Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 35
Filtrar
1.
Physiol Rev ; 95(2): 645-90, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25834234

RESUMO

The mammalian genome encodes 28 distinct members of the transient receptor potential (TRP) superfamily of cation channels, which exhibit varying degrees of selectivity for different ionic species. Multiple TRP channels are present in all cells and are involved in diverse aspects of cellular function, including sensory perception and signal transduction. Notably, TRP channels are involved in regulating vascular function and pathophysiology, the focus of this review. TRP channels in vascular smooth muscle cells participate in regulating contractility and proliferation, whereas endothelial TRP channel activity is an important contributor to endothelium-dependent vasodilation, vascular wall permeability, and angiogenesis. TRP channels are also present in perivascular sensory neurons and astrocytic endfeet proximal to cerebral arterioles, where they participate in the regulation of vascular tone. Almost all of these functions are mediated by changes in global intracellular Ca(2+) levels or subcellular Ca(2+) signaling events. In addition to directly mediating Ca(2+) entry, TRP channels influence intracellular Ca(2+) dynamics through membrane depolarization associated with the influx of cations or through receptor- or store-operated mechanisms. Dysregulation of TRP channels is associated with vascular-related pathologies, including hypertension, neointimal injury, ischemia-reperfusion injury, pulmonary edema, and neurogenic inflammation. In this review, we briefly consider general aspects of TRP channel biology and provide an in-depth discussion of the functions of TRP channels in vascular smooth muscle cells, endothelial cells, and perivascular cells under normal and pathophysiological conditions.


Assuntos
Sinalização do Cálcio , Cálcio/metabolismo , Endotélio Vascular/metabolismo , Músculo Liso Vascular/metabolismo , Canais de Potencial de Receptor Transitório/metabolismo , Animais , Proliferação de Células , Endotélio Vascular/patologia , Endotélio Vascular/fisiopatologia , Humanos , Potenciais da Membrana , Músculo Liso Vascular/patologia , Músculo Liso Vascular/fisiopatologia , Doenças Vasculares/metabolismo , Doenças Vasculares/patologia , Doenças Vasculares/fisiopatologia , Vasoconstrição , Vasodilatação
2.
Proc Natl Acad Sci U S A ; 112(7): E796-805, 2015 Feb 17.
Artigo em Inglês | MEDLINE | ID: mdl-25646445

RESUMO

Cerebral autosomal dominant arteriopathy with subcortical infarcts and leukoencephalopathy (CADASIL), caused by dominant mutations in the NOTCH3 receptor in vascular smooth muscle, is a genetic paradigm of small vessel disease (SVD) of the brain. Recent studies using transgenic (Tg)Notch3(R169C) mice, a genetic model of CADASIL, revealed functional defects in cerebral (pial) arteries on the surface of the brain at an early stage of disease progression. Here, using parenchymal arterioles (PAs) from within the brain, we determined the molecular mechanism underlying the early functional deficits associated with this Notch3 mutation. At physiological pressure (40 mmHg), smooth muscle membrane potential depolarization and constriction to pressure (myogenic tone) were blunted in PAs from TgNotch3(R169C) mice. This effect was associated with an ∼ 60% increase in the number of voltage-gated potassium (KV) channels, which oppose pressure-induced depolarization. Inhibition of KV1 channels with 4-aminopyridine (4-AP) or treatment with the epidermal growth factor receptor agonist heparin-binding EGF (HB-EGF), which promotes KV1 channel endocytosis, reduced KV current density and restored myogenic responses in PAs from TgNotch3(R169C) mice, whereas pharmacological inhibition of other major vasodilatory influences had no effect. KV1 currents and myogenic responses were similarly altered in pial arteries from TgNotch3(R169C) mice, but not in mesenteric arteries. Interestingly, HB-EGF had no effect on mesenteric arteries, suggesting a possible mechanistic basis for the exclusive cerebrovascular manifestation of CADASIL. Collectively, our results indicate that increasing the number of KV1 channels in cerebral smooth muscle produces a mutant vascular phenotype akin to a channelopathy in a genetic model of SVD.


Assuntos
Encéfalo/fisiopatologia , Transtornos Cerebrovasculares/genética , Canais de Potássio/genética , 4-Aminopiridina/farmacologia , Animais , Encéfalo/irrigação sanguínea , Transtornos Cerebrovasculares/fisiopatologia , Modelos Animais de Doenças , Fator de Crescimento Semelhante a EGF de Ligação à Heparina/fisiologia , Potenciais da Membrana , Camundongos , Camundongos Transgênicos , Receptor Notch3 , Receptores Notch/genética , Receptores Notch/fisiologia
3.
Stroke ; 45(8): 2425-30, 2014 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-24968928

RESUMO

BACKGROUND AND PURPOSE: Parenchymal arterioles (PAs) are high-resistance vessels in the brain that connect pial vessels to the microcirculation. We previously showed that PAs have increased vasoconstriction after ischemia and reperfusion that could increase perfusion deficit. Here, we investigated underlying mechanisms by which early postischemic reperfusion causes increased vasoconstriction of PAs. METHODS: Isolated and pressurized PAs from within the middle cerebral artery territory were studied in male Wistar rats that were either nonischemic control (n=34) or after exposure to transient middle cerebral artery occlusion (MCAO) by filament occlusion for 2 hours with 30 minutes of reperfusion (MCAO; n=38). The relationships among pressure-induced tone, smooth muscle calcium (using Fura 2), and membrane potential were determined. Sensitivity of the contractile apparatus to calcium was measured in permeabilized arterioles using Staphylococcus aureus α-toxin. Reactivity to inhibition of transient receptor potential melastanin receptor type 4 (9-phenanthrol), Rho kinase (Y27632), and protein kinase C (Gö6976) was also measured. RESULTS: After MCAO, PAs had increased myogenic tone compared with controls (47±2% versus 35±2% at 40 mm Hg; P<0.01), without an increase in smooth muscle calcium (177±21 versus 201±16 nmol/L; P>0.05) or membrane depolarization (-38±4 versus -36±1 mV; P>0.05). In α-toxin-permeabilized vessels, MCAO caused increased sensitivity of the contractile apparatus to calcium. MCAO did not affect dilation to transient receptor potential melastanin receptor type 4 or protein kinase C inhibition but diminished dilation to Rho kinase inhibition. CONCLUSIONS: The increased vasoconstriction of PAs during early postischemic reperfusion seems to be due to calcium sensitization of smooth muscle and could contribute to infarct expansion and limit neuroprotective agents from reaching their target tissue.


Assuntos
Arteríolas/fisiopatologia , Isquemia Encefálica/fisiopatologia , Cálcio/metabolismo , Infarto da Artéria Cerebral Média/fisiopatologia , Músculo Liso Vascular/fisiopatologia , Vasoconstrição/fisiologia , Animais , Arteríolas/metabolismo , Isquemia Encefálica/metabolismo , Infarto da Artéria Cerebral Média/metabolismo , Masculino , Potenciais da Membrana/fisiologia , Contração Muscular/fisiologia , Músculo Liso Vascular/metabolismo , Ratos , Ratos Wistar , Reperfusão , Traumatismo por Reperfusão/metabolismo , Traumatismo por Reperfusão/fisiopatologia
4.
Circ Res ; 110(2): 285-94, 2012 Jan 20.
Artigo em Inglês | MEDLINE | ID: mdl-22095728

RESUMO

RATIONALE: Acidosis is a powerful vasodilator signal in the brain circulation. However, the mechanisms by which this response occurs are not well understood, particularly in the cerebral microcirculation. One important mechanism to dilate cerebral (pial) arteries is by activation of large-conductance, calcium-sensitive potassium (BK(Ca)) channels by local Ca(2+) signals (Ca(2+) sparks) through ryanodine receptors (RyRs). However, the role of this pathway in the brain microcirculation is not known. OBJECTIVE: The objectives of this study were to determine the mechanism by which acidosis dilates brain parenchymal arterioles (PAs) and to elucidate the roles of RyRs and BK(Ca) channels in this response. METHODS AND RESULTS: Internal diameter and vascular smooth muscle cell Ca(2+) signals were measured in isolated pressurized murine PAs, using imaging techniques. In physiological pH (7.4), vascular smooth muscle cells exhibited primarily RyR-dependent Ca(2+) waves. Reducing external pH from 7.4 to 7.0 in both normocapnic and hypercapnic conditions decreased Ca(2+) wave activity, and dramatically increased Ca(2+) spark activity. Acidic pH caused a dilation of PAs which was inhibited by about 60% by BK(Ca) channel or RyR blockers, in a nonadditive manner. Similarly, dilator responses to acidosis were reduced by nearly 60% in arterioles from BK(Ca) channel knockout mice. Dilations induced by acidic pH were unaltered by inhibitors of K(ATP) channels or nitric oxide synthase. CONCLUSIONS: These results support the novel concept that acidification, by converting Ca(2+) waves to sparks, leads to the activation of BK(Ca) channels to induce dilation of cerebral PAs.


Assuntos
Acidose/metabolismo , Sinalização do Cálcio , Subunidades alfa do Canal de Potássio Ativado por Cálcio de Condutância Alta/metabolismo , Músculo Liso Vascular/metabolismo , Pia-Máter/irrigação sanguínea , Vasodilatação , Acidose/fisiopatologia , Animais , Arteríolas/metabolismo , Arteríolas/fisiopatologia , Agonistas dos Canais de Cálcio/farmacologia , Bloqueadores dos Canais de Cálcio/farmacologia , Sinalização do Cálcio/efeitos dos fármacos , Relação Dose-Resposta a Droga , Inibidores Enzimáticos/farmacologia , Concentração de Íons de Hidrogênio , Técnicas In Vitro , Canais KATP/antagonistas & inibidores , Canais KATP/metabolismo , Subunidades alfa do Canal de Potássio Ativado por Cálcio de Condutância Alta/antagonistas & inibidores , Subunidades alfa do Canal de Potássio Ativado por Cálcio de Condutância Alta/genética , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Músculo Liso Vascular/efeitos dos fármacos , Músculo Liso Vascular/fisiopatologia , Óxido Nítrico Sintase Tipo III/antagonistas & inibidores , Óxido Nítrico Sintase Tipo III/metabolismo , Bloqueadores dos Canais de Potássio/farmacologia , Canal de Liberação de Cálcio do Receptor de Rianodina/efeitos dos fármacos , Canal de Liberação de Cálcio do Receptor de Rianodina/metabolismo , Fatores de Tempo , Vasodilatação/efeitos dos fármacos
5.
Microcirculation ; 20(4): 307-16, 2013 May.
Artigo em Inglês | MEDLINE | ID: mdl-23216877

RESUMO

The cerebral blood supply is delivered by a surface network of pial arteries and arterioles from which arise (parenchymal) arterioles that penetrate into the cortex and terminate in a rich capillary bed. The critical regulation of CBF, locally and globally, requires precise vasomotor regulation of the intracerebral microvasculature. This vascular region is anatomically unique as illustrated by the presence of astrocytic processes that envelope almost the entire basolateral surface of PAs. There are, moreover, notable functional differences between pial arteries and PAs. For example, in pial VSMCs, local calcium release events ("calcium sparks") through ryanodine receptor (RyR) channels in SR membrane activate large conductance, calcium-sensitive potassium channels to modulate vascular diameter. In contrast, VSMCs in PAs express functional RyR and BK channels, but under physiological conditions, these channels do not oppose pressure-induced vasoconstriction. Here, we summarize the roles of ryanodine receptors in the parenchymal microvasculature under physiologic and pathologic conditions, and discuss their importance in the control of CBF.


Assuntos
Sinalização do Cálcio/fisiologia , Córtex Cerebral/irrigação sanguínea , Circulação Cerebrovascular/fisiologia , Microcirculação/fisiologia , Tono Muscular/fisiologia , Músculo Liso Vascular/metabolismo , Canal de Liberação de Cálcio do Receptor de Rianodina/metabolismo , Animais , Humanos
6.
Biochim Biophys Acta ; 1804(3): 524-32, 2010 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-20018259

RESUMO

The cGMP-dependent protein kinase type I (PKG I) is an essential regulator of cellular function in blood vessels throughout the body. DT-2, a peptidic inhibitor of PKG, has played a central role in determining the molecular mechanisms of vascular control involving PKG and its signaling partners. Here, we report the development of (d)-amino acid DT-2 derivatives, namely the retro-inverso ri-(d)-DT-2 and the all (d)-amino acid analog, (d)-DT-2. Both peptide analogs were potent PKG Ialpha inhibitors with K(i) values of 5.5 nM (ri-(d)-DT-2) and 0.8 nM ((d)-DT-2) as determined using a hyperbolic mixed-type inhibition model. Also, both analogs were proteolytically stable in vivo, showed elevated selectivity, and displayed enhanced membrane translocation properties. Studies on isolated arteries from the resistance vasculature demonstrated that intraluminally perfused (d)-DT-2 significantly inhibited vasodilation induced by 8-Br-cGMP. Furthermore, in vivo application of (d)-DT-2 established a uniform translocation pattern in the resistance vasculature, with exception of the brain. Thus, (d)-DT-2 caused significant increases in mean arterial blood pressure in unrestrained, awake mice. Further, mesenteric arteries isolated from (d)-DT-2 treated animals showed a markedly reduced dilator response to 8-Br-cGMP in vitro. Our results clearly demonstrate that (d)-DT-2 is a superior inhibitor of PKG Ialpha and its application in vivo leads to sustained inhibition of PKG in vascular smooth muscle cells. The discovery of (d)-DT-2 may help our understanding of how blood vessels constrict and dilate and may also aid the development of new strategies and therapeutic agents targeted to the prevention and treatment of vascular disorders such as hypertension, stroke and coronary artery disease.


Assuntos
Proteínas Quinases Dependentes de GMP Cíclico/antagonistas & inibidores , Fluoresceínas/farmacologia , Fragmentos de Peptídeos/farmacologia , Inibidores de Proteínas Quinases/farmacologia , Vasodilatação/efeitos dos fármacos , Animais , Pressão Sanguínea/efeitos dos fármacos , Linhagem Celular , Doença da Artéria Coronariana/tratamento farmacológico , Doença da Artéria Coronariana/enzimologia , GMP Cíclico/análogos & derivados , GMP Cíclico/farmacologia , Proteína Quinase Dependente de GMP Cíclico Tipo I , Proteínas Quinases Dependentes de GMP Cíclico/química , Proteínas Quinases Dependentes de GMP Cíclico/metabolismo , Fluoresceínas/uso terapêutico , Hipertensão/tratamento farmacológico , Hipertensão/enzimologia , Masculino , Artérias Mesentéricas/enzimologia , Camundongos , Modelos Biológicos , Músculo Liso Vascular/enzimologia , Miócitos de Músculo Liso/enzimologia , Fragmentos de Peptídeos/uso terapêutico , Inibidores de Proteínas Quinases/química , Inibidores de Proteínas Quinases/uso terapêutico , Spodoptera , Vasoconstrição/efeitos dos fármacos
7.
Am J Physiol Heart Circ Physiol ; 300(3): H803-12, 2011 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-21148767

RESUMO

Intracerebral (parenchymal) arterioles are morphologically and physiologically unique compared with pial arteries and arterioles. The ability of subarachnoid hemorrhage (SAH) to induce vasospasm in large-diameter pial arteries has been extensively studied, although the contribution of this phenomenon to patient outcome is controversial. Currently, little is known regarding the impact of SAH on parenchymal arterioles, which are critical for regulation of local and global cerebral blood flow. Here diameter, smooth muscle intracellular Ca(2+) concentration ([Ca(2+)](i)), and membrane potential measurements were used to assess the function of intact brain parenchymal arterioles isolated from unoperated (control), sham-operated, and SAH model rats. At low intravascular pressure (5 mmHg), membrane potential and [Ca(2+)](i) were not different in arterioles from control, sham-operated, and SAH animals. However, raising intravascular pressure caused significantly greater membrane potential depolarization, elevation in [Ca(2+)](i), and constriction in SAH arterioles. This SAH-induced increase in [Ca(2+)](i) and tone occurred in the absence of the vascular endothelium and was abolished by the L-type voltage-dependent calcium channel (VDCC) inhibitor nimodipine. Arteriolar [Ca(2+)](i) and tone were not different between groups when smooth muscle membrane potential was adjusted to the same value. Protein and mRNA levels of the L-type VDCC Ca(V)1.2 were similar in parenchymal arterioles isolated from control and SAH animals, suggesting that SAH did not cause VDCC upregulation. We conclude that enhanced parenchymal arteriolar tone after SAH is driven by smooth muscle membrane potential depolarization, leading to increased L-type VDCC-mediated Ca(2+) influx.


Assuntos
Arteríolas/fisiopatologia , Encéfalo/irrigação sanguínea , Encéfalo/fisiopatologia , Potenciais da Membrana/fisiologia , Hemorragia Subaracnóidea/fisiopatologia , Vasoconstrição/fisiologia , Animais , Arteríolas/efeitos dos fármacos , Pressão Sanguínea/efeitos dos fármacos , Pressão Sanguínea/fisiologia , Encéfalo/efeitos dos fármacos , Cálcio/fisiologia , Canais de Cálcio Tipo L/fisiologia , Endotélio Vascular/efeitos dos fármacos , Endotélio Vascular/fisiopatologia , Masculino , Potenciais da Membrana/efeitos dos fármacos , Músculo Liso Vascular/efeitos dos fármacos , Músculo Liso Vascular/fisiopatologia , Nimodipina/farmacologia , Ratos , Ratos Sprague-Dawley , Hemorragia Subaracnóidea/tratamento farmacológico , Regulação para Cima/efeitos dos fármacos , Regulação para Cima/fisiologia , Vasoconstrição/efeitos dos fármacos
8.
Acta Neurochir Suppl ; 110(Pt 1): 145-50, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21116930

RESUMO

BACKGROUND: Ca2+ signaling mechanisms are crucial for proper regulation of vascular smooth muscle contractility and vessel diameter. In cerebral artery myocytes, a rise in global cytosolic Ca2+ concentration ([Ca2+]i) causes contraction while an increase in local Ca2+ release events from the sarcoplasmic reticulum (Ca2+ sparks) leads to increased activity of large-conductance Ca2+-activated (BK) K+ channels, hyperpolarization and relaxation. Here, we examined the impact of SAH on Ca2+ spark activity and [Ca2+]i in cerebral artery myocytes following SAH. METHODS: A rabbit double injection SAH model was used in this study. Five days after the initial intracisternal injection of whole blood, small diameter cerebral arteries were dissected from the brain for study. For simultaneous measurement of arterial wall [Ca2+]i and diameter, vessels were cannulated and loaded with the ratiometric Ca2+ indicator fura-2. For measurement of Ca2+ sparks, individual myocytes were enzymatically isolated from cerebral arteries and loaded with the Ca2+ indicator fluo-4. Sparks were visualized using laser scanning confocal microscopy. RESULTS: Arterial wall [Ca2+]i was significantly elevated and greater levels of myogenic tone developed in arteries isolated from SAH animals compared with arteries isolated from healthy animals. The L-type voltage-dependent Ca2+ channel (VDCC) blocker nifedipine attenuated increases in [Ca2+]i and tone in both groups suggesting increased VDCC activity following SAH. Membrane potential measurement using intracellular microelectrodes revealed significant depolarization of vascular smooth muscle following SAH. Further, myocytes from SAH animals exhibited significantly reduced Ca2+ spark frequency (~50%). CONCLUSIONS: Our findings suggest decreased Ca2+ spark frequency leads to reduced BK channel activity in cerebral artery myocytes following SAH. This results in membrane potential depolarization, increased VDCC activity, elevated [Ca2+]i and decreased vessel diameter. We propose this mechanism of enhanced cerebral artery myocyte contractility may contribute to decreased cerebral blood flow and development of neurological deficits in SAH patients.


Assuntos
Sinalização do Cálcio/fisiologia , Artérias Cerebrais/patologia , Células Musculares/fisiologia , Hemorragia Subaracnóidea/patologia , Animais , Pressão Sanguínea/fisiologia , Cálcio/metabolismo , Bloqueadores dos Canais de Cálcio/farmacologia , Canais de Cálcio Tipo L/metabolismo , Modelos Animais de Doenças , Corantes Fluorescentes , Masculino , Potenciais da Membrana/efeitos dos fármacos , Microscopia Confocal/métodos , Modelos Biológicos , Células Musculares/efeitos dos fármacos , Células Musculares/metabolismo , Coelhos , Hemorragia Subaracnóidea/fisiopatologia
9.
Clin Sci (Lond) ; 119(1): 19-36, 2010 Apr 07.
Artigo em Inglês | MEDLINE | ID: mdl-20370719

RESUMO

TRP (transient receptor potential) channels play important roles in the regulation of normal and pathological cellular function. In the vasculature, TRP channels are present both in ECs (endothelial cells) and vascular SMCs (smooth muscle cells) and contribute to vasomotor control mechanisms in most vascular beds. Vascular TRP channels are activated by various stimuli, such as mechanical perturbation, receptor activation and dietary molecules. Some of the specific roles of these channels in normal and impaired vascular function have emerged in recent years and include participation in vascular signalling processes, such as neurotransmission, hormonal signalling, NO production, myogenic tone and autoregulation of blood flow, thermoregulation, responses to oxidative stress and cellular proliferative activity. Current research is aimed at understanding the interactions of TRP channels with other vascular proteins and signalling mechanisms. These studies should reveal new targets for pharmacological therapy of vascular diseases, such as hypertension, ischaemia and vasospasm, and vascular proliferative states.


Assuntos
Endotélio Vascular/fisiologia , Músculo Liso Vascular/fisiologia , Canais de Potencial de Receptor Transitório/fisiologia , Células Endoteliais/fisiologia , Humanos , Miócitos de Músculo Liso/fisiologia
10.
Am J Physiol Heart Circ Physiol ; 297(3): H1096-102, 2009 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-19617407

RESUMO

Transient receptor potential vanilloid 4 (TRPV4) channels have been implicated as mediators of calcium influx in both endothelial and vascular smooth muscle cells and are potentially important modulators of vascular tone. However, very little is known about the functional roles of TRPV4 in the resistance vasculature or how these channels influence hemodynamic properties. In the present study, we examined arterial vasomotor activity in vitro and recorded blood pressure dynamics in vivo using TRPV4 knockout (KO) mice. Acetylcholine-induced hyperpolarization and vasodilation were reduced by approximately 75% in mesenteric resistance arteries from TRPV4 KO versus wild-type (WT) mice. Furthermore, 11,12-epoxyeicosatrienoic acid (EET), a putative endothelium-derived hyperpolarizing factor, activated a TRPV4-like cation current and hyperpolarized the membrane of vascular smooth muscle cells, resulting in the dilation of mesenteric arteries from WT mice. In contrast, 11,12-EET had no effect on membrane potential, diameter, or ionic currents in the mesenteric arteries from TRPV4 KO mice. A disruption of the endothelium reduced 11,12-EET-induced hyperpolarization and vasodilatation by approximately 50%. A similar inhibition of these responses was observed following the block of endothelial (small and intermediate conductance) or smooth muscle (large conductance) K(+) channels, suggesting a link between 11,12-EET activity, TRPV4, and K(+) channels in endothelial and smooth muscle cells. Finally, we found that hypertension induced by the inhibition of nitric oxide synthase was greater in TRPV4 KO compared with WT mice. These results support the conclusion that both endothelial and smooth muscle TRPV4 channels are critically involved in the vasodilation of mesenteric arteries in response to endothelial-derived factors and suggest that in vivo this mechanism opposes the effects of hypertensive stimuli.


Assuntos
Pressão Sanguínea/fisiologia , Hipertensão/fisiopatologia , Canais de Cátion TRPV/genética , Canais de Cátion TRPV/metabolismo , Resistência Vascular/fisiologia , Vasodilatação/fisiologia , Ácido 8,11,14-Eicosatrienoico/análogos & derivados , Ácido 8,11,14-Eicosatrienoico/farmacologia , Animais , Endotélio Vascular/fisiologia , Artérias Mesentéricas/efeitos dos fármacos , Artérias Mesentéricas/fisiologia , Camundongos , Camundongos Knockout , Músculo Liso Vascular/fisiologia , Óxido Nítrico Sintase/metabolismo , Canais de Potássio Cálcio-Ativados/antagonistas & inibidores , Canais de Potássio Cálcio-Ativados/fisiologia , Resistência Vascular/efeitos dos fármacos , Vasodilatação/efeitos dos fármacos , Vasodilatadores/farmacologia
11.
J Cereb Blood Flow Metab ; 39(4): 670-679, 2019 04.
Artigo em Inglês | MEDLINE | ID: mdl-29260608

RESUMO

Activation of ATP-sensitive potassium (KATP) channels in arterial smooth muscle (ASM) contributes to vasodilation evoked by a variety of endogenous and exogenous compounds. Although controversial, activation of KATP channels by neuropeptides such as calcitonin gene-related peptide (CGRP) and pituitary adenylate cyclase activating peptide (PACAP) in the trigeminovascular system, including the middle meningeal artery (MMA), has been linked to migraine headache. The objective of the current study was to determine if ongoing KATP channel activity also influences MMA diameter. In the absence of other exogenous compounds, the KATP channel inhibitors glibenclamide and PNU37883A induced constriction of isolated and pressurized MMAs. In contrast, KATP channel inhibition did not alter cerebral artery diameter. Consistent with tonic KATP activity in MMA, glibenclamide also induced ASM membrane potential depolarization and increased cytosolic Ca2+. Inhibitors of cAMP-dependent protein kinase (PKA) abolished basal KATP activation in MMA and caused a marked decrease in sensitivity to the synthetic KATP channel opener, cromakalim. In vivo MMA constriction in response to gibenclamide was observed using two-photon imaging of arterial diameter. Together these results indicate that PKA-mediated tonic KATP channel activity contributes to the regulation of MMA diameter.


Assuntos
Canais KATP/metabolismo , Artérias Meníngeas/diagnóstico por imagem , Animais , Artérias Cerebrais , Glibureto/farmacologia , Canais KATP/antagonistas & inibidores , Artérias Meníngeas/anatomia & histologia , Artérias Meníngeas/efeitos dos fármacos , Transtornos de Enxaqueca/etiologia , Músculo Liso Vascular , Ratos , Vasoconstrição/efeitos dos fármacos
12.
Clin Exp Pharmacol Physiol ; 35(9): 1116-20, 2008 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-18215190

RESUMO

Members of the transient receptor potential (TRP) channel superfamily are present in vascular smooth muscle cells and play important roles in the regulation of vascular contractility. The TRPC3 and TRPC6 channels are activated by stimulation of several excitatory receptors in vascular smooth muscle cells. Activation of these channels leads to myocyte depolarization, which stimulates Ca2+ entry via voltage-dependent Ca2+ channels (VDCC), leading to vasoconstriction. The TRPV4 channels in arterial myocytes are activated by epoxyeicosatrienoic acids, and activation of the channels enhances Ca2+ spark and transient Ca2+-sensitive K+ channel activity, thereby hyperpolarizing and relaxing vascular smooth muscle cells. The TRPC6 and TRPM4 channels are activated by mechanical stimulation of cerebral artery myocytes. Subsequent depolarization and activation of VDCC Ca2+ entry is directly linked to the development of myogenic tone in vitro and to autoregulation of cerebral blood flow in vivo. These findings imply a fundamental importance of TRP channels in the regulation of vascular smooth muscle tone and suggest that TRP channels could be important targets for drug therapy under conditions in which vascular contractility is disturbed (e.g. hypertension, stroke, vasospasm).


Assuntos
Circulação Cerebrovascular/fisiologia , Homeostase/fisiologia , Canais de Potencial de Receptor Transitório/fisiologia , Resistência Vascular/fisiologia , Animais , Ácidos Eicosanoicos/farmacologia , Humanos , Mecanotransdução Celular/fisiologia , Modelos Biológicos , Músculo Liso Vascular/metabolismo , Músculo Liso Vascular/fisiologia , Canais de Potencial de Receptor Transitório/metabolismo , Doenças Vasculares/etiologia , Vasoconstrição/fisiologia , Vasodilatação/efeitos dos fármacos , Sistema Vasomotor/metabolismo , Sistema Vasomotor/fisiologia
13.
Stroke ; 38(8): 2322-8, 2007 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-17585083

RESUMO

BACKGROUND AND PURPOSE: The transient receptor potential channel TRPM4 is critically linked to the myogenic constrictor response of cerebral arteries that occurs when intravascular pressure increases. This myogenic behavior is thought to be fundamentally involved in the mechanisms of blood flow autoregulation. In this study, we tested the hypothesis that TRPM4 channels in cerebrovascular myocytes contribute to cerebral blood flow autoregulation in vivo. METHODS: In vivo suppression of cerebrovascular TRPM4 expression was achieved by infusing antisense oligodeoxynucleotides into the cerebral spinal fluid of 400- to 550-g Sprague-Dawley rats at 80 microg x day(-1) for 7 days using an osmotic pump that discharged into the lateral cerebral ventricle. Absolute cerebral blood flow measurements were obtained over a range of mean arterial pressures using fluorescent microsphere methods. RESULTS: Oligonucleotides infused into the cerebrospinal fluid were detected in the smooth muscle cells of pial arteries. Semi-quantitative RT-PCR indicated that the message for TRPM4 was decreased in the cerebral arteries of antisense-treated rats. Myogenic constriction was decreased by 70% to 85% in cerebral arteries isolated from TRPM4 antisense- compared with control sense-treated rats. Cerebral blood flow was significantly greater in TRPM4 antisense- versus sense-treated rats at resting and elevated mean arterial pressures, indicating that autoregulatory vasoconstrictor activity was compromised in TRPM4 antisense-treated animals. CONCLUSIONS: In vivo suppression of TRPM4 decreases cerebral artery myogenic constrictions and impairs autoregulation, thus implicating TRPM4 channels and myogenic constriction as major contributors to cerebral blood flow regulation in the living animal.


Assuntos
Artérias Cerebrais/metabolismo , Circulação Cerebrovascular/fisiologia , Homeostase/fisiologia , Canais de Cátion TRPM/fisiologia , Animais , Pressão Sanguínea/efeitos dos fármacos , Pressão Sanguínea/fisiologia , Artérias Cerebrais/efeitos dos fármacos , Circulação Cerebrovascular/efeitos dos fármacos , Regulação para Baixo/efeitos dos fármacos , Regulação para Baixo/fisiologia , Homeostase/efeitos dos fármacos , Masculino , Microesferas , Músculo Liso/efeitos dos fármacos , Músculo Liso/metabolismo , Oligonucleotídeos Antissenso/farmacologia , Ratos , Ratos Sprague-Dawley , Canais de Cátion TRPM/antagonistas & inibidores , Canais de Cátion TRPM/genética , Vasoconstrição/efeitos dos fármacos , Vasoconstrição/fisiologia
14.
Circ Res ; 97(12): 1270-9, 2005 Dec 09.
Artigo em Inglês | MEDLINE | ID: mdl-16269659

RESUMO

Vasodilatory factors produced by the endothelium are critical for the maintenance of normal blood pressure and flow. We hypothesized that endothelial signals are transduced to underlying vascular smooth muscle by vanilloid transient receptor potential (TRPV) channels. TRPV4 message was detected in RNA from cerebral artery smooth muscle cells. In patch-clamp experiments using freshly isolated cerebral myocytes, outwardly rectifying whole-cell currents with properties consistent with those of expressed TRPV4 channels were evoked by the TRPV4 agonist 4alpha-phorbol 12,13-didecanoate (4alpha-PDD) (5 micromol/L) and the endothelium-derived arachidonic acid metabolite 11,12 epoxyeicosatrienoic acid (11,12 EET) (300 nmol/L). Using high-speed laser-scanning confocal microscopy, we found that 11,12 EET increased the frequency of unitary Ca2+ release events (Ca2+ sparks) via ryanodine receptors located on the sarcoplasmic reticulum of cerebral artery smooth muscle cells. EET-induced Ca2+ sparks activated nearby sarcolemmal large-conductance Ca2+-activated K+ (BKCa) channels, measured as an increase in the frequency of transient K+ currents (referred to as "spontaneous transient outward currents" [STOCs]). 11,12 EET-induced increases in Ca2+ spark and STOC frequency were inhibited by lowering external Ca2+ from 2 mmol/L to 10 micromol/L but not by voltage-dependent Ca2+ channel inhibitors, suggesting that these responses require extracellular Ca2+ influx via channels other than voltage-dependent Ca2+ channels. Antisense-mediated suppression of TRPV4 expression in intact cerebral arteries prevented 11,12 EET-induced smooth muscle hyperpolarization and vasodilation. Thus, we conclude that TRPV4 forms a novel Ca2+ signaling complex with ryanodine receptors and BKCa channels that elicits smooth muscle hyperpolarization and arterial dilation via Ca2+-induced Ca2+ release in response to an endothelial-derived factor.


Assuntos
Ácido 8,11,14-Eicosatrienoico/análogos & derivados , Sinalização do Cálcio , Canais de Potássio Ativados por Cálcio de Condutância Alta/fisiologia , Músculo Liso Vascular/fisiologia , Miócitos de Músculo Liso/fisiologia , Canal de Liberação de Cálcio do Receptor de Rianodina/fisiologia , Canais de Cátion TRPV/fisiologia , Ácido 8,11,14-Eicosatrienoico/farmacologia , Animais , Cálcio/metabolismo , Artérias Cerebrais/química , Masculino , Músculo Liso Vascular/química , Ratos , Ratos Sprague-Dawley , Canais de Cátion TRPV/análise , Vasodilatação/efeitos dos fármacos
15.
J Cereb Blood Flow Metab ; 37(1): 140-152, 2017 01.
Artigo em Inglês | MEDLINE | ID: mdl-26661251

RESUMO

Cerebral arterioles contribute critically to regulation of local and global blood flow within the brain. Dysfunction of these blood vessels is implicated in numerous cardiovascular diseases. However, treatments are limited due to incomplete understanding of fundamental control mechanisms at this level of circulation. Emerging evidence points to a key role of Rho-associated protein kinase in regulation of microvascular contractility. This study sought to decipher the mechanisms of Rho-associated protein kinase-mediated myogenic vasoconstriction in cerebral parenchymal arterioles. Here, we report that the Rho-associated protein kinase inhibitor H1152 strongly attenuated pressure-induced constriction, cytosolic [Ca2+] increases, and depolarization of isolated parenchymal arterioles. Further, the RhoA activator CN03 potentiated parenchymal arteriole myogenic constriction and depolarization, indicating important involvement of RhoA/Rho-associated protein kinase signaling in myogenic excitation-contraction mechanisms. Because of the well-established role of TRPM4 in pressure-induced depolarization, possible modulatory effects of Rho-associated protein kinase on TRPM4 currents were explored using patch clamp electrophysiology. TRPM4 currents were suppressed by H1152 and enhanced by CN03. Finally, H1152 elevated the apparent [Ca2+]-threshold for TRPM4 activation, suggesting that Rho-associated protein kinase activates TRPM4 by increasing its Ca2+-sensitivity. Our results support a novel mechanism whereby Rho-associated protein kinase-mediated myogenic vasoconstriction occurs primarily through activation of TRPM4 channels, smooth muscle depolarization, and cytosolic [Ca2+] increases in cerebral arterioles.


Assuntos
Arteríolas/fisiologia , Músculo Liso Vascular/fisiologia , Vasoconstrição/efeitos dos fármacos , Quinases Associadas a rho/metabolismo , Animais , Cálcio/metabolismo , Potenciais da Membrana/fisiologia , Contração Muscular/fisiologia , Técnicas de Patch-Clamp , Ratos , Canais de Cátion TRPM/fisiologia
16.
Nat Neurosci ; 20(5): 717-726, 2017 May.
Artigo em Inglês | MEDLINE | ID: mdl-28319610

RESUMO

Blood flow into the brain is dynamically regulated to satisfy the changing metabolic requirements of neurons, but how this is accomplished has remained unclear. Here we demonstrate a central role for capillary endothelial cells in sensing neural activity and communicating it to upstream arterioles in the form of an electrical vasodilatory signal. We further demonstrate that this signal is initiated by extracellular K+ -a byproduct of neural activity-which activates capillary endothelial cell inward-rectifier K+ (KIR2.1) channels to produce a rapidly propagating retrograde hyperpolarization that causes upstream arteriolar dilation, increasing blood flow into the capillary bed. Our results establish brain capillaries as an active sensory web that converts changes in external K+ into rapid, 'inside-out' electrical signaling to direct blood flow to active brain regions.


Assuntos
Encéfalo/irrigação sanguínea , Capilares/fisiologia , Células Endoteliais/fisiologia , Canais de Potássio Corretores do Fluxo de Internalização/fisiologia , Animais , Masculino , Potenciais da Membrana/fisiologia , Camundongos , Camundongos Knockout , Camundongos Transgênicos , Potássio/fisiologia , Canais de Potássio Corretores do Fluxo de Internalização/genética , Vasodilatação/fisiologia
17.
Circ Res ; 95(9): 922-9, 2004 Oct 29.
Artigo em Inglês | MEDLINE | ID: mdl-15472118

RESUMO

Local control of cerebral blood flow is regulated in part through myogenic constriction of resistance arteries. Although this response requires Ca2+ influx via voltage-dependent Ca2+ channels secondary to smooth muscle cell depolarization, the mechanisms responsible for alteration of vascular smooth muscle (VSM) cell membrane potential are not fully understood. A previous study from our laboratory demonstrated a critical role for a member of the transient receptor potential (TRP) superfamily of ion channels, TRPC6, in this response. Several other of the approximately 22 identified TRP proteins are also present in cerebral arteries, but their functions have not been elucidated. Two of these channels, TRPM4 and TRPM5, exhibit biophysical properties that are consistent with a role for control of membrane potential of excitable cells. We hypothesized that TRPM4/TRPM5-dependent currents contribute to myogenic vasoconstriction of cerebral arteries. Cation channels with unitary conductance, ion selectivity and Ca2+-dependence similar to those of cloned TRPM4 and TRPM5 were present in freshly isolated VSM cells. We found that TRPM4 mRNA was detected in both whole cerebral arteries and in isolated VSM cells whereas TRPM5 message was absent from cerebral artery myocytes. We also found that pressure-induced smooth muscle cell depolarization was attenuated in isolated cerebral arteries treated with TRPM4 antisense oligodeoxynucleotides to downregulate channel subunit expression. In agreement with these data, myogenic vasoconstriction of intact cerebral arteries administered TRPM4 antisense was attenuated compared with controls, whereas KCl-induced constriction did not differ between groups. We concluded that activation of TRPM4-dependent currents contributed to myogenic vasoconstriction of cerebral arteries.


Assuntos
Cálcio/fisiologia , Cerebelo/irrigação sanguínea , Artérias Cerebrais/fisiologia , Canais Iônicos/fisiologia , Músculo Liso Vascular/citologia , Canais de Cátion TRPM/fisiologia , Vasoconstrição/fisiologia , Animais , Artérias/fisiologia , Cálcio/farmacologia , Regulação para Baixo , Canais Iônicos/biossíntese , Canais Iônicos/genética , Transporte de Íons/efeitos dos fármacos , Masculino , Potenciais da Membrana/efeitos dos fármacos , Músculo Liso Vascular/metabolismo , Miócitos de Músculo Liso/efeitos dos fármacos , Miócitos de Músculo Liso/metabolismo , Oligodesoxirribonucleotídeos/farmacologia , Oligodesoxirribonucleotídeos Antissenso/farmacologia , Técnicas de Patch-Clamp , RNA Mensageiro/biossíntese , Ratos , Ratos Sprague-Dawley , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Canais de Cátion TRPM/biossíntese , Canais de Cátion TRPM/genética , Acetato de Tetradecanoilforbol/farmacologia , Uridina Trifosfato/farmacologia
18.
Circ Res ; 90(3): 248-50, 2002 Feb 22.
Artigo em Inglês | MEDLINE | ID: mdl-11861411

RESUMO

Elevation of intravascular pressure causes depolarization and constriction (myogenic tone) of small arteries and arterioles, and this response is a key element in blood flow regulation. However, the nature of pressure-induced depolarization has remained elusive. In the present study, we provide evidence that a transient receptor potential channel (TRPC6) homologue has a major role in this depolarizing response to pressure. Antisense oligodeoxynucleotides to TRPC6 decreased TRPC6 protein expression and greatly attenuated arterial smooth muscle depolarization and constriction caused by elevated pressure in intact cerebral arteries. Suppressing the expression of this channel protein also reduced the current density of a major cation current in resistance artery smooth muscle cells. We propose that TRPC6 channels play an essential role in regulation of myogenic tone.


Assuntos
Pressão Sanguínea/fisiologia , Canais de Cálcio/metabolismo , Artérias Cerebrais/metabolismo , Músculo Liso Vascular/metabolismo , Resistência Vascular/fisiologia , Animais , Western Blotting , Canais de Cálcio/genética , Artérias Cerebrais/química , Imuno-Histoquímica , Técnicas In Vitro , Músculo Liso Vascular/química , Músculo Liso Vascular/efeitos dos fármacos , Oligonucleotídeos Antissenso/farmacologia , Técnicas de Patch-Clamp , RNA Mensageiro/antagonistas & inibidores , RNA Mensageiro/metabolismo , Ratos , Ratos Sprague-Dawley , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Canais de Cátion TRPC , Resistência Vascular/efeitos dos fármacos , Vasoconstrição/efeitos dos fármacos , Vasoconstrição/fisiologia
19.
Circ Res ; 93(2): 124-31, 2003 Jul 25.
Artigo em Inglês | MEDLINE | ID: mdl-12805243

RESUMO

The endothelium is a critical regulator of vascular tone, and dysfunction of the endothelium contributes to numerous cardiovascular pathologies. Recent studies suggest that apamin-sensitive, small-conductance, Ca2+-activated K+ channels may play an important role in active endothelium-dependent vasodilations, and expression of these channels may be altered in disease states characterized by vascular dysfunction. Here, we used a transgenic mouse (SK3T/T) in which SK3 expression levels can be manipulated with dietary doxycycline (DOX) to test the hypothesis that the level of expression of the SK subunit, SK3, in endothelial cells alters arterial function and blood pressure. SK3 protein was elevated in small mesenteric arteries from SK3T/T mice compared with wild-type mice and was greatly suppressed by dietary DOX. SK3 was detected in the endothelium and not in the smooth muscle by immunohistochemistry. In whole-cell patch-clamp experiments, SK currents in endothelial cells from SK3T/T mice were almost completely suppressed by dietary DOX. In intact arteries, SK3 channels contributed to sustained hyperpolarization of the endothelial membrane potential, which was communicated to the arterial smooth muscle. Pressure- and phenylephrine-induced constrictions of SK3T/T arteries were substantially enhanced by treatment with apamin, suppression of SK3 expression with DOX, or removal of the endothelium. In addition, suppression of SK3 expression caused a pronounced and reversible elevation of blood pressure. These results indicate that endothelial SK3 channels exert a profound, tonic, hyperpolarizing influence in resistance arteries and suggest that the level of SK3 channel expression in endothelial cells is a fundamental determinant of vascular tone and blood pressure.


Assuntos
Artérias Mesentéricas/fisiologia , Canais de Potássio Cálcio-Ativados , Canais de Potássio/metabolismo , Animais , Pressão Sanguínea/efeitos dos fármacos , Pressão Sanguínea/fisiologia , Doxiciclina/farmacologia , Endotélio Vascular/efeitos dos fármacos , Endotélio Vascular/metabolismo , Expressão Gênica/efeitos dos fármacos , Imuno-Histoquímica , Técnicas In Vitro , Potenciais da Membrana/fisiologia , Artérias Mesentéricas/efeitos dos fármacos , Artérias Mesentéricas/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Músculo Liso Vascular/fisiologia , Técnicas de Patch-Clamp , Canais de Potássio/genética , RNA Mensageiro/metabolismo , Canais de Potássio Ativados por Cálcio de Condutância Baixa , Transgenes/efeitos dos fármacos , Grau de Desobstrução Vascular/genética , Grau de Desobstrução Vascular/fisiologia , Resistência Vascular/fisiologia
20.
Pharmacol Ther ; 93(2-3): 203-15, 2002.
Artigo em Inglês | MEDLINE | ID: mdl-12191612

RESUMO

The structural similarity of cyclic GMP-dependent protein kinase (cGPK) and cyclic AMP-dependent protein kinase (cAPK) has made it difficult to study cGPK pathways independent of those mediated by cAPK, primarily due to the lack of potent and selective cGPK inhibitors. We recently reported a novel peptide library screen specifically designed to select for tight-binding peptides that identified selective inhibitors of cGPK [Proc Natl Acad Sci USA, 97 (2000) 14772]. Iterative deconvolution of octameric library arrays on paper identified the sequence LRK(5)H (W45). Binding of W45 to cGPK resulted in selective inhibition of the kinase, with K(i) values of 0.8 microM and 560 microM for cGPK and cAPK, respectively. Cellular internalization of highly charged W45 was accomplished by N-terminal fusion of membrane translocation sequences from either the human immunodeficiency virus tyrosine aminotransferase protein (47-59) DT-2 or from the Drosophila Antennapedia homeodomain (43-58) DT-3, respectively. For both fusion peptides, DT-2 and DT-3, we observed a potentiating effect with respect to the inhibitory potency, with K(i) values 40- to 80-fold lower than W45. Fluorescein-labeled DT-2 and DT-3 demonstrated rapid translocation through the cytosol and nuclei in a time-dependent manner using cultured cells and intact tissue samples (cerebral arteries). The physiological effects of DT-2 and DT-3 as selective cGPK inhibitors in smooth muscle were studied in small intact arteries. Nitric oxide, a cyclic GMP/cGPK activator, elicited a concentration-dependent dilation of isolated rat cerebral arteries, which was markedly inhibited by DT-2 and DT-3. Collectively, these results indicate that DT-2 and DT-3 effectively inhibit nitric oxide-induced vasodilation, further emphasizing the central role for cGPK in the modulation of vascular contractility.


Assuntos
Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Proteínas Quinases Dependentes de GMP Cíclico/antagonistas & inibidores , Músculo Liso Vascular/enzimologia , Biblioteca de Peptídeos , Animais , Proteínas Quinases Dependentes de AMP Cíclico/isolamento & purificação , Proteínas Quinases Dependentes de GMP Cíclico/síntese química , Humanos , Músculo Liso Vascular/efeitos dos fármacos , Ratos , Transdução de Sinais
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA