ligação , Fármacos Cardiovasculares/química , Fármacos Cardiovasculares/metabolismo , Células Cultivadas , Estenose Coronária/metabolismo , Estenose Coronária/fisiopatologia , Modelos Animais de Doenças , Cães , Hemodinâmica/efeitos dos fármacos , Masculino , Camundongos Endogâmicos C57BL , Simulação de Acoplamento Molecular , Miócitos Cardíacos/metabolismo , Ligação Proteica , Conformação Proteica , Ranolazina/química , Ranolazina/metabolismo , Relação Estrutura-Atividade , Regulação para Cima , Função Ventricular Esquerda/efeitos dos fármacos
9.
Neurobiol Learn Mem ; 161: 115-121, 2019 05.
Artigo em Inglês | MEDLINE | ID: mdl-30953696

RESUMO

The hippocampus, medial dorsal thalamus and the perirhinal and entorhinal cortices are essential for visual recognition memory whereas the neural substrates underlying olfactory recognition memories are less well characterized. In the present study we combined chemogenetic inactivation with a social odor recognition memory (SORM) task to test the hypothesis that the medial prefrontal cortex (mPFC) is involved in recognition memory. We demonstrate that temporary chemogenetic inactivation of the mPFC prior to an encoding session impairs social odor recognition memory, whereas silencing the mPFC just prior to the recognition session was without effect. Our data support the critical role of the mPFC in the formation rather than retrieval of social odor memory.


Assuntos
Disfunção Cognitiva/fisiopatologia , Percepção Olfatória/fisiologia , Córtex Pré-Frontal/fisiologia , Reconhecimento Psicológico/fisiologia , Percepção Social , Animais , Comportamento Animal/fisiologia , Disfunção Cognitiva/induzido quimicamente , Técnicas Genéticas , Masculino , Rememoração Mental/fisiologia , Córtex Pré-Frontal/fisiopatologia , Ratos , Ratos Long-Evans
10.
Int J Mol Sci ; 20(2)2019 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-30650610

RESUMO

Future long-duration space missions will involve travel outside of the Earth's magnetosphere protection and will result in astronauts being exposed to high energy and charge (HZE) ions and protons. Exposure to this type of radiation can result in damage to the central nervous system and deficits in numerous cognitive domains that can jeopardize mission success. Social processing is a cognitive domain that is important for people living and working in groups, such as astronauts, but it has received little attention in terms of HZE ion exposure. In the current study, we assessed the effects of whole-body oxygen ion (16O; 1000 MeV/n) exposure (1 or 10 cGy) on social odor recognition memory in male Long-Evans rats at one and six months following exposure. Radiation exposure did not affect rats' preferences for a novel social odor experienced during Habituation at either time point. However, rats exposed to 10 cGy displayed short and long-term deficits in 24-h social recognition. In contrast, rats exposed to 1 cGy only displayed long-term deficits in 24-h social recognition. While an age-related decrease in Ki67+ staining (a marker of cell proliferation) was found in the subventricular zone, it was unaffected by radiation exposure. At one month following exposure, plasma KC/GRO (CXCL1) levels were elevated in the 1 cGy rats, but not in the 10 cGy rats, suggesting that peripheral levels of this cytokine could be associated with intact social recognition at earlier time points following radiation exposure. These results have important implications for long-duration missions and demonstrate that behaviors related to social processing could be negatively affected by HZE ion exposure.


Assuntos
Citocinas/sangue , Odorantes/análise , Oxigênio/química , Comportamento Social , Animais , Habituação Psicofisiológica , Íons , Antígeno Ki-67/metabolismo , Masculino , Ratos Long-Evans , Reconhecimento Psicológico , Fatores de Tempo
11.
J Neurosci ; 37(16): 4255-4269, 2017 04 19.
Artigo em Inglês | MEDLINE | ID: mdl-28320842

RESUMO

After nerve injury, Schwann cells convert to a phenotype specialized to promote repair. But during the slow process of axonal regrowth, these repair Schwann cells gradually lose their regeneration-supportive features and eventually die. Although this is a key reason for the frequent regeneration failures in humans, the transcriptional mechanisms that control long-term survival and phenotype of repair cells have not been studied, and the molecular signaling underlying their decline is obscure. We show, in mice, that Schwann cell STAT3 has a dual role. It supports the long-term survival of repair Schwann cells and is required for the maintenance of repair Schwann cell properties. In contrast, STAT3 is less important for the initial generation of repair Schwann cells after injury. In repair Schwann cells, we find that Schwann cell STAT3 activation by Tyr705 phosphorylation is sustained during long-term denervation. STAT3 is required for maintaining autocrine Schwann cell survival signaling, and inactivation of Schwann cell STAT3 results in a striking loss of repair cells from chronically denervated distal stumps. STAT3 inactivation also results in abnormal morphology of repair cells and regeneration tracks, and failure to sustain expression of repair cell markers, including Shh, GDNF, and BDNF. Because Schwann cell development proceeds normally without STAT3, the function of this factor appears restricted to Schwann cells after injury. This identification of transcriptional mechanisms that support long-term survival and differentiation of repair cells will help identify, and eventually correct, the failures that lead to the deterioration of this important cell population.SIGNIFICANCE STATEMENT Although injured peripheral nerves contain repair Schwann cells that provide signals and spatial clues for promoting regeneration, the clinical outcome after nerve damage is frequently poor. A key reason for this is that, during the slow growth of axons through the proximal parts of injured nerves repair, Schwann cells gradually lose regeneration-supporting features and eventually die. Identification of signals that sustain repair cells is therefore an important goal. We have found that in mice the transcription factor STAT3 protects these cells from death and contributes to maintaining the molecular and morphological repair phenotype that promotes axonal regeneration. Defining the molecular mechanisms that maintain repair Schwann cells is an essential step toward developing therapeutic strategies that improve nerve regeneration and functional recovery.


Assuntos
Regeneração Nervosa , Traumatismos dos Nervos Periféricos/metabolismo , Fenótipo , Fator de Transcrição STAT3/genética , Células de Schwann/metabolismo , Animais , Fator Neurotrófico Derivado do Encéfalo/genética , Fator Neurotrófico Derivado do Encéfalo/metabolismo , Feminino , Fator Neurotrófico Derivado de Linhagem de Célula Glial/genética , Fator Neurotrófico Derivado de Linhagem de Célula Glial/metabolismo , Masculino , Camundongos , Fator de Transcrição STAT3/metabolismo , Células de Schwann/citologia
12.
Stroke ; 46(7): 1916-22, 2015 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-25991416

RESUMO

BACKGROUND AND PURPOSE: Acute communicating hydrocephalus and cerebral edema are common and serious complications of subarachnoid hemorrhage (SAH), whose causes are poorly understood. Using a mouse model of SAH, we determined whether soluble epoxide hydrolase (sEH) gene deletion protects against SAH-induced hydrocephalus and edema by increasing levels of vasoprotective eicosanoids and suppressing vascular inflammation. METHODS: SAH was induced via endovascular puncture in wild-type and sEH knockout mice. Hydrocephalus and tissue edema were assessed by T2-weighted magnetic resonance imaging. Endothelial activation was assessed in vivo using T2*-weighted magnetic resonance imaging after intravenous administration of iron oxide particles linked to anti-vascular cell adhesion molecule-1 antibody 24 hours after SAH. Behavioral outcome was assessed at 96 hours after SAH with the open field and accelerated rotarod tests. RESULTS: SAH induced an acute sustained communicating hydrocephalus within 6 hours of endovascular puncture in both wild-type and sEH knockout mice. This was followed by tissue edema, which peaked at 24 hours after SAH and was limited to white matter fiber tracts. sEH knockout mice had reduced edema, less vascular cell adhesion molecule-1 uptake, and improved outcome compared with wild-type mice. CONCLUSIONS: Genetic deletion of sEH reduces vascular inflammation and edema and improves outcome after SAH. sEH inhibition may serve as a novel therapy for SAH.


Assuntos
Edema Encefálico/enzimologia , Epóxido Hidrolases/deficiência , Hemorragia Subaracnóidea/enzimologia , Vasculite/enzimologia , Animais , Edema Encefálico/patologia , Inflamação/enzimologia , Inflamação/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Hemorragia Subaracnóidea/patologia , Vasculite/patologia
13.
Am J Physiol Heart Circ Physiol ; 309(4): H583-91, 2015 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-26092990

RESUMO

Stroke outcome is improved by therapeutic ultrasound. This benefit is presumed to be principally from ultrasound-mediated thrombolysis. We hypothesized that the therapeutic benefit of ultrasound in stroke may, in part, be mediated by the release of beneficial vasoactive substances. Accordingly, we investigated the effect of ultrasound on levels of cytochrome P-450, lipoxygenase, and cyclooxygenase metabolites of arachidonic acid as well as adenosine release and endothelial nitric oxide synthase (eNOS) phosphorylation in primary brain endothelial cells in vitro. Brain endothelial cells were exposed to 1.05-MHz ultrasound at peak rarefactional acoustic pressure amplitudes of 0.35, 0.55, 0.90, and 1.30 MPa. Epoxyeicosatrienoic acids (EETs), hydroxyeicosatetraenoic acids (HETEs), PGE2, adenosine, nitrate/nitrite, and eNOS phosphorylation were measured after ultrasound exposure. Levels of 8,9-EET, 11,12-EET, and 14,15-EET increased by 230 ± 28%, 240 ± 30%, and 246 ± 31% (P < 0.05), respectively, whereas 5-HETE and 15-HETE levels were reduced to 24 ± 14% and 10 ± 3% (P < 0.05), respectively, compared with cells not exposed to ultrasound. PGE2 levels were reduced to 56 ± 14% of control. Adenosine increased more than sixfold after ultrasound exposure compared with unstimulated cells (1.36 ± 0.22 vs. 0.37 ± 0.10 ng/ml, P < 0.05), nitrate/nitrite was below levels of quantification, and eNOS phosphorylation was not altered significantly. Our results suggest that ultrasound may enhance tissue perfusion during stroke by augmenting the generation of vasodilator compounds and inhibiting that of vasoconstrictors. Such regulation supports a beneficial role for therapeutic ultrasound in stroke independent of its effect on the occlusive thrombus.


Assuntos
Encéfalo/citologia , Células Endoteliais/efeitos da radiação , Endotélio Vascular/efeitos da radiação , Som , Adenosina/metabolismo , Animais , Encéfalo/irrigação sanguínea , Encéfalo/efeitos da radiação , Células Cultivadas , Sistema Enzimático do Citocromo P-450/metabolismo , Dinoprostona/metabolismo , Células Endoteliais/metabolismo , Endotélio Vascular/metabolismo , Ácidos Hidroxieicosatetraenoicos/metabolismo , Lipoxigenase/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Óxido Nítrico Sintase Tipo III/metabolismo , Prostaglandina-Endoperóxido Sintases/metabolismo , Vasodilatação
14.
Am J Physiol Heart Circ Physiol ; 306(7): H929-37, 2014 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-24508640

RESUMO

Stroke risk and outcome are strongly modified by estrogen. In addition to ovaries, estrogen is produced locally in peripheral tissue by the enzyme aromatase, and extragonadal synthesis becomes the major source of estrogen after menopause. Aromatase gene deletion in female mice exacerbates ischemic brain damage after stroke. However, it is not clear which cell type is responsible for this effect, since aromatase is expressed in multiple cell types, including cerebrovascular endothelium. We tested the hypothesis that cerebrovascular aromatase contributes to sex differences in cerebrovascular endothelial function. Cerebrocortical microvascular responses to the endothelium-dependent vasodilator ACh were compared between male and female wild-type (WT) and aromatase knockout (ArKO) mice by measuring laser-Doppler perfusion in vivo through a closed cranial window. Additional studies were performed in WT mice treated with the aromatase inhibitor fadrozole or vehicle. WT female mice had significantly greater responses to ACh compared with WT males (P < 0.001), which was associated with higher aromatase expression in female compared with male cerebral vessels (P < 0.05). ACh responses were significantly lower in ArKO compared with WT females (P < 0.05) and in WT females treated with fadrozole versus vehicle (P < 0.001). Conversely, ACh responses were significantly higher in ArKO versus WT males (P < 0.05). Levels of phosphorylated endothelial nitric oxide synthase (eNOS) were lower in ArKO versus WT female brains, but were not altered by aromatase deletion in males. We conclude that cerebrovascular endothelial aromatase plays an important and sexually dimorphic role in cerebrovascular function and that aromatase inhibitors in clinical use may have cardiovascular consequences in both males and females.


Assuntos
Aromatase/metabolismo , Córtex Cerebral/irrigação sanguínea , Circulação Cerebrovascular , Endotélio Vascular/enzimologia , Microcirculação , Microvasos/enzimologia , Acetilcolina/farmacologia , Animais , Aromatase/deficiência , Aromatase/genética , Inibidores da Aromatase/farmacologia , Velocidade do Fluxo Sanguíneo , Circulação Cerebrovascular/efeitos dos fármacos , Relação Dose-Resposta a Droga , Endotélio Vascular/efeitos dos fármacos , Endotélio Vascular/fisiopatologia , Fadrozol/farmacologia , Feminino , Fluxometria por Laser-Doppler , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Microcirculação/efeitos dos fármacos , Microvasos/efeitos dos fármacos , Microvasos/fisiopatologia , Óxido Nítrico Sintase Tipo III/metabolismo , Fosforilação , Caracteres Sexuais , Fatores Sexuais , Vasodilatadores/farmacologia
15.
Arterioscler Thromb Vasc Biol ; 32(8): 1936-42, 2012 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-22723436

RESUMO

OBJECTIVE: Sex differences in cerebral ischemic injury are, in part, attributable to the differences in cerebrovascular perfusion. We determined whether the brain microvascular endothelial cells (ECs) isolated from the female brain are more resistant to ischemic injury compared with male ECs, and whether the difference is attributable to lower expression of soluble epoxide hydrolase and higher levels of vasoprotective epoxyeicosatrienoic acids (EETs). We also determined whether protection by EETs is linked to the inhibition of rho-kinase (ROCK). METHODS AND RESULTS: EC ischemic damage was measured after oxygen-glucose deprivation (OGD) using propidium iodide (PI) and cleaved caspase-3 labeling. Expression of soluble epoxide hydrolase was determined by quantitative polymerase chain reaction and immunocytochemistry, EETs levels by liquid chromatography-tandem mass spectrometry, and ROCK activity by ELISA. EC damage was higher in males compared with females, which correlated with higher soluble epoxide hydrolase mRNA, stronger immunoreactivity, and lower EETs compared with female ECs. Inhibition of soluble epoxide hydrolase abolished the sex difference in EC damage. ROCK activity was higher in male versus female ECs after OGD, and sex differences in EC damage and ROCK activity were abolished by 14,15-EET and ROCK inhibition. CONCLUSIONS: Sex differences in ischemic brain injury are, in part, attributable to differences in EET-mediated inhibition of EC ROCK activation after ischemia.


Assuntos
Isquemia Encefálica/etiologia , Células Endoteliais/fisiologia , Epóxido Hidrolases/fisiologia , Caracteres Sexuais , Ácido 8,11,14-Eicosatrienoico/análogos & derivados , Ácido 8,11,14-Eicosatrienoico/análise , Ácido 8,11,14-Eicosatrienoico/metabolismo , Amidas/farmacologia , Animais , Isquemia Encefálica/enzimologia , Sobrevivência Celular , Células Cultivadas , Epóxido Hidrolases/análise , Epóxido Hidrolases/imunologia , Feminino , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Piridinas/farmacologia , Solubilidade , Quinases Associadas a rho/metabolismo
16.
Adv Pharmacol ; 97: 283-326, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37236762

RESUMO

Cytochrome P450 metabolism of arachidonic acid produces epoxyeicosatrienoates (EETs) and hydroxyeicosatetraenoates (HETEs). Both classes of eicosanoids play important and opposing roles in brain function and disease. EETs promote vasodilation and exhibit antiinflammatory and cytoprotective properties; their biological action is blunted by metabolism to less active diols by the enzyme soluble epoxide hydrolase (sEH). EETs levels are dysregulated in disease states, primarily due to increased activity of sEH. Inhibition of sEH is a promising therapeutic approach for multiple brain disorders including stroke, dementia, subarachnoid hemorrhage and epilepsy. In this chapter, we summarize evidence implicating P450 eicosanoids and their synthetic and metabolizing enzymes in brain health and disease, and experimental and clinical studies targeting these pathways for brain disorders. We also discuss the diagnostic utility of quantifying P450 eicosanoids and their enzymes as disease biomarkers. Remarkable progress has been achieved in translating basic science discoveries in this field clinically.


Assuntos
Encefalopatias , Eicosanoides , Humanos , Eicosanoides/metabolismo , Sistema Enzimático do Citocromo P-450/metabolismo , Encéfalo/metabolismo
17.
Exp Clin Psychopharmacol ; 31(5): 881-887, 2023 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-36634015

RESUMO

A well-documented side effect of cannabis and Δ9-tetrahydrocannabinol (THC) acute administration is deficits in cognition and attention. Cannabidiol (CBD), a nonintoxicating constituent of cannabis, may modulate THC's impairing effects. A goal of this study was to determine the effects of THC and CBD, alone and in combination, on performance in the rodent Psychomotor Vigilance Test (rPVT), a translational paradigm used to quantify sustained attention. Outcome measures in the rPVT include motor speed, premature responding, and lapses in attention. Sprague Dawley rats were trained to perform the rPVT to the acquisition criteria and then received oral doses (mg/kg) of THC (1-17.6), CBD (1-100), and combinations of THC + CBD in sesame oil prior to rPVT sessions, administered in a within-subject randomized design. Blood was collected from rats receiving selected doses of THC alone or THC + CBD for analysis of THC and its metabolites. THC alone produced significant decreases in accuracy and increases in lapses in attention at higher doses (10 mg/kg; ps < .05). The coadministration of CBD (10 mg/kg) with THC (3 or 10 mg/kg) caused greater impairments to sustained attention compared with administration of THC alone (ps < .05). The rPVT is a translational platform sensitive to detect impairments in attention associated with THC and other cannabis constituents. Further work is necessary to determine the mechanism of THC and CBD interactions on impairments in sustained attention. (PsycInfo Database Record (c) 2023 APA, all rights reserved).


Assuntos
Canabidiol , Cannabis , Alucinógenos , Ratos , Animais , Dronabinol/farmacologia , Canabidiol/farmacologia , Ratos Sprague-Dawley , Agonistas de Receptores de Canabinoides , Atenção
18.
Ultrasound Med Biol ; 49(5): 1091-1101, 2023 05.
Artigo em Inglês | MEDLINE | ID: mdl-36739244

RESUMO

OBJECTIVE: There have been attempts to use therapeutic ultrasound (US) for the treatment of both experimental and clinical stroke. We hypothesized that low-intensity US has direct beneficial effects on the brain independent of cerebral blood flow (CBF) during middle cerebral artery occlusion (MCAO). METHODS: Three groups of mice were studied. Group I included 84 mice with MCAO undergoing US treatment/no treatment at two US frequencies (0.25 and 1.05 MHz) with three different acoustic pressures at each frequency in which infarct size (IS) was measured 24 h later. Group II included 11 mice undergoing treatment based on best US results from group I animals in which the IS/risk area (RA) ratio was measured 24 h later. Group III included 38 normal mice undergoing US treatment/no treatment for assessment of CBF, tissue metabolite and protein expression and histopathology. DISCUSSION: Ultrasound at both frequencies and most acoustic pressures resulted in reduction in IS in group I animals, with the best results obtained with 0.25 MHz at 2.0 MPa: IS was reduced 4-fold in the cerebral cortex, 1.5-fold in the caudate putamen and 3.5-fold in the cerebral hemisphere compared with control. US application in group III animals elicited only a marginal increase in CBF despite a 2.6-fold increase in phosphorylated endothelial nitric oxide synthase (p-eNOS)-S1177 and a corresponding decrease in p-eNOS-T494. Histopathology revealed no evidence of hemorrhage, inflammation or necrosis. CONCLUSION: Low-intensity US at specific frequencies and acoustic pressures results in marked neuroprotection in a mouse model of stroke by modulation of p-eNOS independent of its effect on CBF.


Assuntos
Isquemia Encefálica , Acidente Vascular Cerebral , Camundongos , Animais , Infarto da Artéria Cerebral Média/terapia , Óxido Nítrico/metabolismo , Encéfalo/patologia , Circulação Cerebrovascular , Modelos Animais de Doenças
19.
J Neurochem ; 121(4): 680-92, 2012 May.
Artigo em Inglês | MEDLINE | ID: mdl-22394374

RESUMO

Signal transduction and activator of transcription-3 (STAT3) plays an important role in neuronal survival, regeneration and repair after brain injury. We previously demonstrated that STAT3 is activated in brain after cerebral ischemia specifically in neurons. The effect was sex-specific and modulated by sex steroids, with higher activation in females than males. In the current study, we used a proteomics approach to identify downstream proteins affected by ischemia in male and female wild-type (WT) and neuron-specific STAT3 knockout (KO) mice. We established four comparison groups based on the transgenic condition and the hemisphere analyzed, respectively. Moreover, the sexual variable was taken into account and male and female animals were analyzed independently. Results support a role for STAT3 in metabolic, synaptic, structural and transcriptional responses to cerebral ischemia, indeed the adaptive response to ischemia/reperfusion injury is delayed in neuronal-specific STAT3 KO mice. The differences observed between males and females emphasize the importance of sex-specific neuronal survival and repair mechanisms, especially those involving antioxidant and energy-related activities, often caused by sex hormones.


Assuntos
Química Encefálica/genética , Isquemia Encefálica/genética , Encéfalo/fisiologia , Proteoma , Traumatismo por Reperfusão/genética , Fator de Transcrição STAT3/genética , Animais , Western Blotting , Isquemia Encefálica/fisiopatologia , Mapeamento Cromossômico , Corantes , Eletroforese em Gel Bidimensional , Feminino , Lateralidade Funcional/fisiologia , Processamento de Imagem Assistida por Computador , Infarto da Artéria Cerebral Média/genética , Infarto da Artéria Cerebral Média/patologia , Masculino , Espectrometria de Massas , Camundongos , Camundongos Knockout , Camundongos Transgênicos , Traumatismo por Reperfusão/fisiopatologia , Reprodutibilidade dos Testes , Caracteres Sexuais , Tripsina/química
20.
Artigo em Inglês | MEDLINE | ID: mdl-36179013

RESUMO

Background: The use of place conditioning procedures and drug vapor exposure models can increase our understanding of the rewarding and aversive effects of vaped cannabis products. Currently there are limited data on the conditioned rewarding effects of vaporized Δ9-tetrahydrocannabinol (THC), the primary psychoactive constituent of cannabis in rats, and no studies to date examining sex differences. Methods: Male and female Sprague-Dawley rats (N=96; 12 per sex/group) underwent place conditioning sessions immediately after exposure to THC or vehicle (propylene glycol [PG]) vapor. Locomotor activity was measured by beam breaks during conditioning sessions. THC vapor-conditioned rats received one of three THC vapor exposure amounts (low: 5 puffs of 100 mg/mL THC, medium: 5 puffs of 200 mg/mL THC, or high: 10 puffs of 200 mg/mL THC) and matched vehicle vapor (PG) exposure on alternate days for 16 daily sessions. A "no THC" control group of vehicle-conditioned rats received only PG vapor exposure each day. After the 8th and 16th conditioning sessions, untreated rats were tested for conditioned place preference (CPP) or aversion (CPA). Next, extinction tests and a THC vapor-primed reinstatement test were conducted. Results: THC vapor produced CPP and locomotor effects in an exposure dependent manner, and some sex differences were observed. Low THC vapor exposure did not produce CPP in males or females. Medium THC vapor exposure produced CPP in males, but not females. High THC vapor exposure produced CPP in both males and females. Medium and high THC vapor exposure amounts produced hyperactivity in female rats, but not male rats. CPP was more resistant to extinction in females than males. THC vapor reexposure (i.e., drug-prime) after extinction did not result in reinstatement of CPP for either sex. Conclusion: This study demonstrates conditioned rewarding effects of THC vapor in both male and female rats and provides evidence for sex differences in amounts of THC vapor that produce CPP and in time to extinction. CPA was not observed at any of the THC vapor exposure amounts tested. These data provide a foundation for future exploration of the conditioned effects of cannabis constituents and extracts using vapor exposure models.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA