Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 28
Filtrar
1.
Circ Res ; 130(10): 1531-1546, 2022 05 13.
Artigo em Inglês | MEDLINE | ID: mdl-35382561

RESUMO

Capillaries are equipped to sense neurovascular coupling agents released onto the outer wall of a capillary, translating these external signals into electrical/Ca2+ changes that play a crucial role in blood flow regulation and ensuring that neuronal demands are met. However, control mechanisms attributable to forces imposed onto the lumen are less clear. Here, we show that Piezo1 channels act as mechanosensors in central nervous system capillaries. Electrophysiological analyses confirmed expression and function of Piezo1 channels in brain cortical and retinal capillaries. Activation of Piezo1 channels evoked currents that were sensitive to endothelial cell-specific Piezo1 deletion. Using genetically encoded Ca2+ indicator mice and an ex vivo pressurized retina preparation, we found that activation of Piezo1 channels by mechanical forces triggered Ca2+ signals in capillary endothelial cells. Collectively, these findings indicate that Piezo1 channels are capillary mechanosensors that initiate crucial Ca2+ signals and could, therefore, have a profound impact on central nervous system blood flow control.


Assuntos
Capilares , Canais Iônicos , Acoplamento Neurovascular , Animais , Sistema Nervoso Central/irrigação sanguínea , Células Endoteliais/metabolismo , Canais Iônicos/genética , Canais Iônicos/metabolismo , Camundongos
2.
Proc Natl Acad Sci U S A ; 118(17)2021 04 27.
Artigo em Inglês | MEDLINE | ID: mdl-33875602

RESUMO

Cerebral small vessel diseases (SVDs) are a central link between stroke and dementia-two comorbidities without specific treatments. Despite the emerging consensus that SVDs are initiated in the endothelium, the early mechanisms remain largely unknown. Deficits in on-demand delivery of blood to active brain regions (functional hyperemia) are early manifestations of the underlying pathogenesis. The capillary endothelial cell strong inward-rectifier K+ channel Kir2.1, which senses neuronal activity and initiates a propagating electrical signal that dilates upstream arterioles, is a cornerstone of functional hyperemia. Here, using a genetic SVD mouse model, we show that impaired functional hyperemia is caused by diminished Kir2.1 channel activity. We link Kir2.1 deactivation to depletion of phosphatidylinositol 4,5-bisphosphate (PIP2), a membrane phospholipid essential for Kir2.1 activity. Systemic injection of soluble PIP2 rapidly restored functional hyperemia in SVD mice, suggesting a possible strategy for rescuing functional hyperemia in brain disorders in which blood flow is disturbed.


Assuntos
Doenças de Pequenos Vasos Cerebrais/etiologia , Circulação Cerebrovascular , Hiperemia/etiologia , Fosfatidilinositol 4,5-Difosfato/metabolismo , Canais de Potássio Corretores do Fluxo de Internalização/metabolismo , Animais , Doenças de Pequenos Vasos Cerebrais/metabolismo , Modelos Animais de Doenças , Células Endoteliais/metabolismo , Hiperemia/metabolismo , Masculino , Camundongos Transgênicos
3.
Int J Mol Sci ; 24(22)2023 Nov 14.
Artigo em Inglês | MEDLINE | ID: mdl-38003472

RESUMO

Functional hyperemia-activity-dependent increases in local blood perfusion-underlies the on-demand delivery of blood to regions of enhanced neuronal activity, a process that is crucial for brain health. Importantly, functional hyperemia deficits have been linked to multiple dementia risk factors, including aging, chronic hypertension, and cerebral small vessel disease (cSVD). We previously reported crippled functional hyperemia in a mouse model of genetic cSVD that was likely caused by depletion of phosphatidylinositol 4,5-bisphosphate (PIP2) in capillary endothelial cells (EC) downstream of impaired epidermal growth factor receptor (EGFR) signaling. Here, using EC-specific EGFR-knockout (KO) mice, we directly examined the role of endothelial EGFR signaling in functional hyperemia, assessed by measuring increases in cerebral blood flow in response to contralateral whisker stimulation using laser Doppler flowmetry. Molecular characterizations showed that EGFR expression was dramatically decreased in freshly isolated capillaries from EC-EGFR-KO mice, as expected. Notably, whisker stimulation-induced functional hyperemia was significantly impaired in these mice, an effect that was rescued by administration of PIP2, but not by the EGFR ligand, HB-EGF. These data suggest that the deletion of the EGFR specifically in ECs attenuates functional hyperemia, likely via depleting PIP2 and subsequently incapacitating Kir2.1 channel functionality in capillary ECs. Thus, our study underscores the role of endothelial EGFR signaling in functional hyperemia of the brain.


Assuntos
Células Endoteliais , Hiperemia , Camundongos , Animais , Células Endoteliais/metabolismo , Receptores ErbB/genética , Receptores ErbB/metabolismo , Encéfalo/metabolismo , Família de Proteínas EGF/metabolismo , Família de Proteínas EGF/farmacologia , Fator de Crescimento Epidérmico/metabolismo
4.
Proc Natl Acad Sci U S A ; 113(2): E229-38, 2016 Jan 12.
Artigo em Inglês | MEDLINE | ID: mdl-26627720

RESUMO

Sour taste is detected by a subset of taste cells on the tongue and palate epithelium that respond to acids with trains of action potentials. Entry of protons through a Zn(2+)-sensitive proton conductance that is specific to sour taste cells has been shown to be the initial event in sour taste transduction. Whether this conductance acts in concert with other channels sensitive to changes in intracellular pH, however, is not known. Here, we show that intracellular acidification generates excitatory responses in sour taste cells, which can be attributed to block of a resting K(+) current. We identify KIR2.1 as the acid-sensitive K(+) channel in sour taste cells using pharmacological and RNA expression profiling and confirm its contribution to sour taste with tissue-specific knockout of the Kcnj2 gene. Surprisingly, acid sensitivity is not conferred on sour taste cells by the specific expression of Kir2.1, but by the relatively small magnitude of the current, which makes the cells exquisitely sensitive to changes in intracellular pH. Consistent with a role of the K(+) current in amplifying the sensory response, entry of protons through the Zn(2+)-sensitive conductance produces a transient block of the KIR2.1 current. The identification in sour taste cells of an acid-sensitive K(+) channel suggests a mechanism for amplification of sour taste and may explain why weak acids that produce intracellular acidification, such as acetic acid, taste more sour than strong acids.


Assuntos
Canais de Potássio Corretores do Fluxo de Internalização/metabolismo , Prótons , Transdução de Sinais , Paladar/fisiologia , Ácidos/farmacologia , Potenciais de Ação/efeitos dos fármacos , Animais , Canais de Cálcio/metabolismo , Células HEK293 , Humanos , Concentração de Íons de Hidrogênio , Integrases/metabolismo , Espaço Intracelular/metabolismo , Ativação do Canal Iônico/efeitos dos fármacos , Camundongos Knockout , Modelos Biológicos , Especificidade de Órgãos/efeitos dos fármacos , Receptores de Superfície Celular/metabolismo , Transdução de Sinais/efeitos dos fármacos , Canais de Cátion TRPM/metabolismo , Paladar/efeitos dos fármacos , Papilas Gustativas/citologia , Papilas Gustativas/efeitos dos fármacos , Papilas Gustativas/metabolismo , Zinco/farmacologia
5.
Microcirculation ; 25(1)2018 01.
Artigo em Inglês | MEDLINE | ID: mdl-29247493

RESUMO

Cerebral SVDs encompass a group of genetic and sporadic pathological processes leading to brain lesions, cognitive decline, and stroke. There is no specific treatment for SVDs, which progress silently for years before becoming clinically symptomatic. Here, we examine parallels in the functional defects of PAs in CADASIL, a monogenic form of SVD, and in response to SAH, a common type of hemorrhagic stroke that also targets the brain microvasculature. Both animal models exhibit dysregulation of the voltage-gated potassium channel, KV 1, in arteriolar myocytes, an impairment that compromises responses to vasoactive stimuli and impacts CBF autoregulation and local dilatory responses to neuronal activity (NVC). However, the extent to which this channelopathy-like defect ultimately contributes to these pathologies is unknown. Combining experimental data with computational modeling, we describe the role of KV 1 channels in the regulation of myocyte membrane potential at rest and during the modest increase in extracellular potassium associated with NVC. We conclude that PA resting membrane potential and myogenic tone depend strongly on KV 1.2/1.5 channel density, and that reciprocal changes in KV channel density in CADASIL and SAH produce opposite effects on extracellular potassium-mediated dilation during NVC.


Assuntos
Microvasos/patologia , Canais de Potássio de Abertura Dependente da Tensão da Membrana/análise , Animais , CADASIL/fisiopatologia , Dilatação , Humanos , Canais de Potássio de Abertura Dependente da Tensão da Membrana/fisiologia , Hemorragia Subaracnóidea/fisiopatologia
6.
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
7.
Proc Natl Acad Sci U S A ; 111(20): 7462-7, 2014 May 20.
Artigo em Inglês | MEDLINE | ID: mdl-24808139

RESUMO

Studies of stress effects on the brain have traditionally focused on neurons, without considering the cerebral microcirculation. Here we report that stress impairs neurovascular coupling (NVC), the process that matches neuronal activity with increased local blood flow. A stressed phenotype was induced in male rats by administering a 7-d heterotypical stress paradigm. NVC was modeled by measuring parenchymal arteriole (PA) vasodilation in response to neuronal stimulation in amygdala brain slices. After stress, vasodilation of PAs to neuronal stimulation was greatly reduced, and dilation of isolated PAs to external K(+) was diminished, suggesting a defect in smooth muscle inwardly rectifying K(+) (KIR) channel function. Consistent with these observations, stress caused a reduction in PA KIR2.1 mRNA and smooth muscle KIR current density, and blocking KIR channels significantly inhibited NVC in control, but not in stressed, slices. Delivery of corticosterone for 7 d (without stressors) or RU486 (before stressors) mimicked and abrogated NVC impairment by stress, respectively. We conclude that stress causes a glucocorticoid-mediated decrease in functional KIR channels in amygdala PA myocytes. This renders arterioles less responsive to K(+) released from astrocytic endfeet during NVC, leading to impairment of this process. Because the fidelity of NVC is essential for neuronal health, the impairment characterized here may contribute to the pathophysiology of brain disorders with a stress component.


Assuntos
Glucocorticoides/metabolismo , Canais de Potássio Corretores do Fluxo de Internalização/metabolismo , Transdução de Sinais , Tonsila do Cerebelo/efeitos dos fármacos , Animais , Arteríolas/efeitos dos fármacos , Astrócitos/metabolismo , Encéfalo/efeitos dos fármacos , Encéfalo/fisiopatologia , Corticosterona/química , Glucose/metabolismo , Antagonistas de Hormônios/química , Sistema Límbico/efeitos dos fármacos , Masculino , Mifepristona/química , Neurônios/fisiologia , Oxigênio/metabolismo , Fenótipo , Ratos , Ratos Sprague-Dawley , Estresse Psicológico , Fatores de Tempo , Vasodilatação/efeitos dos fármacos
8.
Proc Natl Acad Sci U S A ; 110(15): 6157-62, 2013 Apr 09.
Artigo em Inglês | MEDLINE | ID: mdl-23530219

RESUMO

In the CNS, astrocytes are sensory and regulatory hubs that play important roles in cerebral homeostatic processes, including matching local cerebral blood flow to neuronal metabolism (neurovascular coupling). These cells possess a highly branched network of processes that project from the soma to neuronal synapses as well as to arterioles and capillaries, where they terminate in "endfeet" that encase the blood vessels. Ca(2+) signaling within the endfoot mediates neurovascular coupling; thus, these functional microdomains control vascular tone and local perfusion in the brain. Transient receptor potential vanilloid 4 (TRPV4) channels--nonselective cation channels with considerable Ca(2+) conductance--have been identified in astrocytes, but their function is largely unknown. We sought to characterize the influence of TRPV4 channels on Ca(2+) dynamics in the astrocytic endfoot microdomain and assess their role in neurovascular coupling. We identified local TRPV4-mediated Ca(2+) oscillations in endfeet and further found that TRPV4 Ca(2+) signals are amplified and propagated by Ca(2+)-induced Ca(2+) release from inositol trisphosphate receptors (IP3Rs). Moreover, TRPV4-mediated Ca(2+) influx contributes to the endfoot Ca(2+) response to neuronal activation, enhancing the accompanying vasodilation. Our results identify a dynamic synergy between TRPV4 channels and IP3Rs in astrocyte endfeet and demonstrate that TRPV4 channels are engaged in and contribute to neurovascular coupling.


Assuntos
Astrócitos/citologia , Cálcio/metabolismo , Canais de Cátion TRPV/metabolismo , Animais , Encéfalo/metabolismo , Encéfalo/patologia , Sinalização do Cálcio , Sistema Nervoso Central/fisiologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Neurônios/metabolismo , Oscilometria
9.
Physiology (Bethesda) ; 29(5): 343-60, 2014 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-25180264

RESUMO

Endothelial cells and smooth muscle cells of resistance arteries mediate opposing responses to mechanical forces acting on the vasculature, promoting dilation in response to flow and constriction in response to pressure, respectively. In this review, we explore the role of TRP channels, particularly endothelial TRPV4 and smooth muscle TRPC6 and TRPM4 channels, in vascular mechanosensing circuits, placing their putative mechanosensitivity in context with other proposed upstream and downstream signaling pathways.


Assuntos
Artérias/citologia , Membrana Celular/metabolismo , Células Endoteliais/metabolismo , Miócitos de Músculo Liso/metabolismo , Canais de Cátion TRPM/metabolismo , Animais , Células Endoteliais/patologia , Humanos , Transdução de Sinais/fisiologia
10.
bioRxiv ; 2023 Sep 17.
Artigo em Inglês | MEDLINE | ID: mdl-37745396

RESUMO

Functional hyperemia - activity-dependent increases in local blood perfusion - underlies the on-demand delivery of blood to regions of enhanced neuronal activity, a process that is crucial for brain health. Importantly, functional hyperemia deficits have been linked to multiple dementia risk factors, including aging, chronic hypertension, and cerebral small vessel disease (cSVD). We previously reported crippled functional hyperemia in a mouse model of genetic cSVD that was likely caused by depletion of phosphatidylinositol 4,5-bisphosphate (PIP2) in capillary endothelial cells (EC) downstream of impaired epidermal growth factor receptor (EGFR) signaling. Here, using EC-specific EGFR-knockout (KO) mice, we directly examined the role of endothelial EGFR signaling in functional hyperemia, assessed by measuring increases in cerebral blood flow in response to contralateral whisker stimulation using laser Doppler flowmetry. Molecular characterizations showed that EGFR expression was dramatically decreased in freshly isolated capillaries from EC-EGFR-KO mice, as expected. Notably, whisker stimulation-induced functional hyperemia was significantly impaired in these mice, an effect that was rescued by exogenous administration of PIP2, but not by the EGFR ligand, HB-EGF. These data suggest that the deletion of the EGFR specifically in ECs depletes PIP2 and attenuates functional hyperemia, underscoring the central role of the endothelial EGFR signaling in cerebral blood flow regulation.

11.
J Clin Invest ; 131(18)2021 09 15.
Artigo em Inglês | MEDLINE | ID: mdl-34351870

RESUMO

Dementia resulting from small vessel diseases (SVDs) of the brain is an emerging epidemic for which there is no treatment. Hypertension is the major risk factor for SVDs, but how hypertension damages the brain microcirculation is unclear. Here, we show that chronic hypertension in a mouse model progressively disrupts on-demand delivery of blood to metabolically active areas of the brain (functional hyperemia) through diminished activity of the capillary endothelial cell inward-rectifier potassium channel, Kir2.1. Despite similar efficacy in reducing blood pressure, amlodipine, a voltage-dependent calcium-channel blocker, prevented hypertension-related damage to functional hyperemia whereas losartan, an angiotensin II type 1 receptor blocker, did not. We attribute this drug class effect to losartan-induced aldosterone breakthrough, a phenomenon triggered by pharmacological interruption of the renin-angiotensin pathway leading to elevated plasma aldosterone levels. This hypothesis is supported by the finding that combining losartan with the aldosterone receptor antagonist eplerenone prevented the hypertension-related decline in functional hyperemia. Collectively, these data suggest Kir2.1 as a possible therapeutic target in vascular dementia and indicate that concurrent mineralocorticoid aldosterone receptor blockade may aid in protecting against late-life cognitive decline in hypertensive patients treated with angiotensin II type 1 receptor blockers.


Assuntos
Anti-Hipertensivos/uso terapêutico , Doenças de Pequenos Vasos Cerebrais/tratamento farmacológico , Doenças de Pequenos Vasos Cerebrais/etiologia , Hiperemia/tratamento farmacológico , Hipertensão/complicações , Hipertensão/tratamento farmacológico , Anlodipino/uso terapêutico , Bloqueadores do Receptor Tipo 1 de Angiotensina II/administração & dosagem , Bloqueadores do Receptor Tipo 1 de Angiotensina II/uso terapêutico , Animais , Anti-Hipertensivos/administração & dosagem , Doenças de Pequenos Vasos Cerebrais/fisiopatologia , Circulação Cerebrovascular/efeitos dos fármacos , Circulação Cerebrovascular/fisiologia , Demência Vascular/tratamento farmacológico , Demência Vascular/etiologia , Demência Vascular/fisiopatologia , Modelos Animais de Doenças , Quimioterapia Combinada , Eplerenona/administração & dosagem , Eplerenona/uso terapêutico , Fatores de Risco de Doenças Cardíacas , Humanos , Hiperemia/fisiopatologia , Losartan/administração & dosagem , Losartan/uso terapêutico , Masculino , Camundongos , Microvasos/efeitos dos fármacos , Microvasos/fisiopatologia , Canais de Potássio Corretores do Fluxo de Internalização/efeitos dos fármacos , Canais de Potássio Corretores do Fluxo de Internalização/fisiologia , Sistema Renina-Angiotensina/efeitos dos fármacos , Sistema Renina-Angiotensina/fisiologia
12.
J Neurotrauma ; 34(1): 192-203, 2017 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-26757855

RESUMO

Endothelial dysfunction is a hallmark of many chronic diseases, including diabetes and long-term hypertension. We show that acute traumatic brain injury (TBI) leads to endothelial dysfunction in rat mesenteric arteries. Endothelial-dependent dilation was greatly diminished 24 h after TBI because of impaired nitric oxide (NO) production. The activity of arginase, which competes with endothelial NO synthase (eNOS) for the common substrate l-arginine, were also significantly increased in arteries, suggesting that arginase-mediated depletion of l-arginine underlies diminished NO production. Consistent with this, substrate restoration by exogenous application of l-arginine or inhibition of arginase recovered endothelial function. Moreover, evidence for increased reactive oxygen species production, a consequence of l-arginine starvation-dependent eNOS uncoupling, was detected in endothelium and plasma. Collectively, our findings demonstrate endothelial dysfunction in a remote vascular bed after TBI, manifesting as impaired endothelial-dependent vasodilation, with increased arginase activity, decreased generation of NO, and increased O2- production. We conclude that blood vessels have a "molecular memory" of neurotrauma, 24 h after injury, because of functional changes in vascular endothelial cells; these effects are pertinent to understanding the systemic inflammatory response that occurs after TBI even in the absence of polytrauma.


Assuntos
Arginase/metabolismo , Lesões Encefálicas Traumáticas/enzimologia , Endotélio Vascular/enzimologia , Microcirculação/fisiologia , Óxido Nítrico Sintase Tipo III/metabolismo , Acetilcolina/farmacologia , Animais , Lesões Encefálicas Traumáticas/patologia , Endotélio Vascular/efeitos dos fármacos , Endotélio Vascular/patologia , Masculino , Microcirculação/efeitos dos fármacos , Ratos , Ratos Sprague-Dawley , Vasodilatação/efeitos dos fármacos , Vasodilatação/fisiologia
13.
Physiol Rep ; 5(16)2017 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-28830977

RESUMO

The aim of this study was to investigate mechanisms by which adiponectin influences vascular Ca2+ signaling, K+ channel activity and thus contractile tone of small arteries. Vasodilation to adiponectin was studied in mesenteric resistance arteries constricted with intraluminal pressure. Ca2+ signals were characterized using high speed confocal microscopy of intact arteries. Patch clamp investigated the effect of adiponectin on individual VSMC potassium (K+) channel currents. Adiponectin dilated arteries constricted with pressure-induced tone by approximately 5% and the induced vasodilation was only transient. The dilation to adiponectin was reduced by pharmacological interruption of the Ca2+ spark/large conductance activated K+ (BK) channel pathway but from a physiological perspective, interpretation of the data was limited by the small effect. Neither Adiponectin nor the presence of intact perivascular adipose tissue (PVAT) influenced Ca2+ spark or Ca2+ wave frequency or characteristics. Studied using a perforated patch approach, Adiponectin marginally increased current through the VSMC BK channel but this effect was lost using the whole cell technique with dialysis of the cytoplasm. Adiponectin did not change the frequency or amplitude of Ca2+ spark-induced transient outward currents (STOC). Overall, our study shows that Adiponectin induces only a small and transient dilation of pressure constricted mesenteric arteries. This vasodilatory effect is likely to be independent of Ca2+ sparks or direct BK channel activation.


Assuntos
Adiponectina/farmacologia , Artérias/metabolismo , Sinalização do Cálcio , Canais de Potássio Ativados por Cálcio de Condutância Alta/metabolismo , Miócitos de Músculo Liso/metabolismo , Vasoconstrição , Animais , Artérias/citologia , Artérias/fisiologia , Células Cultivadas , Camundongos , Camundongos Endogâmicos C57BL , Contração Muscular , Músculo Liso Vascular/efeitos dos fármacos , Músculo Liso Vascular/metabolismo , Músculo Liso Vascular/fisiologia , Miócitos de Músculo Liso/efeitos dos fármacos , Miócitos de Músculo Liso/fisiologia
14.
J Cereb Blood Flow Metab ; 36(3): 492-512, 2016 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-26661232

RESUMO

One hundred and twenty five years ago, Roy and Sherrington made the seminal observation that neuronal stimulation evokes an increase in cerebral blood flow.(1) Since this discovery, researchers have attempted to uncover how the cells of the neurovascular unit-neurons, astrocytes, vascular smooth muscle cells, vascular endothelial cells and pericytes-coordinate their activity to control this phenomenon. Recent work has revealed that ionic fluxes through a diverse array of ion channel species allow the cells of the neurovascular unit to engage in multicellular signaling processes that dictate local hemodynamics.In this review we center our discussion on two major themes: (1) the roles of ion channels in the dynamic modulation of parenchymal arteriole smooth muscle membrane potential, which is central to the control of arteriolar diameter and therefore must be harnessed to permit changes in downstream cerebral blood flow, and (2) the striking similarities in the ion channel complements employed in astrocytic endfeet and endothelial cells, enabling dual control of smooth muscle from either side of the blood-brain barrier. We conclude with a discussion of the emerging roles of pericyte and capillary endothelial cell ion channels in neurovascular coupling, which will provide fertile ground for future breakthroughs in the field.


Assuntos
Arteríolas/fisiologia , Encéfalo/irrigação sanguínea , Circulação Cerebrovascular , Canais Iônicos/metabolismo , Músculo Liso Vascular/fisiologia , Animais , Astrócitos/metabolismo , Barreira Hematoencefálica/fisiologia , Encéfalo/fisiologia , Sinalização do Cálcio , Humanos , Potenciais da Membrana
15.
J Cereb Blood Flow Metab ; 36(7): 1195-201, 2016 07.
Artigo em Inglês | MEDLINE | ID: mdl-27052838

RESUMO

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


Assuntos
Arteríolas/metabolismo , Ataque Isquêmico Transitório/fisiopatologia , Músculo Liso Vascular/metabolismo , Acoplamento Neurovascular/fisiologia , Tecido Parenquimatoso/irrigação sanguínea , Canais de Potássio Corretores do Fluxo de Internalização/metabolismo , Animais , Arteríolas/fisiopatologia , Circulação Cerebrovascular/fisiologia , Endotélio Vascular , Ataque Isquêmico Transitório/metabolismo , Masculino , Músculo Liso Vascular/fisiopatologia , Ratos Sprague-Dawley
16.
Sci Signal ; 9(449): ra100, 2016 10 11.
Artigo em Inglês | MEDLINE | ID: mdl-27729550

RESUMO

Activation of Ca2+-sensitive, large-conductance potassium (BK) channels in vascular smooth muscle cells (VSMCs) by local, ryanodine receptor-mediated Ca2+ signals (Ca2+ sparks) acts as a brake on pressure-induced (myogenic) vasoconstriction-a fundamental mechanism that regulates blood flow in small resistance arteries. We report that physiological intraluminal pressure within resistance arteries activated cGMP-dependent protein kinase (PKG) in VSMCs through oxidant-induced formation of an intermolecular disulfide bond between cysteine residues. Oxidant-activated PKG was required to trigger Ca2+ sparks, BK channel activity, and vasodilation in response to pressure. VSMCs from arteries from mice expressing a form of PKG that could not be activated by oxidants showed reduced Ca2+ spark frequency, and arterial preparations from these mice had decreased pressure-induced activation of BK channels. Thus, the absence of oxidative activation of PKG disabled the BK channel-mediated negative feedback regulation of vasoconstriction. Our results support the concept of a negative feedback control mechanism that regulates arterial diameter through mechanosensitive production of oxidants to activate PKG and enhance Ca2+ sparks.


Assuntos
Pressão Sanguínea/fisiologia , Sinalização do Cálcio/fisiologia , Proteínas Quinases Dependentes de GMP Cíclico/metabolismo , Canais de Potássio Ativados por Cálcio de Condutância Alta/metabolismo , Mecanotransdução Celular/fisiologia , Músculo Liso Vascular/metabolismo , Miócitos de Músculo Liso/metabolismo , Vasoconstrição/fisiologia , Animais , Proteínas Quinases Dependentes de GMP Cíclico/genética , Canais de Potássio Ativados por Cálcio de Condutância Alta/genética , Camundongos , Camundongos Mutantes
17.
Trends Cardiovasc Med ; 13(2): 56-62, 2003 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-12586440

RESUMO

First identified in activated T cells, the calcium (Ca2+)-dependent transcription factor, nuclear factor of activated T cells (NFAT), has since been shown to play a role in nonimmune cells, including cells of the cardiovascular system. In arterial smooth muscle, the diverse array of calcium-signaling modalities, the functional interplay between smooth muscle and endothelial cells, and the influence of intravascular pressure on calcium and other signaling pathways creates a calcium-regulatory environment that is arguably unique. This review focuses on mechanisms that control the initial Ca2+/calcineurin-dependent events in NFAT activation, with a particular emphasis on NFAT regulation in native vascular smooth muscle. Also addressed is the role of additional mechanisms that act to modulate calcineurin-dependent NFAT nuclear import/export, mechanisms that may have particular relevance in this tissue.


Assuntos
Proteínas de Ligação a DNA/fisiologia , Músculo Liso Vascular/citologia , Proteínas Nucleares , Fatores de Transcrição/fisiologia , Cardiomegalia/fisiopatologia , Sistema Cardiovascular/citologia , Sistema Cardiovascular/fisiopatologia , Humanos , Fatores de Transcrição NFATC
18.
Sci Signal ; 7(333): ra66, 2014 Jul 08.
Artigo em Inglês | MEDLINE | ID: mdl-25005230

RESUMO

Endothelial cell dysfunction, characterized by a diminished response to endothelial cell-dependent vasodilators, is a hallmark of hypertension. TRPV4 channels play a major role in endothelial-dependent vasodilation, a function mediated by local Ca(2+) influx through clusters of functionally coupled TRPV4 channels rather than by a global increase in endothelial cell Ca(2+). We showed that stimulation of muscarinic acetylcholine receptors on endothelial cells of mouse arteries exclusively activated TRPV4 channels that were localized at myoendothelial projections (MEPs), specialized regions of endothelial cells that contact smooth muscle cells. Muscarinic receptor-mediated activation of TRPV4 depended on protein kinase C (PKC) and the PKC-anchoring protein AKAP150, which was concentrated at MEPs. Cooperative opening of clustered TRPV4 channels specifically amplified Ca(2+) influx at MEPs. Cooperativity of TRPV4 channels at non-MEP sites was much lower, and cooperativity at MEPs was greatly reduced by chelation of intracellular Ca(2+) or AKAP150 knockout, suggesting that Ca(2+) entering through adjacent channels underlies the AKAP150-dependent potentiation of TRPV4 activity. In a mouse model of angiotensin II-induced hypertension, MEP localization of AKAP150 was disrupted, muscarinic receptor stimulation did not activate TRPV4 channels, cooperativity among TRPV4 channels at MEPs was weaker, and vasodilation in response to muscarinic receptor stimulation was reduced. Thus, endothelial-dependent dilation of resistance arteries is enabled by MEP-localized AKAP150, which ensures the proximity of PKC to TRPV4 channels and the coupled channel gating necessary for efficient communication from endothelial to smooth muscle cells in arteries. Disruption of this molecular assembly may contribute to altered blood flow in hypertension.


Assuntos
Proteínas de Ancoragem à Quinase A/metabolismo , Endotélio Vascular/metabolismo , Hipertensão/metabolismo , Ativação do Canal Iônico , Canais de Cátion TRPV/metabolismo , Vasodilatação , Proteínas de Ancoragem à Quinase A/genética , Angiotensina II/efeitos adversos , Angiotensina II/farmacologia , Animais , Sinalização do Cálcio , Modelos Animais de Doenças , Endotélio Vascular/patologia , Hipertensão/induzido quimicamente , Hipertensão/genética , Hipertensão/patologia , Camundongos , Camundongos Knockout , Canais de Cátion TRPV/genética , Vasoconstritores/efeitos adversos , Vasoconstritores/farmacologia
19.
Sci Signal ; 7(327): ra49, 2014 May 27.
Artigo em Inglês | MEDLINE | ID: mdl-24866019

RESUMO

Maintaining constant blood flow in the face of fluctuations in blood pressure is a critical autoregulatory feature of cerebral arteries. An increase in pressure within the artery lumen causes the vessel to constrict through depolarization and contraction of the encircling smooth muscle cells. This pressure-sensing mechanism involves activation of two types of transient receptor potential (TRP) channels: TRPC6 and TRPM4. We provide evidence that the activation of the γ1 isoform of phospholipase C (PLCγ1) is critical for pressure sensing in cerebral arteries. Inositol 1,4,5-trisphosphate (IP3), generated by PLCγ1 in response to pressure, sensitized IP3 receptors (IP3Rs) to Ca(2+) influx mediated by the mechanosensitive TRPC6 channel, synergistically increasing IP3R-mediated Ca(2+) release to activate TRPM4 currents, leading to smooth muscle depolarization and constriction of isolated cerebral arteries. Proximity ligation assays demonstrated colocalization of PLCγ1 and TRPC6 with TRPM4, suggesting the presence of a force-sensitive, local signaling network comprising PLCγ1, TRPC6, TRPM4, and IP3Rs. Src tyrosine kinase activity was necessary for stretch-induced TRPM4 activation and myogenic constriction, consistent with the ability of Src to activate PLCγ isoforms. We conclude that contraction of cerebral artery smooth muscle cells requires the integration of pressure-sensing signaling pathways and their convergence on IP3Rs, which mediate localized Ca(2+)-dependent depolarization through the activation of TRPM4.


Assuntos
Pressão Sanguínea/fisiologia , Artérias Cerebrais/fisiologia , Fosfolipase C gama/metabolismo , Transdução de Sinais/fisiologia , Canais de Cátion TRPM/metabolismo , Vasoconstrição/fisiologia , Análise de Variância , Animais , Células HEK293 , Humanos , Imuno-Histoquímica , Receptores de Inositol 1,4,5-Trifosfato/metabolismo , Masculino , Potenciais da Membrana/fisiologia , Técnicas de Patch-Clamp , Interferência de RNA , Ratos , Ratos Sprague-Dawley , Reação em Cadeia da Polimerase em Tempo Real , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Canais de Cátion TRPC/metabolismo , Canal de Cátion TRPC6 , Quinases da Família src/metabolismo
20.
J Cereb Blood Flow Metab ; 33(4): 479-82, 2013 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-23385200

RESUMO

It has been proposed that prostaglandin E(2) (PGE(2)) is released from astrocytic endfeet to dilate parenchymal arterioles through activation of prostanoid (EP(4)) receptors during neurovascular coupling. However, the direct effects of PGE(2) on isolated parenchymal arterioles have not been tested. Here, we examined the effects of PGE(2) on the diameter of isolated pressurized parenchymal arterioles from rat and mouse brain. Contrary to the prevailing assumption, we found that PGE(2) (0.1, 1, and 5 µmol/L) constricted rather than dilated parenchymal arterioles. Vasoconstriction to PGE(2) was prevented by inhibitors of EP(1) receptors. These results strongly argue against a direct role of PGE(2) on arterioles during neurovascular coupling.


Assuntos
Astrócitos/metabolismo , Encéfalo/irrigação sanguínea , Circulação Cerebrovascular/fisiologia , Dinoprostona/metabolismo , Vasoconstrição/fisiologia , Animais , Arteríolas/metabolismo , Encéfalo/metabolismo , Masculino , Camundongos , Ratos , Ratos Sprague-Dawley , Receptores de Prostaglandina E Subtipo EP1/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA