Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
1.
Arterioscler Thromb Vasc Biol ; 37(4): 694-706, 2017 04.
Artigo em Inglês | MEDLINE | ID: mdl-28153878

RESUMO

OBJECTIVE: Increasing evidence suggests that contractile dysfunction in smooth muscle cells (SMCs) plays a critical role in aortic biomechanical dysfunction and aortic aneurysm and dissection (AAD) development. However, the mechanisms underlying SMC contractile dysfunction in sporadic AAD are poorly understood. In this study, we examined the role of the NLRP3 (nucleotide oligomerization domain-like receptor family, pyrin domain containing 3)-caspase-1 inflammasome, a key inflammatory cascade, in SMC contractile dysfunction in AAD. APPROACH AND RESULTS: We observed significant SMC contractile protein degradation in aortas from patients with sporadic thoracic AAD. The contractile protein degradation was associated with activation of the NLRP3-caspase-1 inflammasome cascade. In SMCs, caspase-1 bound and directly cleaved and degraded contractile proteins, leading to contractile dysfunction. Furthermore, Nlrp3 or caspase-1 deficiency in mice significantly reduced angiotensin II-induced contractile protein degradation, biomechanical dysfunction, and AAD formation in both thoracic and abdominal aortas. Finally, blocking this cascade with the inflammasome inhibitor, glyburide (an antidiabetic medication), reduced angiotensin II-induced AAD formation. CONCLUSIONS: Inflammasome-caspase-1-mediated degradation of SMC contractile proteins may contribute to aortic biomechanical dysfunction and AAD development. This cascade may be a therapeutic target in AAD formation. In addition, glyburide may have protective effects against AAD development.


Assuntos
Aneurisma da Aorta Abdominal/enzimologia , Aneurisma da Aorta Torácica/enzimologia , Dissecção Aórtica/enzimologia , Caspase 1/metabolismo , Inflamassomos/metabolismo , Proteínas Musculares/metabolismo , Músculo Liso Vascular/enzimologia , Miócitos de Músculo Liso/enzimologia , Proteína 3 que Contém Domínio de Pirina da Família NLR/metabolismo , Vasoconstrição , Idoso , Dissecção Aórtica/genética , Dissecção Aórtica/fisiopatologia , Dissecção Aórtica/prevenção & controle , Angiotensina II , Animais , Aorta Abdominal/efeitos dos fármacos , Aorta Abdominal/enzimologia , Aorta Abdominal/patologia , Aorta Abdominal/fisiopatologia , Aorta Torácica/efeitos dos fármacos , Aorta Torácica/enzimologia , Aorta Torácica/patologia , Aorta Torácica/fisiopatologia , Aneurisma da Aorta Abdominal/genética , Aneurisma da Aorta Abdominal/fisiopatologia , Aneurisma da Aorta Abdominal/prevenção & controle , Aneurisma da Aorta Torácica/genética , Aneurisma da Aorta Torácica/fisiopatologia , Aneurisma da Aorta Torácica/prevenção & controle , Fenômenos Biomecânicos , Caspase 1/deficiência , Caspase 1/genética , Células Cultivadas , Modelos Animais de Doenças , Feminino , Predisposição Genética para Doença , Glibureto/farmacologia , Humanos , Inflamassomos/antagonistas & inibidores , Inflamassomos/genética , Masculino , Camundongos da Linhagem 129 , Camundongos Endogâmicos C57BL , Camundongos Knockout , Pessoa de Meia-Idade , Músculo Liso Vascular/efeitos dos fármacos , Músculo Liso Vascular/patologia , Músculo Liso Vascular/fisiopatologia , Miócitos de Músculo Liso/efeitos dos fármacos , Miócitos de Músculo Liso/patologia , Proteína 3 que Contém Domínio de Pirina da Família NLR/deficiência , Proteína 3 que Contém Domínio de Pirina da Família NLR/genética , Fenótipo , Proteólise , Vasoconstrição/efeitos dos fármacos
2.
Crit Care Med ; 42(11): e692-701, 2014 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-25126877

RESUMO

OBJECTIVES: We previously reported the expression of the two-pore-domain K channel TREK-1 in lung epithelial cells and proposed a role for this channel in the regulation of alveolar epithelial cytokine secretion. In this study, we focused on investigating the role of TREK-1 in vivo in the development of hyperoxia-induced lung injury. DESIGN: Laboratory animal experiments. SETTING: University research laboratory. SUBJECTS: Wild-type and TREK-1-deficient mice. INTERVENTIONS: Mice were anesthetized and exposed to 1) room air, no mechanical ventilation, 2) 95% hyperoxia for 24 hours, and 3) 95% hyperoxia for 24 hours followed by mechanical ventilation for 4 hours. MEASUREMENTS AND MAIN RESULTS: Hyperoxia exposure accentuated lung injury in TREK-1-deficient mice but not controls, resulting in increase in lung injury scores, bronchoalveolar lavage fluid cell numbers, and cellular apoptosis and a decrease in quasi-static lung compliance. Exposure to a combination of hyperoxia and injurious mechanical ventilation resulted in further morphological lung damage and increased lung injury scores and bronchoalveolar lavage fluid cell numbers in control but not TREK-1-deficient mice. At baseline and after hyperoxia exposure, bronchoalveolar lavage cytokine levels were unchanged in TREK-1-deficient mice compared with controls. Exposure to hyperoxia and mechanical ventilation resulted in an increase in bronchoalveolar lavage interleukin-6, monocyte chemotactic protein-1, and tumor necrosis factor-α levels in both mouse types, but the increase in interleukin-6 and monocyte chemotactic protein-1 levels was less prominent in TREK-1-deficient mice than in controls. Lung tissue macrophage inflammatory protein-2, keratinocyte-derived cytokine, and interleukin-1ß gene expression was not altered by hyperoxia in TREK-1-deficient mice compared with controls. Furthermore, we show for the first time TREK-1 expression on alveolar macrophages and unimpaired tumor necrosis factor-α secretion from TREK-1-deficient macrophages. CONCLUSIONS: TREK-1 deficiency resulted in increased sensitivity of lungs to hyperoxia, but this effect is less prominent if overwhelming injury is induced by the combination of hyperoxia and injurious mechanical ventilation. TREK-1 may constitute a new potential target for the development of novel treatment strategies against hyperoxia-induced lung injury.


Assuntos
Lesão Pulmonar Aguda/patologia , Citocinas/metabolismo , Hiperóxia/complicações , Canais de Potássio de Domínios Poros em Tandem/deficiência , Lesão Pulmonar Aguda/etiologia , Lesão Pulmonar Aguda/terapia , Animais , Western Blotting , Líquido da Lavagem Broncoalveolar/química , Citocinas/genética , Modelos Animais de Doenças , Ensaio de Imunoadsorção Enzimática , Macrófagos Alveolares/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Microscopia Confocal/métodos , Canais de Potássio de Domínios Poros em Tandem/metabolismo , Distribuição Aleatória , Reação em Cadeia da Polimerase em Tempo Real , Valores de Referência , Respiração Artificial , Medição de Risco , Índice de Gravidade de Doença
3.
J Neurophysiol ; 109(9): 2250-9, 2013 May.
Artigo em Inglês | MEDLINE | ID: mdl-23390312

RESUMO

Slow afterhyperpolarizations (sAHPs) play an important role in establishing the firing pattern of neurons that in turn influence network activity. sAHPs are mediated by calcium-activated potassium channels. However, the molecular identity of these channels and the mechanism linking calcium entry to their activation are still unknown. Here we present several lines of evidence suggesting that the sAHPs in developing starburst amacrine cells (SACs) are mediated by two-pore potassium channels. First, we use whole cell and perforated patch voltage clamp recordings to characterize the sAHP conductance under different pharmacological conditions. We find that this conductance was calcium dependent, reversed at EK, blocked by barium, insensitive to apamin and TEA, and activated by arachidonic acid. In addition, pharmacological inhibition of calcium-activated phosphodiesterase reduced the sAHP. Second, we performed gene profiling on isolated SACs and found that they showed strong preferential expression of the two-pore channel gene kcnk2 that encodes TREK1. Third, we demonstrated that TREK1 knockout animals exhibited an altered frequency of retinal waves, a frequency that is set by the sAHPs in SACs. With these results, we propose a model in which depolarization-induced decreases in cAMP lead to disinhibition of the two-pore potassium channels and in which the kinetics of this biochemical pathway dictate the slow activation and deactivation of the sAHP conductance. Our model offers a novel pathway for the activation of a conductance that is physiologically important.


Assuntos
Células Amácrinas/fisiologia , Potenciais da Membrana , Canais de Potássio de Domínios Poros em Tandem/metabolismo , Potenciais de Ação , Células Amácrinas/efeitos dos fármacos , Células Amácrinas/metabolismo , Animais , Ácido Araquidônico/farmacologia , Cálcio/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Bloqueadores dos Canais de Potássio/farmacologia , Canais de Potássio de Domínios Poros em Tandem/genética , Transcrição Gênica
4.
Am J Physiol Regul Integr Comp Physiol ; 305(1): R60-7, 2013 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-23637138

RESUMO

K2P6.1 or TWIK-2, a two-pore domain K channel, is an important regulator of cardiovascular function. K2P6.1 is highly expressed in vascular smooth muscle and endothelium. Mice (8-12 wk) lacking functional K2P6.1 (K2P6.1(-/-)) are hypertensive and have enhanced vascular contractility. It is not known whether the lack of functional K2P6.1 in endothelium has a role in the vascular dysfunction in K2P6.1(-/-) mice. We tested the hypothesis: K2P6.1(-/-) mice have impaired endothelium-dependent relaxations. K2P6.1(-/-) mice were ∼35 mmHg more hypertensive than WT mice at both 8-12 wk (young adult) and 20-24 wk (mature mice, P < 0.01; n = 8-10). Endothelium-dependent relaxations of the thoracic aorta were evaluated by isometric myography after contraction with phenylephrine (10(-6) M). Maximal ACh-dependent relaxations were increased from 65 ± 1% to 73 ± 1% in the aorta from young adult (P < 0.01; n = 6) and from 45 ± 1% to 74 ± 1% in the aorta from mature (P < 0.001; n = 5) K2P6.1(-/-) mice compared with K2P6.1(+/+) littermates. However, in the aorta from young adult and mature K2P6.1(+/+) mice, 10(-5) M indomethacin, a cyclooxygenase inhibitor, increased maximal ACh relaxations to knockout levels. Enhanced relaxation was also seen with ATP, a P2Y purinergic agonist, and A23187, a nonreceptor-based agonist in mature K2P6.1(-/-) mice. Mature adult aorta from K2P6.1(-/-) showed an attenuated ACh-mediated contraction in the presence of nitro-l-arginine methyl ester (l-NAME) and without precontraction of 0.97 mN vs. 7.5 mN in K2P6.1(-/-) and K2P6.1(+/+) (P < 0.001; n = 5). In summary, K2P6.1(-/-) mice, which are hypertensive, have enhanced endothelium-dependent relaxations in the aorta due to the suppression of an indomethacin-sensitive constrictor component.


Assuntos
Aorta Torácica/fisiologia , Endotélio Vascular/fisiologia , Canais de Potássio de Domínios Poros em Tandem/deficiência , Canais de Potássio de Domínios Poros em Tandem/fisiologia , Vasodilatação/fisiologia , Animais , Calcimicina/farmacologia , Modelos Animais de Doenças , Hipertensão/etiologia , Hipertensão/fisiopatologia , Indometacina/farmacologia , Masculino , Camundongos , Camundongos Knockout , NG-Nitroarginina Metil Éster/farmacologia , Fenilefrina/farmacologia , Canais de Potássio de Domínios Poros em Tandem/genética , Vasoconstrição/efeitos dos fármacos , Vasoconstrição/fisiologia , Vasodilatação/efeitos dos fármacos
5.
Am J Physiol Regul Integr Comp Physiol ; 305(4): R334-42, 2013 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-23761641

RESUMO

Obstructive sleep apnea (OSA), a condition in which the upper airway collapses during sleep, is strongly associated with metabolic and cardiovascular diseases. Little is known how OSA affects the cerebral circulation. The goals of this study were 1) to develop a rat model of chronic OSA that involved apnea and 2) to test the hypothesis that 4 wk of apneas during the sleep cycle alters endothelium-mediated dilations in middle cerebral arteries (MCAs). An obstruction device, which was chronically implanted into the trachea of rats, inflated to obstruct the airway 30 times/h for 8 h during the sleep cycle. After 4 wk of apneas, MCAs were isolated, pressurized, and exposed to luminally applied ATP, an endothelial P2Y2 receptor agonist that dilates through endothelial-derived nitric oxide (NO) and endothelial-dependent hyperpolarization (EDH). Dilations to ATP were attenuated ~30% in MCAs from rats undergoing apneas compared with those from a sham control group (P < 0.04 group effect; n = 7 and 10, respectively). When the NO component of the dilation was blocked to isolate the EDH component, the response to ATP in MCAs from the sham and apnea groups was similar. This finding suggests that the attenuated dilation to ATP must occur through reduced NO. In summary, we have successfully developed a novel rat model for chronic OSA that incorporates apnea during the sleep cycle. Using this model, we demonstrate that endothelial dysfunction occurred by 4 wk of apnea, likely increasing the vulnerability of the brain to cerebrovascular related accidents.


Assuntos
Trifosfato de Adenosina/farmacologia , Modelos Animais de Doenças , Endotélio Vascular/efeitos dos fármacos , Artéria Cerebral Média/efeitos dos fármacos , Agonistas do Receptor Purinérgico P2Y/farmacologia , Apneia Obstrutiva do Sono/metabolismo , Vasodilatação/efeitos dos fármacos , Vasodilatadores/farmacologia , Animais , Fatores Biológicos/metabolismo , Doença Crônica , Relação Dose-Resposta a Droga , Endotélio Vascular/metabolismo , Endotélio Vascular/fisiopatologia , Masculino , Artéria Cerebral Média/metabolismo , Artéria Cerebral Média/fisiopatologia , Óxido Nítrico/metabolismo , Doadores de Óxido Nítrico/farmacologia , Óxido Nítrico Sintase Tipo III/metabolismo , Ratos , Ratos Long-Evans , Respiração , Sono , Apneia Obstrutiva do Sono/etiologia , Apneia Obstrutiva do Sono/fisiopatologia , Fatores de Tempo , Traqueia/fisiopatologia
6.
Am J Physiol Regul Integr Comp Physiol ; 299(2): R461-9, 2010 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-20357027

RESUMO

We tested the hypothesis that TREK-1, a two-pore domain K channel, is involved with dilations in arteries. Because there are no selective activators or inhibitors of TREK-1, we generated a mouse line deficient in TREK-1. Endothelium-mediated dilations were not different in arteries from wild-type (WT) and TREK-1 knockout (KO) mice. This includes dilations of the middle cerebral artery to ATP, dilations of the basilar artery to ACh, and relaxations of the aorta to carbachol, a cholinergic agonist. The nitric oxide (NO) and endothelium-dependent hyperpolarizing factor components of ATP dilations were identical in the middle cerebral arteries of WT and TREK-1 KO mice. Furthermore, the NO and cyclooxygenase-dependent components were identical in the basilar arteries of the different genotypes. Dilations of the basilar artery to alpha-linolenic acid, an activator of TREK-1, were not affected by the absence of TREK-1. Whole cell currents recorded using patch-clamp techniques were similar in cerebrovascular smooth muscle cells (CVSMCs) from WT and TREK-1 KO mice. alpha-linolenic acid or arachidonic acid increased whole cell currents in CVSMCs from both WT and TREK-1 KO mice. The selective blockers of large-conductance Ca-activated K channels, penitrem A and iberiotoxin, blocked the increased currents elicited by either alpha-linolenic or arachidonic acid. In summary, dilations were similar in arteries from WT and TREK-1 KO mice. There was no sign of TREK-1-like currents in CVSMCs from WT mice, and there were no major differences in currents between the genotypes. We conclude that regulation of arterial diameter is not altered in mice lacking TREK-1.


Assuntos
Artéria Basilar/metabolismo , Circulação Cerebrovascular , Artéria Cerebral Média/metabolismo , Canais de Potássio de Domínios Poros em Tandem/deficiência , Potássio/metabolismo , Vasodilatação , Potenciais de Ação , Animais , Aorta/metabolismo , Ácido Araquidônico/metabolismo , Artéria Basilar/efeitos dos fármacos , Circulação Cerebrovascular/efeitos dos fármacos , Relação Dose-Resposta a Droga , Genótipo , Canais de Potássio Ativados por Cálcio de Condutância Alta/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Artéria Cerebral Média/efeitos dos fármacos , Músculo Liso Vascular/metabolismo , Miócitos de Músculo Liso/metabolismo , Óxido Nítrico/metabolismo , Fenótipo , Bloqueadores dos Canais de Potássio/farmacologia , Canais de Potássio de Domínios Poros em Tandem/genética , Prostaglandina-Endoperóxido Sintases/metabolismo , Vasoconstrição , Vasoconstritores/farmacologia , Vasodilatação/efeitos dos fármacos , Vasodilatadores/farmacologia , Ácido alfa-Linolênico/metabolismo
7.
Atherosclerosis ; 192(1): 49-55, 2007 May.
Artigo em Inglês | MEDLINE | ID: mdl-16870193

RESUMO

Type 2 diabetes, a major risk factor for atherosclerosis, is associated with a cluster of lipid risk factors, many of which can be mechanistically linked with underlying dysregulated fatty acid metabolism and elevated plasma non-esterified fatty acids (NEFA). Thus, we tested the hypothesis that elevated NEFA dysregulates lipid metabolism at the levels of lipid synthesis and gene expression in THP-1 monocyte derived macrophages (MDM). THP-1 MDM incubated with oleic acid (OA) and a BODIPY-conjugated NEFA, accumulate, respectively, intracellular inclusions that are positive for oil red O and BODIPY-labeling. Parallel studies with [(14)C]OA show dose-dependent accumulation of intracellular (14)C-labeled neutral lipid, almost exclusively as triglyceride; the rate of [(3)H]OA uptake increases as THP-1 MDM convert to foam cells. Preincubation of THP-1 MDM with higher concentrations of OA (1.8mM versus 0.2mM) was associated with enhanced uptake of Ac-LDL, and increased expression of adipocyte fatty acid binding protein, FAT/CD36, and cyclooxygenase-2 (COX-2); COX-2 mass and activity also increased. These observations suggest a mechanistic link between sustained elevations in albumin-bound NEFA and foam cell formation that may be mediated by enhanced adipogenesis, increased uptake of modified LDL, and upregulated formation of eicosanoids, which may be proinflammatory.


Assuntos
Ciclo-Oxigenase 2/metabolismo , Ácidos Graxos não Esterificados/metabolismo , Células Espumosas/fisiologia , Macrófagos/metabolismo , Adipócitos/metabolismo , Albuminas/metabolismo , Antígenos CD36/metabolismo , Linhagem Celular , Perfilação da Expressão Gênica , Humanos , Metabolismo dos Lipídeos , Dados de Sequência Molecular , Ácido Oleico/metabolismo
8.
Front Cell Neurosci ; 10: 13, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-26869883

RESUMO

We have recently shown that a linear current-to-voltage (I-V) relationship of membrane conductance (passive conductance) reflects the intrinsic property of K(+) channels in mature astrocytes. While passive conductance is known to underpin a highly negative and stable membrane potential (V M) essential for the basic homeostatic function of astrocytes, a complete repertoire of the involved K(+) channels remains elusive. TREK-1 two-pore domain K(+) channel (K2P) is highly expressed in astrocytes, and covalent association of TREK-1 with TWIK-1, another highly expressed astrocytic K2P, has been reported as a mechanism underlying the trafficking of heterodimer TWIK-1/TREK-1 channel to the membrane and contributing to astrocyte passive conductance. To decipher the individual contribution of TREK-1 and address whether the appearance of passive conductance is conditional to the co-expression of TWIK-1/TREK-1 in astrocytes, TREK-1 single and TWIK-1/TREK-1 double gene knockout mice were used in the present study. The relative quantity of mRNA encoding other astrocyte K(+) channels, such as Kir4.1, Kir5.1, and TREK-2, was not altered in these gene knockout mice. Whole-cell recording from hippocampal astrocytes in situ revealed no detectable changes in astrocyte passive conductance, V M, or membrane input resistance (R in) in either kind of gene knockout mouse. Additionally, TREK-1 proteins were mainly located in the intracellular compartments of the hippocampus. Altogether, genetic deletion of TREK-1 alone or together with TWIK-1 produced no obvious alteration in the basic electrophysiological properties of hippocampal astrocytes. Thus, future research focusing on other K(+) channels may shed light on this long-standing and important question in astrocyte physiology.

9.
J Cereb Blood Flow Metab ; 35(3): 402-11, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-25425077

RESUMO

Obstructive sleep apnea (OSA) is associated with cerebrovascular diseases. However, little is known regarding the effects of OSA on the cerebrovascular wall. We tested the hypothesis that OSA augments endothelin-1 (ET-1) constrictions of cerebral arteries. Repeated apneas (30 or 60 per hour) were produced in rats during the sleep cycle (8 hours) by remotely inflating a balloon implanted in the trachea. Four weeks of apneas produced a 23-fold increase in ET-1 sensitivity in isolated and pressurized posterior cerebral arteries (PCAs) compared with PCAs from sham-operated rats (EC50=10(-9.2) mol/L versus 10(-10.6) mol/L; P<0.001). This increased sensitivity was abolished by the ET-B receptor antagonist, BQ-788. Constrictions to the ET-B receptor agonist, IRL-1620, were greater in PCAs from rats after 2 or 4 weeks of apneas compared with that from sham-operated rats (P=0.013). Increased IRL-1620 constrictions in PCAs from OSA rats were normalized with the transient receptor potential channel (TRPC) blocker, SKF96365, or the Rho kinase (ROCK) inhibitor, Y27632. These data show that OSA increases the sensitivity of PCAs to ET-1 through enhanced ET-B activity, and enhanced activity of TRPCs and ROCK. We conclude that enhanced ET-1 signaling is part of a pathologic mechanism associated with adverse cerebrovascular outcomes of OSA.


Assuntos
Circulação Cerebrovascular/fisiologia , Endotelina-1/metabolismo , Receptores de Endotelina/metabolismo , Transdução de Sinais/fisiologia , Apneia Obstrutiva do Sono/metabolismo , Animais , Western Blotting , Modelos Animais de Doenças , Ensaio de Imunoadsorção Enzimática , Imuno-Histoquímica , Ratos , Reação em Cadeia da Polimerase em Tempo Real , Vasoconstrição/fisiologia
10.
Hypertension ; 66(4): 913-7, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-26259594

RESUMO

We tested the hypothesis that apneas during the sleep cycle exacerbate hypertension and accelerate changes that occur with cerebral small vessel disease. Obstructive sleep apnea was modeled by intermittent inflations of a chronically implanted tracheal balloon to occlude the airway during the sleep cycle (termed OSA) in spontaneously hypertensive stroke-prone (SHRSP) rats, a model of cerebral small vessel disease. SHRSP rats and their parent strain, Wistar Kyoto (WKY) rats, were exposed to OSA for 2 weeks (from 9 to 11 or from 18 to 20 weeks). At 9 weeks, hypertension was developing in the SHRSP rats and was firmly established by 18 weeks. OSA exposure increased systolic blood pressure in SHRSP rats by ≈30 mm Hg in both age groups compared with shams that were surgically prepared but not exposed to OSA (P<0.05). OSA exposure also increased systolic blood pressure in WKY rats by 20 and 37 mm Hg at 11 and 20 weeks, respectively (P<0.05). OSA exposure in SHRSP rats compromised blood-brain barrier integrity in white matter at both 11 and 20 weeks of age when compared with SHRSP sham rats (P<0.05). Microglia were activated in SHRSP rats exposed to OSA but not in sham rats at 11 weeks (P<0.05). At 20 weeks, microglia were activated in sham SHRSP rats (P<0.05) compared with WKY sham rats and were not further activated by OSA. Neither was blood-brain barrier integrity altered nor microglia activated in any of the WKY groups. We conclude that OSA accelerates the onset of the cerebral pathologies associated with cerebral small vessel disease in SHRSP, but not WKY, rats.


Assuntos
Doenças de Pequenos Vasos Cerebrais/patologia , Apneia Obstrutiva do Sono/complicações , Sono , Substância Branca/patologia , Animais , Doenças de Pequenos Vasos Cerebrais/etiologia , Doenças de Pequenos Vasos Cerebrais/fisiopatologia , Modelos Animais de Doenças , Masculino , Ratos , Ratos Endogâmicos SHR , Ratos Endogâmicos WKY , Apneia Obstrutiva do Sono/fisiopatologia
11.
Hypertension ; 64(6): 1260-5, 2014 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-25245387

RESUMO

TWIK-2 (KCNK6) is a member of the 2-pore domain (K2P) family of potassium channels, which are highly expressed in the vascular system. We tested the hypothesis that TWIK-2 deficiency leads to pulmonary hypertension. TWIK-2 knockout mice and their wildtype littermates at 8 weeks of age had similar mean right ventricular systolic pressures (24±3 and 21±3 mm Hg, respectively.) Significantly, by 20 weeks of age, the mean right ventricular systolic pressures in TWIK-2 knockout mice increased to 35±3 mm Hg (P≤0.036), whereas mean right ventricular systolic pressures in wildtype littermates remained at 22±3 mm Hg. Elevated mean right ventricular systolic pressures in the TWIK-2 knockout mice was accompanied by pulmonary vascular remodeling as determined by a 25% increase in the cross-sectional area of the vessels occupied by the vessel wall. Additionally, secondary branches of the pulmonary artery from 20-week-old TWIK-2 knockout mice showed an enhanced contractile response to U46619 (10(-6) moles/L), a thromboxane A2 mimetic, which was completely abolished with the Rho-kinase inhibitor, Y27632 (10(-6) and 10(-5) moles/L). Treatment of TWIK-2 knockout mice with the Rho-kinase inhibitor, fasudil, in the drinking water for 12 weeks, abolished the development of pulmonary hypertension and attenuated the vessel remodeling. We concluded that mice deficient in the TWIK-2 channel develop pulmonary hypertension between 8 and 20 weeks of age through a mechanism involving Rho-kinase. Our results suggest that downregulation of TWIK-2 in the pulmonary vasculature may be an underlying mechanism in the development of pulmonary hypertension.


Assuntos
DNA/genética , Regulação da Expressão Gênica , Hipertensão Pulmonar/genética , Canais de Potássio de Domínios Poros em Tandem/deficiência , Artéria Pulmonar/metabolismo , Quinases Associadas a rho/genética , Animais , Pressão Sanguínea , Modelos Animais de Doenças , Genótipo , Hipertensão Pulmonar/metabolismo , Hipertensão Pulmonar/fisiopatologia , Masculino , Camundongos , Camundongos Knockout , Canais de Potássio de Domínios Poros em Tandem/genética , Artéria Pulmonar/fisiopatologia , Função Ventricular Direita , Quinases Associadas a rho/biossíntese
12.
Basic Clin Pharmacol Toxicol ; 113(4): 250-8, 2013 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-23724868

RESUMO

Polyunsaturated fatty acid (PUFA)-activated two-pore domain potassium channels (K2P ) have been proposed to be expressed in the pulmonary vasculature. However, their physiological or pathophysiological roles are poorly defined. Here, we tested the hypothesis that PUFA-activated K2P are involved in pulmonary vasorelaxation and that alterations of channel expression are pathophysiologically linked to pulmonary hypertension. Expression of PUFA-activated K2P in the murine lung was investigated by quantitative reverse-transcription polymerase chain reaction (qRT-PCR), immunohistochemistry (IHC), by patch clamp (PC) and myography. K2P -gene expression was examined in chronic hypoxic mice. qRT-PCR showed that the K2P 2.1 and K2P 6.1 were the predominantly expressed K2P in the murine lung. IHC revealed protein expression of K2P 2.1 and K2P 6.1 in the endothelium of pulmonary arteries and of K2P 6.1 in bronchial epithelium. PC showed pimozide-sensitive K2P -like K(+) -current activated by docosahexaenoic acid (DHA) in freshly isolated endothelial cells as well as DHA-induced membrane hyperpolarization. Myography on pulmonary arteries showed that DHA induced concentration-dependent instantaneous relaxations that were resistant to endothelial removal and inhibition of NO and prostacyclin synthesis and to a cocktail of blockers of calcium-activated K(+) channels but were abolished by high extracellular (30 mM) K(+) -concentration. Gene expression and protein of K2P 2.1 were not altered in chronic hypoxic mice, while K2P 6.1 was up-regulated by fourfold. In conclusion, the PUFA-activated K2P 2.1 and K2P 6.1 are expressed in murine lung and functional K2P -like channels contribute to endothelium hyperpolarization and pulmonary artery relaxation. The increased K2P 6.1-gene expression may represent a novel counter-regulatory mechanism in pulmonary hypertension and suggest that arterial K2P 2.1 and K2P 6.1 could be novel therapeutic targets.


Assuntos
Ácidos Docosa-Hexaenoicos/farmacologia , Hipertensão Pulmonar/patologia , Hipóxia/patologia , Canais de Potássio de Domínios Poros em Tandem/metabolismo , Animais , Eletrofisiologia , Epoprostenol/antagonistas & inibidores , Epoprostenol/biossíntese , Hipertensão Pulmonar/tratamento farmacológico , Hipertensão Pulmonar/etiologia , Hipóxia/complicações , Hipóxia/tratamento farmacológico , Pulmão/efeitos dos fármacos , Pulmão/metabolismo , Masculino , Camundongos , Canais de Potássio de Domínios Poros em Tandem/genética , Artéria Pulmonar/efeitos dos fármacos , Artéria Pulmonar/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Regulação para Cima , Vasodilatação/efeitos dos fármacos
13.
Hypertension ; 58(4): 672-8, 2011 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-21876070

RESUMO

K(2P)6.1, a member of the 2-pore domain K channel family, is highly expressed in the vascular system; however, its function is unknown. We tested the following hypotheses. K(2P)6.1 regulates the following: (1) systemic blood pressure; (2) the contractile state of arteries; (3) vascular smooth muscle cell migration; (4) proliferation; and/or (5) volume regulation. Mice lacking K(2P)6.1 (KO) were generated by deleting exon 1 of Kcnk6. Mean arterial blood pressure in both anesthetized and awake KO mice was increased by 17±2 and 26±3 mm Hg, respectively (P<0.05). The resting membrane potential in freshly dispersed vascular smooth muscle cells was depolarized by 17±2 mV in the KO compared with wild-type littermates (P<0.05). The contractile responses to KCl (P<0.05) and BAY K 8644 (P<0.01), an activator of L-type calcium channels, were enhanced in isolated segments of aorta from KO mice. However, there was no difference in the current density of L-type calcium channels. Responses to U46619, an agent that activates rho kinase, showed an enhanced contraction in aorta from KO mice (P<0.001). The BAY K 8644-mediated increase in contraction was decreased to wild-type levels when treated with Y27632, a rho kinase inhibitor, (P<0.05). K(2P)6.1 does not appear to be involved with migration, proliferation, or volume regulation in cultured vascular smooth muscle cells. We conclude that K(2P)6.1 deficiency induces vascular dysfunction and hypertension through a mechanism that may involve smooth muscle cell depolarization and enhanced rho kinase activity.


Assuntos
Aorta/fisiopatologia , Hipertensão/etiologia , Hipertensão/fisiopatologia , Músculo Liso Vascular/fisiopatologia , Canais de Potássio de Domínios Poros em Tandem/fisiologia , Ácido 15-Hidroxi-11 alfa,9 alfa-(epoximetano)prosta-5,13-dienoico/farmacologia , Éster Metílico do Ácido 3-Piridinacarboxílico, 1,4-Di-Hidro-2,6-Dimetil-5-Nitro-4-(2-(Trifluormetil)fenil)/farmacologia , Animais , Aorta/efeitos dos fármacos , Pressão Sanguínea/efeitos dos fármacos , Pressão Sanguínea/fisiologia , Agonistas dos Canais de Cálcio/farmacologia , Modelos Animais de Doenças , Potenciais da Membrana/efeitos dos fármacos , Potenciais da Membrana/fisiologia , Camundongos , Camundongos Knockout , Músculo Liso Vascular/efeitos dos fármacos , Canais de Potássio de Domínios Poros em Tandem/deficiência , Canais de Potássio de Domínios Poros em Tandem/genética , Cloreto de Potássio/farmacologia , Vasoconstrição/efeitos dos fármacos , Vasoconstrição/fisiologia , Vasoconstritores/farmacologia , Quinases Associadas a rho/fisiologia
14.
Am J Physiol Heart Circ Physiol ; 296(6): H1774-80, 2009 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-19363137

RESUMO

Two-pore domain K(+) (K(2P)) channels are a new channel family. The goal of this study was to determine if K(2P) channels are activated by the nitric oxide (NO)/cGMP/PKG pathway in vascular smooth muscle. Relative levels of message for K(2P) channels were assessed in rat middle cerebral arteries (MCAs) using quantitative RT-PCR, and K(+) currents were measured in freshly dispersed vascular smooth muscle cells of the MCA. The rat MCA expresses a number of K(2P) channels. Message for TREK-1 was the most abundant K(2P) channel, followed by TASK-1 and TWIK-2, which were expressed at approximately 10% of the level of TREK-1. Message for other K(2P) channels was 1% or less than that of TREK-1. A number of K(2P) channels, including TREK-1, TWIK-2, and TASK-1, have putative PKG phosphorylation sites in the intracellular domains. The NO donor sodium nitroprusside (100 muM) or the membrane permeable analog of cGMP 8-bromo-cGMP (10 muM) elicited transient increases in whole cell current of vascular smooth muscle from the rat MCA. However, after large-conductance Ca(2+)-activated K(+) channels had been blocked with 10 mM tetraethylammonium (TEA), no increase in whole cell current was observed. Since K(2P) channels are resistant to the blocking effects of TEA, we conclude that K(2P) channels in vascular smooth muscle were not activated by the NO/cGMP/PKG pathway. Although K(2P) channels are highly expressed, K(2P) currents are not activated via the NO/cGMP pathway in rat MCA smooth muscle, despite the presence of numerous putative PKG phosphorylation sites.


Assuntos
Circulação Cerebrovascular/fisiologia , GMP Cíclico/metabolismo , Artéria Cerebral Média/fisiologia , Músculo Liso Vascular/fisiologia , Canais de Potássio de Domínios Poros em Tandem/metabolismo , Animais , Proteínas Quinases Dependentes de GMP Cíclico/metabolismo , Expressão Gênica/fisiologia , Masculino , Proteínas do Tecido Nervoso , Óxido Nítrico/metabolismo , Fosforilação , Canais de Potássio de Domínios Poros em Tandem/química , Canais de Potássio de Domínios Poros em Tandem/genética , Estrutura Terciária de Proteína , Ratos , Ratos Long-Evans
15.
Exp Biol Med (Maywood) ; 234(12): 1493-502, 2009 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-19934370

RESUMO

TWIK-2, a member of the Two-Pore Domain K channel family, is expressed in a number of mammalian tissues including the vascular system. The function of TWIK-2 is not known. The purpose of this study was to clone the TWIK-2 channel from the rat middle cerebral artery, express it in CHO cells, and characterize the channel's electrical properties. In light of the fact that there are no specific TWIK-2 inhibitors or activators, a better characterization of the channel should enhance our understanding of its role in the vascular system. TWIK-2 was cloned from the rat middle cerebral artery and expressed with an N-terminal green fluorescence protein (GFP) in CHO cells. We report that rTWIK-2-GFP currents were relatively linear at physiological K(+) concentrations but become slightly inwardly rectifying in symmetrical K(+). rTWIK-2-GFP was insensitive to 10 mM TEA, 3 mM 4-aminopyridine, and 10 microM glibenclamide. However, rTWIK-2-GFP was inhibited by Ba(2+) with 50% of the current being blocked at 80 microM. rTWIK-2-GFP activity was enhanced 60% by 100 microM arachidonic acid. The electrophysiological characteristics of TWIK-2 indicate that it could serve an important role in ion homeostasis and regulation of the membrane potential in arteries and arterioles.


Assuntos
Potenciais da Membrana/fisiologia , Artéria Cerebral Média/metabolismo , Canais de Potássio de Domínios Poros em Tandem/biossíntese , Animais , Ácido Araquidônico/farmacologia , Bário/farmacologia , Células CHO , Clonagem Molecular , Cricetinae , Cricetulus , Relação Dose-Resposta a Droga , Expressão Gênica , Masculino , Potenciais da Membrana/efeitos dos fármacos , Potássio/metabolismo , Canais de Potássio de Domínios Poros em Tandem/genética , Ratos , Ratos Long-Evans
16.
Artigo em Inglês | MEDLINE | ID: mdl-19964748

RESUMO

Isoflurane is the most commonly used anesthetic in mice. We studied the effect of low and high levels of isoflurane (also a potent coronary vasodilator) on aortic impedance in mice. Aortic impedance was determined using pressure and flow velocity signals at baseline (B, pentobarbital anesthesia), low (Isol, 1%), and high (Iso2.5, 2.5%) levels of isoflurane. Significant differences were observed in peak and mean flow velocities, systolic, diastolic, mean and pulse pressures at B and Iso2.5. However in impedance indices only peripheral vascular resistance was significantly different. No changes were observed in the harmonic components that represent pulsatile characteristics of the aorta. Peak left ventricular (LV) pressure was significantly lower at Iso2.5 when compared to B, but +/-dP/dt and tau (time constant of LV relaxation) did not change significantly indicating that LV contractility was unaffected. These results show that various levels of isoflurane cause significant changes in vascular hemodynamics and care must be taken to minimize these differences when using isoflurane as an anesthesia.


Assuntos
Anestésicos Inalatórios/farmacologia , Aorta/efeitos dos fármacos , Isoflurano/farmacologia , Resistência Vascular/efeitos dos fármacos , Animais , Aorta/fisiologia , Engenharia Biomédica , Circulação Coronária/efeitos dos fármacos , Hemodinâmica/efeitos dos fármacos , Camundongos , Vasodilatadores/farmacologia
17.
J Lipid Res ; 48(2): 348-57, 2007 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-17102149

RESUMO

Small, dense, electronegative low density lipoprotein [LDL(-)] is increased in patients with familial hypercholesterolemia and diabetes, populations at increased risk for coronary artery disease. It is present to a lesser extent in normolipidemic subjects. The mechanistic link between small, dense LDL(-) and atherogenesis is not known. To begin to address this, we studied the composition and dynamics of small, dense LDL(-) from normolipidemic subjects. NEFA levels, which correlate with triglyceride content, are quantitatively linked to LDL electronegativity. Oxidized LDL is not specific to small, dense LDL(-) or lipoprotein [a] (i.e., abnormal lipoprotein). Apolipoprotein C-III is excluded from the most abundant LDL (i.e., that of intermediate density: 1.034 < d < 1.050 g/ml) but associated with both small and large LDL(-). In contrast, lipoprotein-associated phospholipase A(2) (LpPLA(2)) is highly enriched only in small, dense LDL(-). The association of LpPLA(2) with LDL may occur through amphipathic helical domains that are displaced from the LDL surface by contraction of the neutral lipid core.


Assuntos
Lipoproteínas LDL/química , Lipoproteínas LDL/metabolismo , Fosfolipases A2/metabolismo , Apolipoproteína C-III/análise , Eletroquímica , Eletroforese em Gel de Poliacrilamida , Ácidos Graxos não Esterificados/química , Humanos , Lipoproteínas HDL/química , Lipoproteínas HDL/metabolismo , Oxirredução , Ligação Proteica , Eletricidade Estática
18.
Am J Physiol Heart Circ Physiol ; 291(2): H770-80, 2006 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-16565299

RESUMO

Little is known about the presence and function of two-pore domain K(+) (K(2P)) channels in vascular smooth muscle cells (VSMCs). Five members of the K(2P) channel family are known to be directly activated by arachidonic acid (AA). The purpose of this study was to determine 1) whether AA-sensitive K(2P) channels are expressed in cerebral VSMCs and 2) whether AA dilates the rat middle cerebral artery (MCA) by increasing K+ currents in VSMCs via an atypical K+ channel. RT-PCR revealed message for the following AA-sensitive K(2P) channels in rat MCA: tandem of P domains in weak inward rectifier K+ (TWIK-2), TWIK-related K+ (TREK-1 and TREK-2), TWIK-related AA-stimulated K+ (TRAAK), and TWIK-related halothane-inhibited K+ (THIK-1) channels. However, in isolated VSMCs, only message for TWIK-2 was found. Western blotting showed that TWIK-2 is present in MCA, and immunohistochemistry further demonstrated its presence in VSMCs. AA (10-100 microM) dilated MCAs through an endothelium-independent mechanism. AA-induced dilation was not affected by inhibition of cyclooxygenase, epoxygenase, or lipoxygenase or inhibition of classical K+ channels with 10 mM TEA, 3 mM 4-aminopyridine, 10 microM glibenclamide, or 100 microM Ba2+. AA-induced dilations were blocked by 50 mM K+, indicating involvement of a K+ channel. AA (10 microM) increased whole cell K+ currents in dispersed cerebral VSMCs. AA-induced currents were not affected by inhibitors of the AA metabolic pathways or blockade of classical K+ channels. We conclude that AA dilates the rat MCA and increases K+ currents in VSMCs via an atypical K+ channel that is likely a member of the K(2P) channel family.


Assuntos
Artérias Cerebrais/metabolismo , Canais de Potássio de Domínios Poros em Tandem/metabolismo , Actinas/metabolismo , Animais , Ácido Araquidônico/metabolismo , Western Blotting , Artérias Cerebrais/efeitos dos fármacos , Eletrofisiologia , Imuno-Histoquímica , Técnicas In Vitro , Masculino , Potenciais da Membrana/efeitos dos fármacos , Músculo Liso Vascular/citologia , Músculo Liso Vascular/efeitos dos fármacos , Músculo Liso Vascular/metabolismo , Bloqueadores dos Canais de Potássio/farmacologia , Canais de Potássio/metabolismo , Canais de Potássio de Domínios Poros em Tandem/efeitos dos fármacos , Ratos , Ratos Long-Evans , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Vasodilatação/efeitos dos fármacos
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa