Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 107
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Neurobiol Dis ; 145: 105074, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-32890773

RESUMEN

In utero alcohol exposure can induce severe neurodevelopmental disabilities leading to long-term behavioral deficits. Because alcohol induces brain defects, many studies have focused on nervous cells. However, recent reports have shown that alcohol markedly affects cortical angiogenesis in both animal models and infants with fetal alcohol spectrum disorder (FASD). In addition, the vascular system is known to contribute to controlling gamma-aminobutyric acid (GABA)ergic interneuron migration in the developing neocortex. Thus, alcohol-induced vascular dysfunction may contribute to the neurodevelopmental defects in FASD. The present study aimed at investigating the effects of alcohol on endothelial activity of pial microvessels. Ex vivo experiments on cortical slices from mouse neonates revealed that in endothelial cells from pial microvessels acute alcohol exposure inhibits both glutamate-induced calcium mobilization and activities of matrix metalloproteinase-9 (MMP-9) and tissue plasminogen activator (tPA). The inhibitory effect of alcohol on glutamate-induced MMP-9 activity was abrogated in tPA-knockout and Grin1flox/VeCadcre mice suggesting that alcohol interacts through the endothelial NMDAR/tPA/MMP-9 vascular pathway. Contrasting with the effects from acute alcohol exposure, in mouse neonates exposed to alcohol in utero during the last gestational week, glutamate exacerbated both calcium mobilization and endothelial protease activities from pial microvessels. This alcohol-induced vascular dysfunction was associated with strong overexpression of the N-methyl-d-aspartate receptor subunit GluN1 and mispositioning of the Gad67-GFP interneurons that normally populate the superficial cortical layers. By comparing several human control fetuses with a fetus chronically exposed to alcohol revealed that alcohol exposure led to mispositioning of the calretinin-positive interneurons, whose density was decreased in the superficial cortical layers II-III and increased in deepest layers. This study provides the first mechanistic and functional evidence that alcohol impairs glutamate-regulated activity of pial microvessels. Endothelial dysfunction is characterized by altered metalloproteinase activity and interneuron mispositioning, which was also observed in a fetus with fetal alcohol syndrome. These data suggest that alcohol-induced endothelial dysfunction may contribute in ectopic cortical GABAergic interneurons, that has previously been described in infants with FASD.


Asunto(s)
Células Endoteliales/efectos de los fármacos , Trastornos del Espectro Alcohólico Fetal/enzimología , Trastornos del Espectro Alcohólico Fetal/patología , Interneuronas/patología , Neurogénesis/efectos de los fármacos , Piamadre/efectos de los fármacos , Animales , Depresores del Sistema Nervioso Central/toxicidad , Células Endoteliales/enzimología , Etanol/toxicidad , Femenino , Neuronas GABAérgicas/efectos de los fármacos , Neuronas GABAérgicas/patología , Humanos , Interneuronas/efectos de los fármacos , Metaloproteasas/metabolismo , Ratones , Piamadre/enzimología , Embarazo , Efectos Tardíos de la Exposición Prenatal
2.
Cephalalgia ; 40(12): 1310-1320, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32611244

RESUMEN

BACKGROUND: The Transient Receptor Potential Ankyrin 1 (TRPA1) channel might play a role in migraine. However, different mechanisms for this have been suggested. The purpose of our study was to investigate the localization and significance of TRPA1 channels in rat pial and dural arteries. METHODS: Immunofluorescence microscopy was used to localize TRPA1 channels in dural arteries, pial arteries, dura mater and trigeminal ganglion. The genuine closed cranial window model was used to examine the effect of Na2S, a donor of the TRPA1 channel opener H2S, on the diameter of pial and dural arteries. Further, we performed blocking experiments with TRPA1 antagonist HC-030031, calcitonin gene-related peptide (CGRP) receptor antagonist olcegepant and KCa3.1 channel blocker TRAM-34. RESULTS: TRPA1 channels were localized to the endothelium of both dural and pial arteries and in nerve fibers in dura mater. Further, we found TRPA1 expression in the membrane of trigeminal ganglia neuronal cells, some of them also staining for CGRP. Na2S caused dilation of both dural and pial arteries. In dural arteries, this was inhibited by HC-030031 and olcegepant. In pial arteries, the dilation was inhibited by TRAM-34, suggesting involvement of the KCa3.1 channel. CONCLUSION: Na2S causes a TRPA1- and CGRP-dependent dilation of dural arteries and a KCa3.1 channel-dependent dilation of pial arteries in rats.


Asunto(s)
Duramadre/metabolismo , Piamadre/metabolismo , Sulfuros/farmacología , Canal Catiónico TRPA1/metabolismo , Vasodilatadores/farmacología , Animales , Duramadre/efectos de los fármacos , Masculino , Trastornos Migrañosos/metabolismo , Trastornos Migrañosos/fisiopatología , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Piamadre/efectos de los fármacos , Ratas , Ratas Sprague-Dawley , Canal Catiónico TRPA1/efectos de los fármacos
3.
Stroke ; 49(5): 1267-1275, 2018 05.
Artículo en Inglés | MEDLINE | ID: mdl-29669868

RESUMEN

BACKGROUND AND PURPOSE: Reperfusion is the most significant determinant of good outcome after ischemic stroke. However, complete reperfusion often cannot be achieved, despite satisfactory recanalization. We hypothesized that microvascular protection was essential for achieving effective reperfusion and, hence, neuroprotection. To test this hypothesis, we have developed an in vivo model to differentially monitor parenchymal and vascular reactive oxygen species (ROS) formation. By comparing the ROS-suppressing effect of N-tert-butyl-α-phenylnitrone (PBN) with its blood-brain barrier impermeable analog 2-sulfo-phenyl-N-tert-butylnitrone (S-PBN), we assessed the impact of vascular ROS suppression alone on reperfusion and stroke outcome after recanalization. METHODS: The distal middle cerebral artery was occluded for 1 hour by compressing with a micropipette and then recanalized (n=60 Swiss mice). ROS formation was monitored for 1 hour after recanalization by intravital fluorescence microscopy in pial vasculature and cortical parenchyma with topically applied hydroethidine through a cranial window. PBN (100 mg/kg) or S-PBN (156 mg/kg) was administered shortly before recanalization, and suppression of the vascular and parenchymal hydroethidine fluorescence was examined (n=22). Microcirculatory patency, reperfusion, ischemic tissue size, and neurological outcome were also assessed in a separate group of mice 1 to 72 hours after recanalization (n=30). RESULTS: PBN and S-PBN completely suppressed the reperfusion-induced increase in ROS signal within vasculature. PBN readily suppressed ROS produced in parenchyma by 88%. S-PBN also suppressed the parenchymal ROS by 64% but starting 40 minutes later. Intriguingly, PBN and S-PBN comparably reduced the size of ischemic area by 65% and 48% (P>0.05), respectively. S-PBN restored the microvascular patency and perfusion after recanalization, suggesting that its delayed parenchymal antioxidant effect could be secondary to improved microcirculatory reperfusion. CONCLUSIONS: Promoting microvascular reperfusion by protecting vasculature can secondarily reduce parenchymal ROS formation and provide neuroprotection. The model presented can be used to directly assess pharmacological end points postulated in brain parenchyma and vasculature in vivo.


Asunto(s)
Bencenosulfonatos/farmacología , Corteza Cerebral/efectos de los fármacos , Circulación Cerebrovascular/efectos de los fármacos , Óxidos N-Cíclicos/farmacología , Infarto de la Arteria Cerebral Media/metabolismo , Microcirculación/efectos de los fármacos , Fármacos Neuroprotectores/farmacología , Piamadre/efectos de los fármacos , Especies Reactivas de Oxígeno/metabolismo , Animales , Barrera Hematoencefálica , Corteza Cerebral/irrigación sanguínea , Corteza Cerebral/metabolismo , Corteza Cerebral/patología , Colorantes Fluorescentes , Infarto de la Arteria Cerebral Media/patología , Microscopía Intravital , Masculino , Ratones , Microscopía Fluorescente , Fenantridinas , Piamadre/irrigación sanguínea , Piamadre/metabolismo , Piamadre/patología , Reperfusión
4.
BMC Anesthesiol ; 15: 37, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25805961

RESUMEN

BACKGROUND: JM-1232(-) is a novel anesthetic agent which acts through gamma-aminobutyric acid receptors. Cerebral pial vascular effects of JM-1232(-) are unknown. We thus evaluated topical and intravenous effects of JM-1232(-) on cerebral pial microvessels in rabbits, and the extent to which carbon dioxide (CO2) reactivity is preserved. METHODS: Closed cranial windows were used to visualize cerebral pial circulation in 29 Japanese white rabbits. In the first experiment, the cranial window was superfused with increasing concentrations of JM-1232(-): 10(-11), 10(-9), 10(-7), 10(-5) mol/L, n = 8 per concentration. In the second experiment, we examined the effects of an intravenous bolus of 1 mg/kg bolus of JM-1232(-), followed by the continuous infusion at 0.3 mg/kg/minute on cerebral pial vascular alteration (n = 9). In the third, we examined CO2 reactivity of cerebral pial vessels under JM-1232(-) (n = 6) or sevoflurane anesthesia (n = 6). RESULTS: Topical application of JM-1232(-) did not change pial venular diameter, and constricted arterials only at the highest concentration. Intravenous administration of JM-1232(-) produced cerebral pial constriction which gradually diminished over time. Under intravenous administration of JM-1232(-) and inhaled sevoflurane, diameters of vessels increased in parallel with CO2 partial pressure. Slopes of linear regression and correlation coefficients in arterioles and venules were comparable for JM-1232(-) anesthesia and sevoflurane anesthesia. CONCLUSIONS: Topical application of JM-1232(-) had little effect on cerebral pial vessels. Intravenous administration produced vasoconstriction of cerebral pial arterioles and venules, however those changes were clinically unimportant. In addition, JM-1232(-) did not impair CO2 responsiveness. At least from the perspective of vascular reactivity, JM-1232(-) thus appears safe for neurosurgical patients.


Asunto(s)
Arteriolas/efectos de los fármacos , Isoindoles/administración & dosificación , Isoindoles/farmacología , Piamadre/irrigación sanguínea , Piamadre/efectos de los fármacos , Piperazinas/administración & dosificación , Piperazinas/farmacología , Vénulas/efectos de los fármacos , Administración Intravenosa , Administración Tópica , Animales , Arteriolas/fisiología , Relación Dosis-Respuesta a Droga , Hipercapnia/fisiopatología , Hipocapnia/fisiopatología , Conejos , Vénulas/fisiología
5.
J Anesth ; 29(2): 186-90, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25150577

RESUMEN

PURPOSE: Rho-kinase inhibitor is widely used for prevention of cerebral vascular spasm. However, the cerebral pial vascular action of Rho-kinase inhibitor has not been investigated. We therefore evaluated the direct effects of Y-27632, a Rho-kinase inhibitor, on pial microvessels. METHOD: Experiments were performed on anesthetized rabbits. A closed cranial window was used to visualize the pial microcirculation. After baseline hemodynamic and pial vascular measurements, the cranial window was superfused with four increasing concentrations of Y-27632 (10(-9), 10(-7), 10(-6), 10(-5) mol l(-1); n = 7) dissolved in artificial cerebrospinal fluid for 7 min each. We measured the diameters of pial vessels, mean arterial pressure (MAP), heart rate (HR), and rectal temperature at 7 min after application of each Y-27632 concentration. RESULTS: MAP, HR, rectal temperature, arterial pH, PaCO2, PaO2, and plasma Na(+), K(+) and glucose concentrations did not change significantly during the experimental period. Y-27632 at 10(-9) to 10(-7) mol l(-1) did not produce any significant change in pial arterioles. Topical application of Y-27632 at 10(-6) and 10(-5) mol l(-1) produced pial large (8.4 ± 5.7 and 19.8 ± 12.7 %) and small (10.1 ± 8.5 and 18.1 ± 12.3 %) arterioles dilation. However, Y-27632 did not produce any change in pial large and small venules. CONCLUSION: We evaluated the direct effects of Y-27632 on pial microvessels. Y-27632 dilates only pial arterioles in a concentration-dependent manner, and most at a concentration of 10(-5) mol l(-1). Y-27632 is a potent cerebral pial arteriolar dilator but is not a venular dilator.


Asunto(s)
Amidas/farmacología , Capilares/efectos de los fármacos , Piamadre/irrigación sanguínea , Piamadre/efectos de los fármacos , Inhibidores de Proteínas Quinasas/farmacología , Piridinas/farmacología , Quinasas Asociadas a rho/antagonistas & inhibidores , Animales , Presión Arterial/efectos de los fármacos , Arteriolas/efectos de los fármacos , Temperatura Corporal/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Frecuencia Cardíaca/efectos de los fármacos , Hemodinámica/efectos de los fármacos , Microcirculación/efectos de los fármacos , Conejos , Vasodilatación/efectos de los fármacos , Vénulas/efectos de los fármacos
6.
Childs Nerv Syst ; 28(4): 533-40, 2012 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-22252717

RESUMEN

PURPOSE: The purpose of this paper is to study the ionic permeability of the leptomeninges related to the effect of ouabain (sodium-potassium-ATPase inhibitor) and amiloride (epithelial sodium channel (ENaC) inhibitor) on the tissue, as well as identify the presence of ion channels. METHODS: Cranial leptomeningeal samples from 26 adult sheep were isolated. Electrophysiological measurements were performed with Ussing system and transmembrane resistance values (R(TM) in Ω*cm(2)) obtained over time. Experiments were conducted with the application of ouabain 10(-3) M or amiloride 10(-5) M at the arachnoidal and pial sides. Immunohistochemical studies of leptomeningeal tissue were prepared with alpha-1 sodium-potassium-ATPase (ATP1A1), beta-ENaC, and delta-ENaC subunit antibodies. RESULTS: The application of ouabain at the arachnoidal side raised the transmembrane resistance statistically significantly and thus decreased its ionic permeability. The addition of ouabain at the pial side led also to a significant but less profound increment in transmembrane resistance. The addition of amiloride at the arachnoidal or pial side did not produce any statistical significant change in the R(TM) from controls (p > 0.05). Immunohistochemistry confirmed the presence of the ATP1A1 and beta- and delta-ENaC subunits at the leptomeninges. CONCLUSIONS: In summary, leptomeningeal tissue possesses sodium-potassium-ATPase and ENaC ion channels. The application of ouabain alters the ionic permeability of the leptomeninges thus reflecting the role of sodium-potassium-ATPase. Amiloride application did not alter the ionic permeability of leptomeninges possibly due to localization of ENaC channels towards the subarachnoid space, away from the experimental application sites. The above properties of the tissue could potentially be related to cerebrospinal fluid turnover at this interface.


Asunto(s)
Aracnoides/metabolismo , Permeabilidad de la Membrana Celular/fisiología , Canales Epiteliales de Sodio/fisiología , Piamadre/metabolismo , ATPasa Intercambiadora de Sodio-Potasio/fisiología , Amilorida/farmacología , Animales , Aracnoides/efectos de los fármacos , Permeabilidad de la Membrana Celular/efectos de los fármacos , Canales Epiteliales de Sodio/metabolismo , Femenino , Masculino , Ouabaína/farmacología , Piamadre/efectos de los fármacos , Ovinos , ATPasa Intercambiadora de Sodio-Potasio/metabolismo
7.
8.
Biomolecules ; 11(12)2021 12 10.
Artículo en Inglés | MEDLINE | ID: mdl-34944506

RESUMEN

Renin-angiotensin systems produce angiotensin II (Ang II) and angiotensin 1-7 (Ang 1-7), which are able to induce opposite effects on circulation. This study in vivo assessed the effects induced by Ang II or Ang 1-7 on rat pial microcirculation during hypoperfusion-reperfusion, clarifying the mechanisms causing the imbalance between Ang II and Ang 1-7. The fluorescence microscopy was used to quantify the microvascular parameters. Hypoperfusion and reperfusion caused vasoconstriction, disruption of blood-brain barrier, reduction of capillary perfusion and an increase in reactive oxygen species production. Rats treated with Ang II showed exacerbated microvascular damage with stronger vasoconstriction compared to hypoperfused rats, a further increase in leakage, higher decrease in capillary perfusion and marker oxidative stress. Candesartan cilexetil (specific Ang II type 1 receptor (AT1R) antagonist) administration prior to Ang II prevented the effects induced by Ang II, blunting the hypoperfusion-reperfusion injury. Ang 1-7 or ACE2 activator administration, preserved the pial microcirculation from hypoperfusion-reperfusion damage. These effects of Ang 1-7 were blunted by a Mas (Mas oncogene-encoded protein) receptor antagonist, while Ang II type 2 receptor antagonists did not affect Ang 1-7-induced changes. In conclusion, Ang II and Ang 1-7 triggered different mechanisms through AT1R or MAS receptors able to affect cerebral microvascular injury.


Asunto(s)
Angiotensina II/administración & dosificación , Angiotensina I/administración & dosificación , Bencimidazoles/administración & dosificación , Compuestos de Bifenilo/administración & dosificación , Fragmentos de Péptidos/administración & dosificación , Piamadre/irrigación sanguínea , Daño por Reperfusión/metabolismo , Tetrazoles/administración & dosificación , Angiotensina I/efectos adversos , Angiotensina II/efectos adversos , Animales , Bencimidazoles/farmacología , Compuestos de Bifenilo/farmacología , Femenino , Masculino , Microcirculación/efectos de los fármacos , Microscopía Fluorescente , Fragmentos de Péptidos/efectos adversos , Piamadre/efectos de los fármacos , Piamadre/metabolismo , Proto-Oncogenes Mas/metabolismo , Ratas , Especies Reactivas de Oxígeno/metabolismo , Daño por Reperfusión/tratamiento farmacológico , Tetrazoles/farmacología
9.
Microvasc Res ; 78(2): 212-7, 2009 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-19555699

RESUMEN

Cerebral ischemia/reperfusion (I/R) results in cellular energy failure and dysfunction of the neurovascular unit that contribute to subsequent neuronal cell death in the neonate. PX-18 is a putative neuroprotective inhibitor of secretory phospholipase A(2) (sPLA(2)) but its in vivo testing has been limited by its poor solubility. Our purpose was to assess whether PX-18 preserved neuronal-vascular reactivity to I/R-sensitive endothelium-dependent (hypercapnia, bradykinin) and/or neuron-dependent (N-methyl-D-aspartate; NMDA) stimuli. To make the drug available for in vivo studies, PX-18 was formulated as a 3% nanosuspension applying high pressure homogenization. Newborn piglets (1-day old, n=40) were anesthetized and ventilated, and cerebrovascular reactivity to the above stimuli was determined by measuring changes in pial arteriolar diameters using the closed cranial window/intravital videomicroscopy technique. Intravenous infusion of PX-18 nanosuspension (6 mg/kg, 20 min) did not affect baseline arteriolar diameters, or hypercapnia-, bradykinin-, or NMDA-induced pial arteriolar vasodilation under normoxic conditions. Global cerebral ischemia (10 min) followed by 1 h of reperfusion significantly attenuated hypercapnia-, bradykinin-, and NMDA-induced vasodilation in untreated or vehicle-treated controls. However, PX-18 resulted in nearly full preservation of cerebrovascular reactivity to all these stimuli. In conclusion, inhibition of sPLA(2) by PX-18 improves neurovascular function both at the neuronal and the microvascular level following I/R. This effect of PX-18 likely contributes to its neuroprotective effect.


Asunto(s)
Ácidos Alcanesulfónicos/farmacología , Isquemia Encefálica/fisiopatología , Circulación Cerebrovascular/efectos de los fármacos , Inhibidores Enzimáticos/farmacología , Ácidos Oléicos/farmacología , Fosfolipasas A2 Secretoras/antagonistas & inhibidores , Animales , Animales Recién Nacidos , Arteriolas/efectos de los fármacos , Arteriolas/fisiología , Bradiquinina/farmacología , Arterias Cerebrales/efectos de los fármacos , Arterias Cerebrales/inervación , Química Farmacéutica/métodos , Evaluación Preclínica de Medicamentos , Hipercapnia/fisiopatología , Microscopía por Video , N-Metilaspartato/farmacología , Neuronas/efectos de los fármacos , Tamaño de la Partícula , Piamadre/irrigación sanguínea , Piamadre/efectos de los fármacos , Daño por Reperfusión/tratamiento farmacológico , Porcinos , Vasodilatación/efectos de los fármacos , Vasodilatación/fisiología , Vasodilatadores/farmacología
10.
Science ; 157(3792): 1074-7, 1967 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-6036240

RESUMEN

Deafferented somatosensory thalamic neurons showed hyperactivity, followed by greatly reduced activity, after initiation of cortical spreading depression; local cooling of sensorimotor cortex was followed only by the inactive phase. Stimulation of contralateral midbrain reticular formation during the inactive phase failed to induce the typical increase in discharge rate of somatosensory thalamic neurons, but produced desynchronization in unaffected cortex. These results indicate that corticothalamic discharge is necessary for sustaining the ongoing activity of deafferented somatosensory thalamic neurons and for maintaining their responsiveness to stimulation of the reticular formation.


Asunto(s)
Depresión de Propagación Cortical , Neuronas/fisiología , Formación Reticular/fisiología , Tálamo/fisiología , Animales , Gatos , Estimulación Eléctrica , Piamadre/efectos de los fármacos , Cloruro de Potasio/farmacología
11.
J Neurol ; 255(6): 828-30, 2008 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-18446313
12.
Brain Res ; 1165: 81-8, 2007 Aug 24.
Artículo en Inglés | MEDLINE | ID: mdl-17658492

RESUMEN

Pituitary adenylate cyclase-activating polypeptide (PACAP) is a cerebrovascular dilator and was found neuroprotective in numerous in vitro and in vivo models of cerebral ischemia. However, the mechanism of its cerebrovascular action is poorly known, especially in newborns. Therefore, we tested pial arteriolar responses to the two naturally occurring forms PACAP27 and 38 as well as to shorter sequences (PACAP6-27, 6-38, 1-15, 6-15, 20-31). We also investigated the involvement of nitric oxide synthase (NOS), cyclooxygenase-1 and -2 (COX-1 and -2) activity in PACAP-induced pial arteriolar responses using the NOS inhibitor N-omega-nitro-l-arginine methyl ester (L-NAME 15 mg/kg iv), the non-selective COX inhibitor indomethacin (5 mg/kg iv), and the selective COX-1 and COX-2 inhibitors SC-560 (1 mg/kg iv) and NS-398 (1 mg/kg iv), respectively. Anesthetized, ventilated piglets (n=127) were equipped with closed cranial windows, and pial arteriolar diameters were determined via intravital microscopy. Topical application of both natural PACAPs, but none of the PACAP segments, resulted in prominent, repeatable, dose-dependent vasodilation. Percentage changes ranged 5+/-1-29+/-6 (n=7) and 4+/-1-36+/-7 (n=9) to 10(-)(8) to 10(-)(6) M PACAP27 and 38 (mean+/-SEM), respectively. Vasodilation to both natural PACAPs was significantly reduced by co-application with PACAP6-27 or 6-38, but not by L-NAME. Indomethacin abolished PACAP38 but not PACAP27-induced vasodilation. Arteriolar responses to PACAP38 were also sensitive to SC-560 but not to NS-398 suggesting the unique involvement of COX-1 activity in this response. In summary, PACAP27 and 38 are potent vasodilators in the neonatal cerebral circulation with at least two distinct mechanisms of action: a COX-dependent and a COX-independent pathway.


Asunto(s)
Arteriolas/efectos de los fármacos , Piamadre/irrigación sanguínea , Polipéptido Hipofisario Activador de la Adenilato-Ciclasa/farmacología , Prostaglandina-Endoperóxido Sintasas/metabolismo , Vasodilatadores/farmacología , Análisis de Varianza , Animales , Animales Recién Nacidos , Arteriolas/fisiología , Relación Dosis-Respuesta a Droga , Interacciones Farmacológicas , Inhibidores Enzimáticos/farmacología , Péptidos/farmacología , Piamadre/efectos de los fármacos , Porcinos , Vasoconstricción/efectos de los fármacos , Vasoconstricción/fisiología
13.
Brain Res ; 1655: 48-54, 2017 01 15.
Artículo en Inglés | MEDLINE | ID: mdl-27865779

RESUMEN

Streptozotocin (STZ)-induced chronic hyperglycemia has a detrimental effect on neurovascular coupling, linked to increased PKC-mediated phosphorylation and PKC isoform expression changes. Here, we sought to determine whether: 1) selective PKC-α/ß/γ inhibitor, GF109203X, could reverse the effects of chronic hyperglycemia on cerebrovascular reactivity; 2) pancreatic islet transplantation could prevent the development of cerebrovascular impairment seen in a rat model of Type 1 Diabetes. We studied the effect of GF109203X in diabetic (DM), non-diabetic (ND), and transplanted (TR) Lewis rats during either sciatic nerve stimulation (SNS) or the topical applications of the large-conductance Ca2+-operated K+(BKCa) channel opener, NS1619, or the K+ inward rectifier (Kir) channel agonist, KCl. Pial arteriole diameter changes were monitored using a closed cranial window in vivo microscopy technique. The pial arteriole dilatory response associated with SNS was decreased by ~45%, when comparing DM vs either ND or TR rats. Also, pial arteriolar dilations to topical KCl and NS1619 were largely attenuated in DM rats, but not in ND or TR animals. These responses were completely restored by the acute application of GF109203X to the brain surface. The PKC inhibitor had no effect on vascular responses in normoglycemic and TR animals. In conclusion, DM-associated chronic impairment of neurovascular coupling may be readily reversed by a PKC-α/ß/γ inhibitor or prevented via pancreatic islet transplantation. We believe that specific PCK isoforms (α/ß/γ) are mechanistically linked to the neurovascular uncoupling seen with hyperglycemia.


Asunto(s)
Fármacos Cardiovasculares/farmacología , Diabetes Mellitus Experimental/terapia , Diabetes Mellitus Tipo 1/terapia , Trasplante de Islotes Pancreáticos , Acoplamiento Neurovascular , Proteína Quinasa C/antagonistas & inhibidores , Animales , Arteriolas/efectos de los fármacos , Arteriolas/fisiopatología , Bencimidazoles/farmacología , Diabetes Mellitus Experimental/fisiopatología , Diabetes Mellitus Tipo 1/fisiopatología , Inhibidores Enzimáticos/farmacología , Hipoglucemiantes/farmacología , Indoles/farmacología , Masculino , Maleimidas/farmacología , Neurotransmisores/farmacología , Acoplamiento Neurovascular/fisiología , Piamadre/efectos de los fármacos , Piamadre/fisiopatología , Cloruro de Potasio/farmacología , Proteína Quinasa C/metabolismo , Ratas Endogámicas Lew , Receptores KIR/agonistas , Receptores KIR/metabolismo , Nervio Ciático/efectos de los fármacos , Nervio Ciático/fisiopatología
14.
J Appl Physiol (1985) ; 100(2): 631-6, 2006 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-16210431

RESUMEN

Our goals were to determine whether chronic exposure to nicotine alters nitric oxide synthase (NOS)-dependent reactivity of cerebral (pial) arterioles and to identify a potential role for NADPH oxidase in impaired NOS-dependent responses during chronic exposure to nicotine. We measured in vivo diameter of pial arterioles to NOS-dependent (acetylcholine and ADP) and -independent (nitroglycerin) agonists in saline-treated rats and rats chronically treated with nicotine (2 mg.kg(-1).day(-1) for 2 wk via an osmotic minipump). We found that NOS-dependent, but not -independent, vasodilatation was impaired in nicotine-treated compared with saline-treated rats. In addition, the production of superoxide anion (lucigenin chemiluminescence) was increased in rats treated with nicotine compared with saline-treated rats. Furthermore, using Western blot analysis, we found that chronic exposure to nicotine increased p47phox protein in the parietal cortex. Finally, we found that apocynin (40 mg.kg(-1).day(-1)) in the drinking water to inhibit NADPH oxidase alleviated impaired NOS-dependent cerebral vasodilatation in nicotine treated rats but did not alter NOS-dependent responses in saline treated rats and did not alter NOS-independent reactivity in saline- or nicotine-treated rats. These findings suggest that chronic exposure to nicotine impairs NOS-dependent dilatation of pial arterioles by a mechanism that appears to be related to the formation of superoxide anion via activation of NADPH oxidase.


Asunto(s)
NADPH Oxidasas/metabolismo , Nicotina/farmacología , Piamadre/efectos de los fármacos , Vasodilatación , Acetofenonas/farmacología , Acetilcolina/farmacología , Animales , Arteriolas/efectos de los fármacos , Arteriolas/enzimología , Relación Dosis-Respuesta a Droga , Inhibidores Enzimáticos/farmacología , Bombas de Infusión Implantables , Masculino , NADPH Oxidasas/antagonistas & inhibidores , Nicotina/administración & dosificación , Nicotina/farmacocinética , Agonistas Nicotínicos/administración & dosificación , Agonistas Nicotínicos/farmacocinética , Agonistas Nicotínicos/farmacología , Óxido Nítrico Sintasa/metabolismo , Lóbulo Parietal/efectos de los fármacos , Lóbulo Parietal/enzimología , Piamadre/irrigación sanguínea , Piamadre/enzimología , Ratas , Ratas Sprague-Dawley , Superóxidos/metabolismo , Factores de Tiempo , Vasodilatadores/farmacología
15.
Brain Res ; 1634: 171-178, 2016 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-26773687

RESUMEN

S100B is an astrocyte-derived protein that can act through the receptor for advanced glycation endproducts (RAGE) to mediate either "trophic" or "toxic" responses. Its levels increase in many neurological conditions with associated microvascular dysregulation, such as subarachnoid hemorrhage (SAH) and traumatic brain injury. The role of S100B in the pathogenesis of microvasculopathy has not been addressed. This study was designed to examine whether S100B alters pial arteriolar vasodilating function. Rats were randomized to receive (1) artificial cerebrospinal fluid (aCSF), (2) exogenous S100B, and (3) exogenous S100B+the decoy soluble RAGE (sRAGE). S100B was infused intracerebroventricularly (icv) using an osmotic pump and its levels in the CSF were adjusted to achieve a concentration similar to what we observed in SAH. After 48 h of continuous icv infusion, a cranial window/intravital microscopy was applied to animals for evaluation of pial arteriolar dilating responses to sciatic nerve stimulation (SNS), hypercapnia, and topical suffusion of vasodilators including acetylcholine (ACh), s-nitroso-N-acetyl penicillamine (SNAP), or adenosine (ADO). Pial arteriolar dilating responses were calculated as the percentage change of arteriolar diameter in relation to baseline. The continuous S100B infusion for 48 h was associated with reduced responses to the neuronal-dependent vasodilator SNS (p<0.05) and the endothelial-dependent vasodilator ACh (p<0.05), compared to controls. The inhibitory effects of S100B were prevented by sRAGE. On the other hand, S100B did not alter the responses elicited by vascular smooth muscle cell-dependent vasodilators, namely hypercapnia, SNAP, or ADO. These findings indicate that S100B regulates neuronal and endothelial dependent cerebral arteriolar dilation and suggest that this phenomenon is mediated through RAGE-associated pathways.


Asunto(s)
Piamadre/irrigación sanguínea , Piamadre/fisiología , Receptor para Productos Finales de Glicación Avanzada/fisiología , Subunidad beta de la Proteína de Unión al Calcio S100/administración & dosificación , Subunidad beta de la Proteína de Unión al Calcio S100/fisiología , Acetilcolina/administración & dosificación , Adenosina/administración & dosificación , Animales , Arteriolas/efectos de los fármacos , Arteriolas/fisiología , Estimulación Eléctrica , Hipercapnia/metabolismo , Infusiones Intraventriculares , Masculino , Piamadre/efectos de los fármacos , Ratas , Ratas Sprague-Dawley , S-Nitroso-N-Acetilpenicilamina/administración & dosificación , Subunidad beta de la Proteína de Unión al Calcio S100/líquido cefalorraquídeo , Nervio Ciático/fisiología , Transducción de Señal/efectos de los fármacos , Vasodilatadores/administración & dosificación
16.
Sci Rep ; 6: 38786, 2016 12 09.
Artículo en Inglés | MEDLINE | ID: mdl-27934907

RESUMEN

Noninvasive microvascular imaging using optical coherence Doppler tomography (ODT) has shown great promise in brain studies; however, high-speed microcirculatory imaging in deep brain remains an open quest. A high-speed 1.3 µm swept-source ODT (SS-ODT) system is reported which was based on a 200 kHz vertical-cavity-surface-emitting laser. Phase errors induced by sweep-trigger desynchronization were effectively reduced by spectral phase encoding and instantaneous correlation among the A-scans. Phantom studies have revealed a significant reduction in phase noise, thus an enhancement of minimally detectable flow down to 268.2 µm/s. Further in vivo validation was performed, in which 3D cerebral-blood-flow (CBF) networks in mouse brain over a large field-of-view (FOV: 8.5 × 5 × 3.2 mm3) was scanned through thinned skull. Results showed that fast flows up to 3 cm/s in pial vessels and minute flows down to 0.3 mm/s in arterioles or venules were readily detectable at depths down to 3.2 mm. Moreover, the dynamic changes of the CBF networks elicited by acute cocaine such as heterogeneous responses in various vessel compartments and at different cortical layers as well as transient ischemic events were tracked, suggesting the potential of SS-ODT for brain functional imaging that requires high flow sensitivity and dynamic range, fast frame rate and a large FOV to cover different brain regions.


Asunto(s)
Arteriolas/diagnóstico por imagen , Circulación Cerebrovascular , Neuroimagen Funcional/métodos , Piamadre/irrigación sanguínea , Tomografía de Coherencia Óptica/métodos , Vénulas/diagnóstico por imagen , Animales , Arteriolas/efectos de los fármacos , Circulación Cerebrovascular/efectos de los fármacos , Cocaína/farmacología , Efecto Doppler , Procesamiento de Imagen Asistido por Computador , Imagenología Tridimensional , Ataque Isquémico Transitorio/diagnóstico por imagen , Rayos Láser , Masculino , Ratones , Ratones Endogámicos C57BL , Fantasmas de Imagen , Piamadre/diagnóstico por imagen , Piamadre/efectos de los fármacos , Vénulas/efectos de los fármacos
17.
Brain Res Dev Brain Res ; 156(2): 139-46, 2005 May 12.
Artículo en Inglés | MEDLINE | ID: mdl-16099300

RESUMEN

Previous studies have observed that fluid percussion brain injury (FPI) impaired NMDA induced pial artery dilation in an age-dependent manner. This study was designed to investigate the contribution of plasminogen activators to impaired NMDA dilation after FPI in newborn and juvenile pigs equipped with a closed cranial window. In the newborn pig, NMDA (10(-8), 10(-6) M) induced pial artery dilation was reversed to vasoconstriction following FPI, but pretreatment with the plasminogen activator inhibitor PAI-1 derived hexapeptide (EEIIMD) (10(-7) M) prevented post injury vasoconstriction (9 +/- 1 and 16 +/- 1, vs. -6 +/- 2 and-11 +/- 3, vs. 5 +/- 1 and 9 +/- 1% for responses to NMDA 10(-8), 10(-6) M prior to FPI, after FPI, and after FPI in EEIIMD pretreated animals, respectively). In contrast, in the juvenile pig, NMDA dilation was only attenuated following FPI and EEIIMD pretreatment partially prevented such inhibition (9 +/- 1 and 16 +/- 1 vs. 2 +/- 1 and 4 +/- 1 vs. 5 +/- 1 and 7 +/- 1% for responses to NMDA prior to FPI, after FPI, and after FPI in EEIIMD pretreated animals, respectively). Additionally, EEIIMD blunted age-dependent pial artery vasoconstriction following FPI. EEIIMD blocked dilation to the plasminogen activator agonists uPA and tPA while responses to SNP and papaverine were unchanged. Pretreatment with suPAR, which blocked dilation to uPA, elicited effects on pial artery diameter and NMDA vascular activity post FPI similar to that observed with EEIIMD. These data show that EEIIMD and suPAR partially prevented FPI induced alterations in NMDA dilation and reductions in pial artery diameter. EEIIMD and suPAR are efficacious and selective inhibitors of plasminogen activator induced dilation. These data suggest that plasminogen activators contribute to age-dependent impairment of NMDA induced dilation following FPI.


Asunto(s)
Lesiones Encefálicas/fisiopatología , Agonistas de Aminoácidos Excitadores/farmacología , N-Metilaspartato/farmacología , Activadores Plasminogénicos/fisiología , Vasodilatación/efectos de los fármacos , Factores de Edad , Análisis de Varianza , Animales , Animales Recién Nacidos , Arterias/efectos de los fármacos , Lesiones Encefálicas/líquido cefalorraquídeo , Lesiones Encefálicas/tratamiento farmacológico , Circulación Cerebrovascular/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Interacciones Farmacológicas , Ensayo de Inmunoadsorción Enzimática/métodos , Piamadre/irrigación sanguínea , Piamadre/efectos de los fármacos , Inhibidor 1 de Activador Plasminogénico/análogos & derivados , Inhibidor 1 de Activador Plasminogénico/uso terapéutico , Activadores Plasminogénicos/sangre , Inhibidores de Serina Proteinasa/uso terapéutico , Porcinos , Vasoconstricción/efectos de los fármacos
18.
Brain Res ; 1601: 85-91, 2015 Mar 19.
Artículo en Inglés | MEDLINE | ID: mdl-25598204

RESUMEN

Increasing evidence indicates that poor outcomes after brain hemorrhage, especially after subarachnoid hemorrhage (SAH), can be attributed largely to dysfunction of the cerebral microcirculation. However, the cause of this dysfunction remains unclear. Here, we investigated changes in the cerebral microcirculation after regional hemorrhage in the subarachnoid space using the closed cranial window technique in mice. A single pial arteriole on the surface of the brain was punctured to induce a regional hemorrhage in the subarachnoid space. Physiological parameters were monitored during the procedure, and microvessel diameter was measured after hemorrhage. The vasoreactivity of the arterioles in response to hypercapnia as well as to topical application of the vasodilator acetylcholine (ACh) and S-nitroso-N-acetyl-penicillamine (SNAP) were assessed. The constriction of pial arterioles was detected without changes in other physiological parameters. Decreased reactivity of pial arterioles to all of the applied vasodilatory stimuli was observed after hemorrhage. Our results indicate that regional hemorrhage in the subarachnoid space can induce the vasospasm of microvessels and also reduce the vasoreactivity of pial arterioles.


Asunto(s)
Piamadre/irrigación sanguínea , Piamadre/patología , Piamadre/fisiopatología , Hemorragia Subaracnoidea/patología , Hemorragia Subaracnoidea/fisiopatología , Acetilcolina/farmacología , Animales , Arteriolas/efectos de los fármacos , Arteriolas/patología , Arteriolas/fisiopatología , Hipercapnia/fisiopatología , Masculino , Ratones , Ratones Endogámicos C57BL , Microvasos/fisiopatología , Lóbulo Parietal/efectos de los fármacos , Lóbulo Parietal/fisiopatología , Piamadre/efectos de los fármacos , S-Nitroso-N-Acetilpenicilamina/farmacología
19.
Biol Trace Elem Res ; 165(2): 167-72, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25645362

RESUMEN

Cadmium, a heavy metal, is a toxic environmental and industrial pollutant. Exposure to cadmium can lead to the toxic effects in a variety of tissues, also including the brain. The present study investigated the effect of cadmium exposure on the histopathology of cerebral cortex in juvenile mice. Juvenile mice were randomly divided into control, low (1.87 mg/kg), medium (3.74 mg/kg), and high (7.48 mg/kg) dose groups. After cadmium exposure by drinking water for 10 days, the cerebral cortex was obtained for histopathology studies. The medium and high dose of cadmium, rather than low dose, could induce the histopathology alterations of cerebral cortex in a dose-dependent manner. In the high-dose group, microstructure significantly showed pia mater encephali divorcing from cerebral cortex layer, serious hyperemia of blood capillary in pia mater encephali and cerebral cortex, broadening vessel peripheral clearance, a large number of eosinophil leukocyte infiltrating around blood vessel, vacuolar degeneration in part granule cells, and obviously increasing apoptotic cells. Ultrastructure obviously displayed marginalized heterochromatin, incomplete or fused nuclear membranes, broadened perinuclear space, ambiguous mitochondria cristae, decreased synaptic cleft, and fused presynaptic and postsynaptic membrane. Our results revealed that cadmium at the middle and high dose could induce obvious microstructure and ultrastructure alterations of cerebral cortex in juvenile mice, which may be one important mechanism of cadmium neurotoxicity.


Asunto(s)
Cadmio/toxicidad , Corteza Cerebral/patología , Administración Oral , Animales , Apoptosis , Corteza Cerebral/efectos de los fármacos , Corteza Cerebral/ultraestructura , Relación Dosis-Respuesta a Droga , Eosinófilos/metabolismo , Femenino , Lóbulo Frontal/ultraestructura , Heterocromatina/metabolismo , Hiperemia/inducido químicamente , Leucocitos/metabolismo , Masculino , Ratones , Microscopía Electrónica de Transmisión , Mitocondrias/efectos de los fármacos , Neuronas/metabolismo , Piamadre/efectos de los fármacos , Sinapsis/efectos de los fármacos
20.
Brain Res ; 1603: 141-9, 2015 Apr 07.
Artículo en Inglés | MEDLINE | ID: mdl-25662771

RESUMEN

Our previous findings indicated that in rats subjected to subarachnoid hemorrhage (SAH), suppression of post-SAH neuroinflammation via vascular adhesion protein-1 (VAP-1) blockade provides significant neuroprotection. We and others have reported that neuroinflammation contributes to cerebral microvascular impairment. Thus, in the present study, we tested the hypotheses that: (1) treatment with LJP-1586, a selective VAP-1 blocker, prevents SAH-associated pial arteriolar dilating dysfunction; and (2) the vasculoprotective effect of LJP-1586 arises from inhibiting SAH-elicited neutrophil recruitment. We utilized an endovascular perforation model of SAH. Rats subjected to SAH were either treated with LJP-1586 or rendered neutropenic via anti-neutrophil-antibody treatment. Findings from these groups were compared to their respective control groups. At 48 h post-SAH, rats were evaluated for neurobehavioral function, pial venular leukocyte trafficking, and pial arteriolar reactivity to topically-applied acetylcholine (ACh) and S-nitroso-N-acetyl penicillamine (SNAP). Pial arteriolar responses decreased at 48 h post-SAH. However, in the presence of LJP-1586, those responses were significantly preserved. Neutrophil-depletion yielded a substantial suppression of SAH-associated leukocyte adhesion and infiltration. This was accompanied by a significant preservation of pial arteriolar dilating function, suggesting a direct link between neutrophil recruitment and the loss of cerebral microvascular reactivity. Moreover, neutrophil depletion also was associated with significant protection of neurobehavioral function. The present findings suggest that attenuating SAH-linked elevation in neutrophil trafficking will protect against the development of microvascular dysfunction and subsequent neurological impairment.


Asunto(s)
Alilamina/análogos & derivados , Amina Oxidasa (conteniendo Cobre)/antagonistas & inhibidores , Fármacos Cardiovasculares/farmacología , Moléculas de Adhesión Celular/antagonistas & inhibidores , Infiltración Neutrófila/efectos de los fármacos , Hemorragia Subaracnoidea/tratamiento farmacológico , Acetilcolina/farmacología , Alilamina/farmacología , Amina Oxidasa (conteniendo Cobre)/metabolismo , Animales , Arteriolas/efectos de los fármacos , Arteriolas/fisiopatología , Moléculas de Adhesión Celular/metabolismo , Circulación Cerebrovascular/efectos de los fármacos , Circulación Cerebrovascular/fisiología , Agonistas Colinérgicos/farmacología , Modelos Animales de Enfermedad , Leucocitos/efectos de los fármacos , Leucocitos/fisiología , Masculino , Neuroinmunomodulación/efectos de los fármacos , Neuroinmunomodulación/fisiología , Infiltración Neutrófila/fisiología , Neutrófilos/efectos de los fármacos , Neutrófilos/fisiología , Donantes de Óxido Nítrico/farmacología , Piamadre/irrigación sanguínea , Piamadre/efectos de los fármacos , Piamadre/fisiopatología , Ratas Sprague-Dawley , Flujo Sanguíneo Regional/efectos de los fármacos , Flujo Sanguíneo Regional/fisiología , S-Nitroso-N-Acetilpenicilamina/farmacología , Hemorragia Subaracnoidea/mortalidad , Hemorragia Subaracnoidea/fisiopatología , Vénulas/efectos de los fármacos , Vénulas/fisiopatología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA