Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 27
Filtrar
1.
J Neurochem ; 140(5): 814-825, 2017 03.
Artículo en Inglés | MEDLINE | ID: mdl-28002622

RESUMEN

Epoxyeicosatrienoic acids (EETs) are synthesized in astrocytes, and inhibitors of soluble epoxide hydrolase (sEH), which hydrolyzes EETs, reduce infarct volume in ischemic stroke. Astrocytes can release protective neurotrophic factors, such as vascular endothelial growth factor (VEGF). We found that addition of sEH inhibitors to rat cultured astrocytes immediately after oxygen-glucose deprivation (OGD) markedly increased VEGF concentration in the medium 48 h later and the effect was blocked by an EET antagonist. The sEH inhibitors increased EET concentrations to levels capable of increasing VEGF. When the sEH inhibitors were removed from the medium at 48 h, the increase in VEGF persisted for an additional 48 h. Neurons exposed to OGD and subsequently to astrocyte medium previously conditioned with OGD plus sEH inhibitors showed increased phosphorylation of their VEGF receptor-2, less TUNEL staining, and increased phosphorylation of Akt, which was blocked by a VEGF receptor-2 antagonist. Our findings indicate that sEH inhibitors, applied to cultured astrocytes after an ischemia-like insult, can increase VEGF secretion. The released VEGF then enhances Akt-enabled cell survival signaling in neurons through activation of VEGF receptor-2 leading to less neuronal cell death. These results suggest a new strategy by which astrocytes can be leveraged to support neuroprotection.


Asunto(s)
Astrocitos/metabolismo , Hipoxia de la Célula/efectos de los fármacos , Inhibidores Enzimáticos/farmacología , Epóxido Hidrolasas/antagonistas & inhibidores , Glucosa/deficiencia , Factor A de Crecimiento Endotelial Vascular/metabolismo , Animales , Astrocitos/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Medios de Cultivo Condicionados , Femenino , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Proteína Oncogénica v-akt/metabolismo , Fosforilación , Embarazo , Ratas , Ratas Sprague-Dawley , Receptores de Factores de Crecimiento Endotelial Vascular/antagonistas & inhibidores , Receptores de Factores de Crecimiento Endotelial Vascular/metabolismo , Transducción de Señal/efectos de los fármacos
2.
Artículo en Inglés | MEDLINE | ID: mdl-27174801

RESUMEN

Astrocytes secrete vasodilator and vasoconstrictor factors via end feet processes, altering blood flow to meet neuronal metabolic demand. Compared to what is known about the ability of astrocytes to release factors that dilate local cerebral vasculature, very little is known regarding the source and identity of astrocyte derived constricting factors. The present study investigated if astrocytes express CYP 4A ω-hydroxylase and metabolize arachidonic acid (AA) to 20-hydroxyeicotetraenoic acid (20-HETE) that regulates KCa channel activity in astrocytes and cerebral arterial myocyte contractility. Here we report that cultured astrocytes express CYP 4A2/3 ω-hydroxylase mRNA and CYP 4A protein and produce 20-HETE and the CYP epoxygenase metabolites epoxyeicosatrienoic acids (EETs) when incubated with AA. The production of 20-HETE and EETs was enhanced following stimulation of metabotropic glutamate receptors (mGluR) on the astrocytes. Exogenous application of 20-HETE attenuated, whereas inhibition of 20-HETE production with HET-0016 increased the open state probabilities (NPo) of 71pS and 161pS KCa single-channel currents recorded from astrocytes. Exposure of isolated cerebral arterial myocytes to conditioned media from cultured astrocytes caused shortening of the length of freshly isolated cerebral arterial myocytes that was not evident following inhibition of astrocyte 20-HETE synthesis and action. These findings suggest that astrocytes not only release vasodilator EETs in response to mGluR stimulation but also synthetize and release the cerebral arterial myocyte constrictor 20-HETE that also functions as an endogenous inhibitor of the activity of two types of KCa channel currents found in astrocytes.


Asunto(s)
Astrocitos/metabolismo , Sistema Enzimático del Citocromo P-450/biosíntesis , Ácidos Hidroxieicosatetraenoicos/biosíntesis , Receptores de Glutamato Metabotrópico/metabolismo , Animales , Ácido Araquidónico/administración & dosificación , Ácido Araquidónico/metabolismo , Astrocitos/efectos de los fármacos , Astrocitos/enzimología , Encéfalo/metabolismo , Circulación Cerebrovascular/genética , Sistema Enzimático del Citocromo P-450/genética , Regulación Enzimológica de la Expresión Génica , Ácidos Hidroxieicosatetraenoicos/metabolismo , Músculo Liso Vascular/metabolismo , Ratas , Receptores de Glutamato Metabotrópico/genética
3.
Am J Physiol Heart Circ Physiol ; 306(7): H989-H1000, 2014 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-24464756

RESUMEN

Recent studies have indicated that the myogenic response (MR) in cerebral arteries is impaired in Fawn Hooded Hypertensive (FHH) rats and that transfer of a 2.4 megabase pair region of chromosome 1 (RNO1) containing 15 genes from the Brown Norway rat into the FHH genetic background restores MR in a FHH.1(BN) congenic strain. However, the mechanisms involved remain to be determined. The present study examined the role of the large conductance calcium-activated potassium (BK) channel in impairing the MR in FHH rats. Whole-cell patch-clamp studies of cerebral vascular smooth muscle cells (VSMCs) revealed that iberiotoxin (IBTX; BK inhibitor)-sensitive outward potassium (K+) channel current densities are four- to fivefold greater in FHH than in FHH.1(BN) congenic strain. Inside-out patches indicated that the BK channel open probability (NPo) is 10-fold higher and IBTX reduced NPo to a greater extent in VSMCs isolated from FHH than in FHH.1(BN) rats. Voltage sensitivity of the BK channel is enhanced in FHH as compared with FHH.1(BN) rats. The frequency and amplitude of spontaneous transient outward currents are significantly greater in VSMCs isolated from FHH than in FHH.1(BN) rats. However, the expression of the BK-α and -ß-subunit proteins in cerebral vessels as determined by Western blot is similar between the two groups. Middle cerebral arteries (MCAs) isolated from FHH rats exhibited an impaired MR, and administration of IBTX restored this response. These results indicate that there is a gene on RNO1 that impairs MR in the MCAs of FHH rats by enhancing BK channel activity.


Asunto(s)
Circulación Cerebrovascular , Hipertensión/metabolismo , Canales de Potasio de Gran Conductancia Activados por el Calcio/metabolismo , Músculo Liso Vascular/metabolismo , Vasoconstricción , Animales , Animales Congénicos , Señalización del Calcio , Modelos Animales de Enfermedad , Hipertensión/genética , Hipertensión/fisiopatología , Activación del Canal Iónico , Subunidades alfa de los Canales de Potasio de Gran Conductancia Activados por Calcio/metabolismo , Subunidades beta de los Canales de Potasio de Gran Conductancia Activados por el Calcio/metabolismo , Canales de Potasio de Gran Conductancia Activados por el Calcio/antagonistas & inhibidores , Masculino , Potenciales de la Membrana , Arteria Cerebral Media/metabolismo , Arteria Cerebral Media/fisiopatología , Músculo Liso Vascular/efectos de los fármacos , Músculo Liso Vascular/fisiopatología , Miocitos del Músculo Liso/metabolismo , Técnicas de Placa-Clamp , Bloqueadores de los Canales de Potasio/farmacología , Ratas , Ratas Endogámicas BN
4.
Circ Res ; 110(3): 471-80, 2012 Feb 03.
Artículo en Inglés | MEDLINE | ID: mdl-22158710

RESUMEN

RATIONALE: Hydrogen peroxide (H(2)O(2)) serves as a key endothelium-derived hyperpolarizing factor mediating flow-induced dilation in human coronary arterioles (HCAs). The precise mechanisms by which H(2)O(2) elicits smooth muscle hyperpolarization are not well understood. An important mode of action of H(2)O(2) involves the oxidation of cysteine residues in its target proteins, including protein kinase G (PKG)-Iα, thereby modulating their activities. OBJECTIVE: Here we hypothesize that H(2)O(2) dilates HCAs through direct oxidation and activation of PKG-Iα leading to the opening of the large-conductance Ca(2+)-activated K(+) (BK(Ca)) channel and subsequent smooth muscle hyperpolarization. METHODS AND RESULTS: Flow and H(2)O(2) induced pressure gradient/concentration-dependent vasodilation in isolated endothelium-intact and -denuded HCAs, respectively. The dilation was largely abolished by iberiotoxin, a BK(Ca) channel blocker. The PKG inhibitor Rp-8-Br-PET-cGMP also markedly inhibited flow- and H(2)O(2)-induced dilation, whereas the soluble guanylate cyclase inhibitor ODQ had no effect. Treatment of coronary smooth muscle cells (SMCs) with H(2)O(2) elicited dose-dependent, reversible dimerization of PKG-Iα, and induced its translocation to the plasma membrane. Patch-clamp analysis identified a paxilline-sensitive single-channel K(+) current with a unitary conductance of 246-pS in freshly isolated coronary SMCs. Addition of H(2)O(2) into the bath solution significantly increased the probability of BK(Ca) single-channel openings recorded from cell-attached patches, an effect that was blocked by the PKG-Iα inhibitor DT-2. H(2)O(2) exhibited an attenuated stimulatory effect on BK(Ca) channel open probability in inside-out membrane patches. CONCLUSIONS: H(2)O(2) dilates HCAs through a novel mechanism involving protein dimerization and activation of PKG-Iα and subsequent opening of smooth muscle BK(Ca) channels.


Asunto(s)
Arteriolas/fisiología , Vasos Coronarios/fisiología , Proteínas Quinasas Dependientes de GMP Cíclico/fisiología , Dimerización , Peróxido de Hidrógeno/farmacología , Canales de Potasio de Gran Conductancia Activados por el Calcio/fisiología , Vasodilatación/efectos de los fármacos , Arteriolas/efectos de los fármacos , Células Cultivadas , Vasos Coronarios/efectos de los fármacos , Proteínas Quinasas Dependientes de GMP Cíclico/antagonistas & inhibidores , Relación Dosis-Respuesta a Droga , Endotelio Vascular/citología , Endotelio Vascular/efectos de los fármacos , Endotelio Vascular/fisiología , Inhibidores Enzimáticos/farmacología , Fluoresceínas/farmacología , Humanos , Músculo Liso Vascular/citología , Músculo Liso Vascular/efectos de los fármacos , Músculo Liso Vascular/fisiología , Técnicas de Placa-Clamp , Fragmentos de Péptidos/farmacología , Péptidos/farmacología , Vasodilatación/fisiología
5.
Am J Physiol Heart Circ Physiol ; 302(6): H1285-93, 2012 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-22245774

RESUMEN

Recent studies have indicated that inhibitors of the synthesis of 20-hydroxyeicosatetraenoic acid (20-HETE) may have direct neuroprotective actions since they reduce infarct volume after ischemia reperfusion in the brain without altering blood flow. To explore this possibility, the present study used organotypic hippocampal slice cultures subjected to oxygen-glucose deprivation (OGD) and reoxygenation to examine whether 20-HETE is released by organotypic hippocampal slices after OGD and whether it contributes to neuronal death through the generation of ROS and activation of caspase-3. The production of 20-HETE increased twofold after OGD and reoxygenation. Blockade of the synthesis of 20-HETE with N-hydroxy-N'-(4-butyl-2-methylphenol)formamidine (HET0016) or its actions with a 20-HETE antagonist, 20-hydroxyeicosa-6(Z),15(Z)-dienoic acid, reduced cell death, as measured by the release of lactate dehydrogenase and propidium iodide uptake. Administration of a 20-HETE mimetic, 20-hydroxyeicosa-5(Z),14(Z)-dienoic acid (5,14-20-HEDE), had the opposite effect and increased injury after OGD. The death of neurons after OGD was associated with an increase in the production of ROS and activation of caspase-3. These effects were attenuated by HET0016 and potentiated after the administration of 5,14-20-HEDE. These findings indicate that the production of 20-HETE by hippocampal slices is increased after OGD and that inhibitors of the synthesis or actions of 20-HETE protect neurons from ischemic cell death. The protective effect of 20-HETE inhibitors is associated with a decrease in superoxide production and activation of caspase-3.


Asunto(s)
Amidinas/farmacología , Glucosa/deficiencia , Hipocampo/efectos de los fármacos , Ácidos Hidroxieicosatetraenoicos/antagonistas & inhibidores , Ácidos Hidroxieicosatetraenoicos/farmacología , Fármacos Neuroprotectores/farmacología , Animales , Animales Recién Nacidos , Caspasa 3/metabolismo , Muerte Celular/efectos de los fármacos , Hipoxia de la Célula , Citoprotección , Hipocampo/metabolismo , Hipocampo/patología , Ácidos Hidroxieicosatetraenoicos/metabolismo , Ratas , Ratas Sprague-Dawley , Superóxidos/metabolismo , Factores de Tiempo , Técnicas de Cultivo de Tejidos
6.
Am J Physiol Heart Circ Physiol ; 300(5): H1557-65, 2011 May.
Artículo en Inglés | MEDLINE | ID: mdl-21257913

RESUMEN

While myogenic force in response to a changing arterial pressure has been described early in the 20th century, it was not until 1984 that the effect of a sequential increase in intraluminal pressure on cannulated cerebral arterial preparations was found to result in pressure-dependent membrane depolarization associated with spike generation and reduction in lumen diameter. Despite a great deal of effort by different laboratories and investigators, the identification of the existence of a mediator of the pressure-induced myogenic constriction in arterial muscle remained a challenge. It was the original finding by our laboratory that demonstrated the capacity of cerebral arterial muscle cells to express the cytochrome P-450 4A enzyme that catalyzes the formation of the potent vasoconstrictor 20-hydroxyeicosatetraenoic acid (20-HETE) from arachidonic acid, the production of which in cerebral arterial muscle cells increases with the elevation in intravascular pressure. 20-HETE activates protein kinase C and causes the inhibition of Ca(²+)-activated K(+) channels, depolarizes arterial muscle cell membrane, and activates L-type Ca(²+) channel to increase intracellular Ca(²+) levels and evoke vasoconstriction. The inhibition of 20-HETE formation attenuates pressure-induced arterial myogenic constriction in vitro and blunts the autoregulation of cerebral blood flow in vivo. We suggest that the formation and action of cytochrome P-450-derived 20-HETE in cerebral arterial muscle could play a critically important role in the control of cerebral arterial tone and the autoregulation of cerebral blood flow under physiological conditions.


Asunto(s)
Circulación Cerebrovascular/fisiología , Homeostasis/fisiología , Ácidos Hidroxieicosatetraenoicos/fisiología , Vasoconstricción/fisiología , Animales , Humanos , Canales Iónicos/fisiología , Músculo Liso Vascular/fisiología , Flujo Sanguíneo Regional/fisiología
7.
Am J Physiol Heart Circ Physiol ; 301(2): H373-81, 2011 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-21602473

RESUMEN

Group I metabotropic glutamate receptors (mGluR) on astrocytes have been shown to participate in cerebral vasodilation to neuronal activation in brain slices. Pharmacological stimulation of mGluR in brain slices can produce arteriolar constriction or dilation depending on the initial degree of vascular tone. Here, we examined whether pharmacological stimulation of mGluR in vivo increases cerebral blood flow. A 1-mM solution of the group I mGluR agonist (S)-3,5-dihydroxyphenylglycine (DHPG) superfused at 5 µl/min over the cortical surface of anesthetized rats produced a 30 ± 2% (±SE) increase in blood flow measured by laser-Doppler flowmetry after 15-20 min. The response was completely blocked by superfusion of group I mGluR antagonists and attenuated by superfusion of an epoxyeicosatrienoic acid (EET) antagonist (5 ± 4%), an EET synthesis inhibitor (11 ± 3%), and a cyclooxygenase-2 inhibitor (15 ± 3%). The peak blood flow response was not significantly affected by administration of inhibitors of cyclooxygenase-1, neuronal nitric oxide synthase, heme oxygenase, adenosine A(2B) receptors, or an inhibitor of the synthesis of 20-hydroxyeicosatetraenoic acid (20-HETE). The blood flow response gradually waned following 30-60 min of DHPG superfusion. This loss of the flow response was attenuated by a 20-HETE synthesis inhibitor and was prevented by superfusion of an inhibitor of epoxide hydrolase, which hydrolyzes EETs. These results indicate that pharmacological stimulation of mGluR in vivo increases cerebral blood flow and that the response depends on the release of EETs and a metabolite of cyclooxygenase-2. Epoxide hydrolase activity and 20-HETE synthesis limit the duration of the response to prolonged mGluR activation.


Asunto(s)
Arterias Cerebrales/efectos de los fármacos , Corteza Cerebral/irrigación sanguínea , Circulación Cerebrovascular/efectos de los fármacos , Agonistas de Aminoácidos Excitadores/farmacología , Glicina/análogos & derivados , Ácidos Hidroxieicosatetraenoicos/metabolismo , Receptores de Glutamato Metabotrópico/agonistas , Resorcinoles/farmacología , Vasodilatación/efectos de los fármacos , Vasodilatadores/farmacología , Ácido 8,11,14-Eicosatrienoico/análogos & derivados , Ácido 8,11,14-Eicosatrienoico/farmacología , Antagonistas del Receptor de Adenosina A2/farmacología , Análisis de Varianza , Animales , Arteriolas/efectos de los fármacos , Arteriolas/metabolismo , Velocidad del Flujo Sanguíneo/efectos de los fármacos , Arterias Cerebrales/metabolismo , Ciclooxigenasa 2/metabolismo , Inhibidores de la Ciclooxigenasa 2/farmacología , Epóxido Hidrolasas/antagonistas & inhibidores , Epóxido Hidrolasas/metabolismo , Antagonistas de Aminoácidos Excitadores/farmacología , Glicina/farmacología , Hemo Oxigenasa (Desciclizante)/antagonistas & inhibidores , Hemo Oxigenasa (Desciclizante)/metabolismo , Ácidos Hidroxieicosatetraenoicos/antagonistas & inhibidores , Flujometría por Láser-Doppler , Masculino , Óxido Nítrico Sintasa/antagonistas & inhibidores , Óxido Nítrico Sintasa/metabolismo , Óxido Nítrico Sintasa de Tipo I , Ratas , Ratas Wistar , Receptor de Adenosina A2B/efectos de los fármacos , Receptor de Adenosina A2B/metabolismo , Receptores de Glutamato Metabotrópico/antagonistas & inhibidores , Receptores de Glutamato Metabotrópico/metabolismo , Flujo Sanguíneo Regional/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Factores de Tiempo
8.
Artículo en Inglés | MEDLINE | ID: mdl-32404236

RESUMEN

The present study assessed the effect of nearby construction activity on the responses of rat middle cerebral arteries (MCA)to the endothelium-dependent vasodilator acetylcholine and the NO donor sodium nitroprusside (SNP) and the activity of MaxiK potassium channels in MCA smooth muscle cells from male Sprague-Dawley rats. Two monitoring systems were used to assess vibrations in the animal rooms during and immediately after construction activities near the research building where the animal facility is located. One was a commercially available system; the other was a Raspberry-Pi (RPi)-based vibration monitoring system designed in our laboratory that included a small computing unit attached to a rolling sensor (low sensitivity) and a piezoelectric film sensor (high sensitivity). Both systems recorded increased levels of vibration during construction activity outside the building. During the construction period, vasodilator responses to acetylcholine and SNP were abolished, and MaxiK single-channel current opening frequency and open-state probability in cell-attached patches of isolated MCA myocytes were dramatically decreased. Recovery of acetylcholine- and SNP-induced dilation was minimal in MCA from rats studied after completion of construction but housed in the animal facility during construction, whereas responses to acetylcholine and SNP were intact in rats purchased, housed, and studied after construction. Baseline levels of vibration returned after the completion of construction, concomitant with the recovery of normal endothelium-dependent vasodilation to acetylcholine and of NO sensitivity assessed by using SNP in MCA from animals obtained after construction. The results of this study indicate that the vibration associated with nearby construction can have highly disruptive effects on crucial physiologic phenotypes.

9.
J Cereb Blood Flow Metab ; 28(1): 111-25, 2008 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-17519974

RESUMEN

Adenosine, astrocyte metabotropic glutamate receptors (mGluRs), and epoxyeicosatrienoic acids (EETs) have been implicated in neurovascular coupling. Although A(2A) and A(2B) receptors mediate cerebral vasodilation to adenosine, the role of each receptor in the cerebral blood flow (CBF) response to neural activation remains to be fully elucidated. In addition, adenosine can amplify astrocyte calcium, which may increase arachidonic acid metabolites such as EETs. The interaction of these pathways was investigated by determining if combined treatment with antagonists exerted an additive inhibitory effect on the CBF response. During whisker stimulation of anesthetized rats, the increase in cortical CBF was reduced by approximately half after individual administration of A(2B), mGluR and EET antagonists and EET synthesis inhibitors. Combining treatment of either a mGluR antagonist, an EET antagonist, or an EET synthesis inhibitor with an A(2B) receptor antagonist did not produce an additional decrement in the CBF response. Likewise, the CBF response also remained reduced by approximately 50% when an EET antagonist was combined with an mGluR antagonist or an mGluR antagonist plus an A(2B) receptor antagonist. In contrast, A(2A) and A(3) receptor antagonists had no effect on the CBF response to whisker stimulation. We conclude that (1) adenosine A(2B) receptors, rather than A(2A) or A(3) receptors, play a significant role in coupling cortical CBF to neuronal activity, and (2) the adenosine A(2B) receptor, mGluR, and EETs signaling pathways are not functionally additive, consistent with the possibility of astrocytic mGluR and adenosine A(2B) receptor linkage to the synthesis and release of vasodilatory EETs.


Asunto(s)
Ácido 8,11,14-Eicosatrienoico/análogos & derivados , Ácido 8,11,14-Eicosatrienoico/metabolismo , Ácidos Araquidónicos/metabolismo , Corteza Cerebelosa/irrigación sanguínea , Unión Neuroefectora/metabolismo , Receptores de Glutamato Metabotrópico/metabolismo , Receptores Purinérgicos P1/metabolismo , Vasodilatación/fisiología , Vasodilatadores/metabolismo , Vibrisas , Animales , Corteza Cerebelosa/metabolismo , Circulación Cerebrovascular/efectos de los fármacos , Circulación Cerebrovascular/fisiología , Masculino , Antagonistas de Receptores Purinérgicos P1 , Ratas , Ratas Wistar , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología , Vasodilatación/efectos de los fármacos
10.
Compr Physiol ; 8(2): 801-821, 2018 03 25.
Artículo en Inglés | MEDLINE | ID: mdl-29687906

RESUMEN

There have been numerous reviews related to the cerebral circulation. Most of these reviews are similar in many ways. In the present review, we thought it important to provide an overview of function with specific attention to details of cerebral arterial control related to brain homeostasis, maintenance of neuronal energy demands, and a unique perspective related to the role of astrocytes. A coming review in this series will discuss cerebral vascular development and unique properties of the neonatal circulation and developing brain, thus, many aspects of development are missing here. Similarly, a review of the response of the brain and cerebral circulation to heat stress has recently appeared in this series (8). By trying to make this review unique, some obvious topics were not discussed in lieu of others, which are from recent and provocative research such as endothelium-derived hyperpolarizing factor, circadian regulation of proteins effecting cerebral blood flow, and unique properties of the neurovascular unit. © 2018 American Physiological Society. Compr Physiol 8:801-821, 2018.


Asunto(s)
Circulación Cerebrovascular/fisiología , Sistema Enzimático del Citocromo P-450/fisiología , Metabolismo de los Lípidos/fisiología , Ácido Araquidónico/metabolismo , Astrocitos/metabolismo , Presión Sanguínea/fisiología , Hipoxia de la Célula/fisiología , Relojes Circadianos/fisiología , Homeostasis/fisiología , Humanos , Potenciales de la Membrana/fisiología , Neovascularización Patológica/fisiopatología
11.
PLoS One ; 12(5): e0176796, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28472069

RESUMEN

The transient receptor potential vallinoid type 4 (TRPV4) is a calcium entry channel known to modulate vascular function by mediating endothelium-dependent vasodilation. The present study investigated if isolated cerebral arterial myocytes of the Fawn Hooded hypertensive (FHH) rat, known to display exaggerated KCa channel current activity and impaired myogenic tone, express TRPV4 channels at the transcript and protein level and exhibit TRPV4-like single-channel cationic current activity. Reverse transcription polymerase chain reaction (RT-PCR), Western blot, and immunostaining analysis detected the expression of mRNA transcript and translated protein of TRPV4 channel in FHH rat cerebral arterial myocytes. Patch clamp recording of single-channel current activity identified the presence of a single-channel cationic current with unitary conductance of ~85 pS and ~96 pS at hyperpolarizing and depolarizing potentials, respectively, that was inhibited by the TRPV4 channel antagonist RN 1734 or HC 067074 and activated by the potent TRPV4 channel agonist GSK1016790A. Application of negative pressure via the interior of the patch pipette increased the NPo of the TRPV4-like single-channel cationic current recorded in cell-attached patches at a patch potential of 60 mV that was inhibited by prior application of the TRPV4 channel antagonist RN 1734 or HC 067047. Treatment with the TRPV4 channel agonist GSK1016790A caused concentration-dependent increase in the NPo of KCa single-channel current recorded in cell-attached patches of cerebral arterial myocytes at a patch potential of 40 mV, which was not influenced by pretreatment with the voltage-gated L-type Ca2+ channel blocker nifedipine or the T-type Ca2+ channel blocker Ni2+. These findings demonstrate that FHH rat cerebral arterial myocytes express mRNA transcript and translated protein for TRPV4 channel and display TRPV4-like single-channel cationic current activity that was stretch-sensitive and activation of which increased the open state probability of KCa single-channel current in these arterial myocytes.


Asunto(s)
Encéfalo/irrigación sanguínea , Circulación Cerebrovascular , Hipertensión/fisiopatología , Músculo Liso Vascular/fisiopatología , Canales Catiónicos TRPV/fisiología , Animales , Masculino , Ratas
12.
J Appl Physiol (1985) ; 100(1): 307-17, 2006 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-16357084

RESUMEN

Astrocytes send processes to synapses and blood vessels, communicate with other astrocytes through gap junctions and by release of ATP, and thus are an integral component of the neurovascular unit. Electrical field stimulations in brain slices demonstrate an increase in intracellular calcium in astrocyte cell bodies transmitted to perivascular end-feet, followed by a decrease in vascular smooth muscle calcium oscillations and arteriolar dilation. The increase in astrocyte calcium after neuronal activation is mediated, in part, by activation of metabotropic glutamate receptors. Calcium signaling in vitro can also be influenced by adenosine acting on A2B receptors and by epoxyeicosatrienoic acids (EETs) shown to be synthesized in astrocytes. Prostaglandins, EETs, arachidonic acid, and potassium ions are candidate mediators of communication between astrocyte end-feet and vascular smooth muscle. In vivo evidence supports a role for cyclooxygenase-2 metabolites, EETs, adenosine, and neuronally derived nitric oxide in the coupling of increased blood flow to increased neuronal activity. Combined inhibition of the EETs, nitric oxide, and adenosine pathways indicates that signaling is not by parallel, independent pathways. Indirect pharmacological results are consistent with astrocytes acting as intermediaries in neurovascular signaling within the neurovascular unit. For specific stimuli, astrocytes are also capable of transmitting signals to pial arterioles on the brain surface for ensuring adequate inflow pressure to parenchymal feeding arterioles. Therefore, evidence from brain slices and indirect evidence in vivo with pharmacological approaches suggest that astrocytes play a pivotal role in regulating the fundamental physiological response coupling dynamic changes in cerebral blood flow to neuronal synaptic activity. Future work using in vivo imaging and genetic manipulation will be required to provide more direct evidence for a role of astrocytes in neurovascular coupling.


Asunto(s)
Astrocitos/fisiología , Encéfalo/irrigación sanguínea , Encéfalo/fisiología , Circulación Cerebrovascular/fisiología , Músculo Liso Vascular/fisiología , Neuronas/fisiología , Sistema Vasomotor/fisiología , Animales , Retroalimentación/fisiología , Humanos , Músculo Liso Vascular/inervación
13.
Pediatr Clin North Am ; 53(5): 1029-37, xi, 2006 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-17027622

RESUMEN

Reactive oxygen species (ROS) are a family of oxygen-derived free radicals that are produced in mammalian cells under normal and pathologic conditions. Many ROS, such as the superoxide anion (O2-) and hydrogen peroxide (H2O2), act as cellular signaling molecules within blood vessels, altering mechanisms mediating mechanical signal transduction and autoregulation of cerebral blood flow. This article focuses on the actions of ROS, such as O2.- and H2O2, and how they influence mechanisms responsible for the modulation of pressure-induced myogenic tone in the cerebral circulation and blood flow autoregulation in response to elevated arterial pressure. ROS may be a key target for therapeutic interventions in pediatric patients who have hypoxic injury or altered cerebral metabolism induced by trauma or infection.


Asunto(s)
Circulación Cerebrovascular/efectos de los fármacos , Circulación Cerebrovascular/fisiología , Especies Reactivas de Oxígeno/metabolismo , Especies Reactivas de Oxígeno/farmacología , Niño , Sistema Enzimático del Citocromo P-450/fisiología , Humanos , Canales Iónicos/efectos de los fármacos , Especies Reactivas de Oxígeno/química , Transducción de Señal
14.
J Neurosci ; 23(5): 1678-87, 2003 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-12629172

RESUMEN

The influence of activation of glutamate receptor (GluR) on outward K(+) current in cultured neonate rat hippocampal astrocytes was investigated. Patch-clamp analysis of K(+) channel currents in cultured astrocytes identified the existence of 71 +/- 6 and 161 +/- 11 pS single-channel K(+) currents that were sensitive to changes in voltage and [Ca(2+)](i) and blocked by external TEA but not by charybdotoxin, iberiotoxin, apamin, or 4-aminopyridine. Reverse transcriptase (RT)-PCR and Northern blot analysis revealed transcripts of the Ca(2+)-activated K(+) channel (K(Ca)) beta(4)-subunit (beta4) (KCNMB4) in cultured astrocytes. Expression of the metabotropic glutamate receptor (mGluR) subtypes mGluR1 and mGluR5 and the ionotropic glutamate receptor (iGluR) subtypes iGluR1 and iGluR4 were detected by RT-PCR and immunofluorescence analysis in cultured astrocytes. The mGluR agonists L-glutamate and quisqualate increased the open state probability (NP(o)) of the 71 and 161 pS K(+) channel currents that were prevented by the mGluR receptor antagonists 1-aminoindan-1,5-dicarboxylic acid or L-(+)-2-amino-3-phosphonopropionic acid and not by the iGluR antagonists (+)-5-methyl-10,11-dihydro-5H-dibenzo [a,d] cyclohepten-5,10-imine maleate or CNQX. Activation of the two types of K(+) channel currents by mGluR agonists was attenuated by pertussis toxin and by inhibition of phospholipase C (PLC) or cytochrome P450 arachidonate epoxygenase. These results indicate that brain astrocytes contain the KCNMB4 transcript and express two novel types of K(Ca) channels that are gated by activation of a G-protein coupled metabotropic glutamate receptor functionally linked to PLC and cytochrome P450 arachidonate epoxygenase activity.


Asunto(s)
Astrocitos/metabolismo , Canales de Potasio Calcio-Activados/metabolismo , Receptores de Glutamato Metabotrópico/metabolismo , Animales , Astrocitos/citología , Astrocitos/efectos de los fármacos , Calcio/metabolismo , Calcio/farmacología , Células Cultivadas , Citocromo P-450 CYP2J2 , Inhibidores Enzimáticos del Citocromo P-450 , Sistema Enzimático del Citocromo P-450 , Inhibidores Enzimáticos/farmacología , Agonistas de Aminoácidos Excitadores/farmacología , Antagonistas de Aminoácidos Excitadores/farmacología , Hipocampo/citología , Subunidades beta de los Canales de Potasio de Gran Conductancia Activados por el Calcio , Potenciales de la Membrana/efectos de los fármacos , Potenciales de la Membrana/fisiología , Proteínas del Tejido Nervioso/antagonistas & inhibidores , Proteínas del Tejido Nervioso/metabolismo , Oxigenasas/antagonistas & inhibidores , Técnicas de Placa-Clamp , Toxina del Pertussis/farmacología , Potasio/metabolismo , Bloqueadores de los Canales de Potasio/farmacología , Canales de Potasio/metabolismo , Canales de Potasio Calcio-Activados/antagonistas & inhibidores , Ratas , Ratas Sprague-Dawley , Receptores de Glutamato Metabotrópico/efectos de los fármacos , Receptores de Glutamato Metabotrópico/genética , Fosfolipasas de Tipo C/antagonistas & inhibidores
15.
J Appl Physiol (1985) ; 119(10): 1202-9, 2015 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-25792716

RESUMEN

Adenosine A2A receptors and ATP-activated K(+) (KATP) channels contribute to part of the cerebral vasodilatory response to systemic hypoxia, but other mediators are likely involved. Epoxyeicosatrienoic acids (EETs) are cerebral vasodilators and are released from astrocytes exposed to hypoxia. Moreover, stimulation of metabotropic glutamate receptors (mGluR) produces vasodilation by an EET-dependent mechanism. Here, we tested the hypothesis that EET signaling and mGluR activation contribute to hypoxic vasodilation. Laser-Doppler flow was measured over cerebral cortex of anesthetized rats subjected to stepwise reductions in arterial oxygen saturation to 50-70%. Hypoxic reactivity was calculated as the slope of the change in laser-Doppler flow vs. the reciprocal of arterial oxygen content. Hypoxic reactivity significantly decreased from 9.2 ± 1.9 (±95% confidence interval) in controls with vehicle treatment to 2.6 ± 1.4 with the EET antagonist 14,15-epoxyeicosa-5(Z)-enoic acid, to 3.0 ± 1.5 with the EET synthesis inhibitor MS-PPOH, to 1.9 ± 2.3 with the combined mGluR subtype 1 and 5 antagonists 2-methyl-6-(phenylethynyl)pyridine and LY367385, to 5.6 ± 1.2 with the KATP channel inhibitor glibenclamide, and to 5.8 ± 2.3 with the A2A receptor antagonist SCH58261. However, reactivity was not significantly altered by the A2B receptor antagonist MRS1754 (6.7 ± 1.8; P = 0.28 Dunnett's test) or by the 20-hydroxyeicosatetraenoic acid synthesis inhibitor HET0016 (7.5 ± 2.3; P = 0.6). These data indicate that, in addition to the known contributions of A2A receptors and KATP channels to the increase in cerebral blood flow during hypoxia, EETs and mGluRs make a major contribution, possibly by mGluR stimulation and hypoxia-induced release of EETs. In contrast, A2B receptors do not make a major contribution, and 20-hydroxyeicosatetraenoic acid does not significantly limit hypoxic vasodilation.


Asunto(s)
Circulación Cerebrovascular/fisiología , Eicosanoides/metabolismo , Hipoxia/metabolismo , Ácido 8,11,14-Eicosatrienoico/metabolismo , Animales , Ácidos Eicosanoicos/metabolismo , Ácidos Hidroxieicosatetraenoicos/metabolismo , Masculino , Ratas , Ratas Wistar
16.
Eur J Pharmacol ; 486(3): 297-306, 2004 Feb 23.
Artículo en Inglés | MEDLINE | ID: mdl-14985052

RESUMEN

This study examined the effects of a 20-hydroxyeicosatetraenoic acid (20-HETE) antagonist, 20-hydroxyeicosa-6(Z),15(Z)-dienoic acid (WIT002) and two agonists, 4-amino-N-(20-hydroxy-eicosa-5(Z),14(Z)-dienoyl) benzenesulfonamide (ABSA) and 20-hydroxyeicosa-5(Z),14(Z)-dienoic acid (WIT003), on the diameter of rat middle cerebral arteries in vitro and on cerebral blood flow in vivo. WIT003, ABSA and 20-HETE all had a similar effect to reduce the diameter of the middle cerebral artery by 26%. WIT003 and 20-HETE both increased intracellular Ca2+ concentration ([Ca2+]i) in vascular smooth muscle cells isolated from the middle cerebral artery. In contrast, WIT002 had no effect on the basal diameter of the middle cerebral artery but it attenuated the vasoconstrictor responses and the rise in [Ca2+]i in vascular smooth muscle cells following administration of 20-HETE and 5-hydroxytryptamine (5-HT). WIT003 partially restored the vasoconstrictor response to 5-HT in the middle cerebral artery after administration of an inhibitor of the endogenous synthesis of 20-HETE. Infusion of the 20-HETE agonists, WIT003 and ABSA, into cisterna magna of rats reduced baseline cerebral blood flow by 20%, whereas administration of the 20-HETE antagonist, WIT002, had no effect. Intracisternal injection of WIT002 attenuated the fall in cerebral blood flow following injection of blood into the cisterna magna, whereas administration of the 20-HETE agonist, ABSA, potentiated this response. These findings indicate that the 20-HETE agonists, WIT003 and ABSA, increase cerebral vascular tone both in vivo and in vitro and suggest blocking the vasoconstrictor actions of 20-HETE may be useful to prevent the acute fall in cerebral blood flow following subarachnoid hemorrhage.


Asunto(s)
Circulación Cerebrovascular/efectos de los fármacos , Ácidos Hidroxieicosatetraenoicos/agonistas , Ácidos Hidroxieicosatetraenoicos/antagonistas & inhibidores , Ácidos Hidroxieicosatetraenoicos/farmacología , Músculo Liso Vascular/efectos de los fármacos , Vasoconstricción/efectos de los fármacos , Animales , Calcio/metabolismo , Interacciones Farmacológicas , Técnicas In Vitro , Masculino , Arteria Cerebral Media/efectos de los fármacos , Arteria Cerebral Media/fisiología , Músculo Liso Vascular/fisiología , Ratas , Ratas Sprague-Dawley , Flujo Sanguíneo Regional/efectos de los fármacos , Hemorragia Subaracnoidea/fisiopatología , Factores de Tiempo , Vasoconstricción/fisiología
17.
Curr Vasc Pharmacol ; 12(6): 810-7, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24066934

RESUMEN

The existence of arterial myogenic tone was first described by Bayliss in 1902, however, its association with pressure-dependent membrane depolarization was not observed until 1984. The factors that mediate myogenic arterial constriction remain unknown. One possible clue was a finding by our laboratory that cerebral arterial muscle cells express CYP 4A ω-hydroxylase enzyme that catalyzes the formation of the potent vasoconstrictor 20-hydroxyeicosatetraenoic acid (20-HETE) from arachidonic acid (AA), the production of which increased by elevations of intravascular pressure. 20-HETE activates protein kinase C (PKC), inhibits Ca(2+)-activated K(+) (KCa) channels, depolarizes arterial muscle cell membrane, activates L-type Ca(2+) channels, increases intracellular Ca(2+) ([Ca(2+)]i) and mediates autoregulation of cerebral blood flow. Emerging evidence indicates that 20-HETE level increases in ischemia/reperfusion injury and stimulates production of reactive oxygen species (ROS) resulting in oxidative stress induced ischemic stroke injury, which can be prevented by inhibition of 20-HETE synthesis or action. The brain also expresses CYP epoxygenases that convert AA to the vasodilator epoxyeicosatrienoic acids (EETs), the production of which increases in ischemia and provide protection against ischemia-induced tissue damage. Basal or stimulus released ROS act to modify cerebral myogenic tone. Similar to other enzymes CYP epoxygenase and ω-hydroxylase also generate ROS that modify myogenic cerebral reactivity. Hypoxia per se or adenosine released during hypoxia induces increased production of ROS that alter cerebrovascular function. The capacity of the brain to express CYP enzymes that produce bioactive EETs and 20-HETE and generate ROS has a significant bearing in regulating the dynamics of cerebral blood flow and serve as potential therapeutic targets for the management of pathologic disorders of the cerebral circulation.


Asunto(s)
Encéfalo/irrigación sanguínea , Encéfalo/metabolismo , Circulación Cerebrovascular/fisiología , Homeostasis/fisiología , Músculo Liso Vascular/metabolismo , Animales , Sistema Enzimático del Citocromo P-450/metabolismo , Humanos , Especies Reactivas de Oxígeno/metabolismo
18.
J Appl Physiol (1985) ; 114(2): 252-61, 2013 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-23172031

RESUMEN

The present study examined the role of the dual-specificity protein phosphatase-5 (DUSP-5) in the pressure-induced myogenic responses of organ-cultured cerebral arterial segments. In these studies, we initially compared freshly isolated and organ-cultured cerebral arterial segments with respect to responses to step increases in intravascular pressure, vasodilator and vasoconstrictor stimuli, activities of the large-conductance arterial Ca(2+)-activated K(+) (K(Ca)) single-channel current, and stable protein expression of DUSP-5 enzyme. The results demonstrate maintained pressure-dependent myogenic vasoconstriction, DUSP-5 protein expression, endothelium-dependent and -independent dilations, agonist-induced constriction, and unitary K(Ca) channel conductance in organ-cultured cerebral arterial segments similar to that in freshly isolated cerebral arteries. Furthermore, using a permeabilization transfection technique in organ-cultured cerebral arterial segments, gene-specific small interfering RNA (siRNA) induced knockdown of DUSP-5 mRNA and protein, which were associated with enhanced pressure-dependent cerebral arterial myogenic constriction and increased phosphorylation of PKC-ßII. In addition, siRNA knockdown of DUSP-5 reduced levels of phosphorylated ROCK and ERK1 with no change in the level of phosphorylated ERK2. Pharmacological inhibition of ERK1/2 phosphorylation significantly attenuated pressure-induced myogenic constriction in cerebral arteries. The findings within the present studies illustrate that DUSP-5, native in cerebral arterial muscle cells, appears to regulate signaling of pressure-dependent myogenic cerebral arterial constriction, which is crucial for the maintenance of constant cerebral blood flow to the brain.


Asunto(s)
Arterias Cerebrales/fisiología , Circulación Cerebrovascular/fisiología , Fosfatasas de Especificidad Dual/fisiología , Desarrollo de Músculos/fisiología , Músculo Liso Vascular/fisiología , Vasoconstricción/fisiología , Animales , Encéfalo/fisiología , Células Cultivadas , Arterias Cerebrales/citología , Fosfatasas de Especificidad Dual/efectos de los fármacos , Fosfatasas de Especificidad Dual/genética , Masculino , Modelos Animales , Músculo Liso Vascular/citología , Técnicas de Placa-Clamp , Canales de Potasio Calcio-Activados/fisiología , ARN Mensajero/efectos de los fármacos , ARN Mensajero/genética , ARN Interferente Pequeño/farmacología , Ratas , Ratas Sprague-Dawley , Flujo Sanguíneo Regional/fisiología , Transducción de Señal/fisiología
19.
PLoS One ; 8(7): e68498, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23861911

RESUMEN

The present study examined the level of generation of reactive oxygen species (ROS) and roles of inactivation of the phosphatase PTEN and the PI3K/Akt signaling pathway in response to an increase in intramural pressure-induced myogenic cerebral arterial constriction. Step increases in intraluminal pressure of cannulated cerebral arteries induced myogenic constriction and concomitant formation of superoxide (O2 (.-)) and its dismutation product hydrogen peroxide (H2O2) as determined by fluorescent HPLC analysis, microscopic analysis of intensity of dihydroethidium fluorescence and attenuation of pressure-induced myogenic constriction by pretreatment with the ROS scavenger 4,hydroxyl-2,2,6,6-tetramethylpiperidine1-oxyl (tempol) or Mito-tempol or MitoQ in the presence or absence of PEG-catalase. An increase in intraluminal pressure induced oxidation of PTEN and activation of Akt. Pharmacological inhibition of endogenous PTEN activity potentiated pressure-dependent myogenic constriction and caused a reduction in NPo of a 238 pS arterial KCa channel current and an increase in [Ca(2+)]i level in freshly isolated cerebral arterial muscle cells (CAMCs), responses that were attenuated by Inhibition of the PI3K/Akt pathway. These findings demonstrate an increase in intraluminal pressure induced increase in ROS production triggered redox-sensitive signaling mechanism emanating from the cross-talk between oxidative inactivation of PTEN and activation of the PI3K/Akt signaling pathway that involves in the regulation of pressure-dependent myogenic cerebral arterial constriction.


Asunto(s)
Arteria Cerebral Media/metabolismo , Músculo Liso Vascular/metabolismo , Estrés Oxidativo , Fosfohidrolasa PTEN/metabolismo , Transducción de Señal , Animales , Calcio/metabolismo , Catalasa/farmacología , Activación Enzimática/efectos de los fármacos , Peróxido de Hidrógeno/metabolismo , Masculino , Arteria Cerebral Media/efectos de los fármacos , Contracción Muscular/efectos de los fármacos , Músculo Liso Vascular/efectos de los fármacos , Oxidación-Reducción , Fosfohidrolasa PTEN/antagonistas & inhibidores , Polietilenglicoles/farmacología , Canales de Potasio Calcio-Activados/metabolismo , Presión , Ratas , Especies Reactivas de Oxígeno/metabolismo , Transducción de Señal/efectos de los fármacos , Superóxidos/metabolismo
20.
J Am Heart Assoc ; 2(3): e000080, 2013 Apr 25.
Artículo en Inglés | MEDLINE | ID: mdl-23619744

RESUMEN

BACKGROUND: Arachidonic acid (AA) and/or its enzymatic metabolites are important lipid mediators contributing to endothelium-derived hyperpolarizing factor (EDHF)-mediated dilation in multiple vascular beds, including human coronary arterioles (HCAs). However, the mechanisms of action of these lipid mediators in endothelial cells (ECs) remain incompletely defined. In this study, we investigated the role of the transient receptor potential vanilloid 4 (TRPV4) channel in AA-induced endothelial Ca(2+) response and dilation of HCAs. METHODS AND RESULTS: AA induced concentration-dependent dilation in isolated HCAs. The dilation was largely abolished by the TRPV4 antagonist RN-1734 and by inhibition of endothelial Ca(2+)-activated K(+) channels. In native and TRPV4-overexpressing human coronary artery ECs (HCAECs), AA increased intracellular Ca(2+) concentration ([Ca(2+)]i), which was mediated by TRPV4-dependent Ca(2+) entry. The AA-induced [Ca(2+)]i increase was inhibited by cytochrome P450 (CYP) inhibitors. Surprisingly, the CYP metabolites of AA, epoxyeicosatrienoic acids (EETs), were much less potent activators of TRPV4, and CYP inhibitors did not affect EET production in HCAECs. Apart from its effect on [Ca(2+)]i, AA induced endothelial hyperpolarization, and this effect was required for Ca(2+) entry through TRPV4. AA-induced and TRPV4-mediated Ca(2+) entry was also inhibited by the protein kinase A inhibitor PKI. TRPV4 exhibited a basal level of phosphorylation, which was inhibited by PKI. Patch-clamp studies indicated that AA activated TRPV4 single-channel currents in cell-attached and inside-out patches of HCAECs. CONCLUSIONS: AA dilates HCAs through a novel mechanism involving endothelial TRPV4 channel-dependent Ca(2+) entry that requires endothelial hyperpolarization, PKA-mediated basal phosphorylation of TRPV4, and direct activation of TRPV4 channels by AA.


Asunto(s)
Ácido Araquidónico/farmacología , Señalización del Calcio/efectos de los fármacos , Señalización del Calcio/fisiología , Calcio/metabolismo , Vasos Coronarios/efectos de los fármacos , Vasos Coronarios/fisiología , Endotelio Vascular/efectos de los fármacos , Endotelio Vascular/fisiología , Transducción de Señal/efectos de los fármacos , Canales Catiónicos TRPV/efectos de los fármacos , Canales Catiónicos TRPV/fisiología , Arteriolas/citología , Arteriolas/efectos de los fármacos , Arteriolas/fisiología , Células Cultivadas , Vasos Coronarios/citología , Endotelio Vascular/citología , Femenino , Humanos , Masculino , Persona de Mediana Edad
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA