Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 38
Filtrar
Mais filtros

Base de dados
Tipo de documento
País de afiliação
Intervalo de ano de publicação
1.
Purinergic Signal ; 2023 Nov 08.
Artigo em Inglês | MEDLINE | ID: mdl-37938538

RESUMO

Adenosine receptor (AR) suppresses inflammation and fibrosis by activating cyclic adenosine monophosphate (cAMP) signaling. We investigated whether altered AR expression contributes to the development of fibrotic diseases and whether A2AAR and A2BAR upregulation inhibits fibrotic responses. Primary human lung fibroblasts (HLFs) from normal (NHLFs) or patients with idiopathic pulmonary fibrosis (DHLF) were used for in vitro testing. Murine models of fibrotic liver or pulmonary disease were developed by injecting thioacetamide intraperitoneally, by feeding a high-fat diet, or by intratracheal instillation of bleomycin. Modafinil, which activates cAMP signaling via A2AAR and A2BAR, was administered orally. The protein amounts of A2AAR, A2BAR, and exchange protein directly activated by cAMP (Epac) were reduced, while collagen and α-smooth muscle actin (α-SMA) were elevated in DHLFs compared to NHLFs. In liver or lung tissue from murine models of fibrotic diseases, A2AAR and A2BAR were downregulated, but A1AR and A3AR were not. Epac amounts decreased, and amounts of collagen, α-SMA, KCa2.3, and KCa3.1 increased compared to the control. Modafinil restored the amounts of A2AAR, A2BAR, and Epac, and reduced collagen, α-SMA, KCa2.3, and KCa3.1 in murine models of fibrotic diseases. Transforming growth factor-ß reduced the amounts of A2AAR, A2BAR, and Epac, and elevated collagen, α-SMA, KCa2.3, and KCa3.1 in NHLFs; however, these alterations were inhibited by modafinil. Our investigation revealed that A2AAR and A2BAR downregulation induced liver and lung fibrotic diseases while upregulation attenuated fibrotic responses, suggesting that A2AAR and A2BAR-upregulating agents, such as modafinil, may serve as novel therapies for fibrotic diseases.

2.
Circ Res ; 123(5): e5-e19, 2018 08 17.
Artigo em Inglês | MEDLINE | ID: mdl-30030219

RESUMO

RATIONALE: Circulating CTRP1 (C1q/TNF-α [tumor necrosis factor-α]-related protein 1) levels are increased in hypertensive patients compared with those in healthy subjects. Nonetheless, little is known about the molecular and physiological function of CTRP1 in blood pressure (BP) regulation. OBJECTIVE: To investigate the physiological/pathophysiological role of CTRP1 in BP regulation. METHODS AND RESULTS: CTRP1 production was increased to maintain normotension under dehydration conditions, and this function was impaired in inducible CTRP1 KO (knockout) mice (CTRP1 ΔCAG). The increase in CTRP1 under dehydration conditions was mediated by glucocorticoids, and the antagonist mifepristone prevented the increase in CTRP1 and attenuated BP recovery. Treatment with a synthetic glucocorticoid increased the transcription, translation, and secretion of CTRP1 from skeletal muscle cells. Functionally, CTRP1 increases BP through the stimulation of the AT1R (Ang II [angiotensin II] receptor 1)-Rho (Ras homolog gene family)/ROCK (Rho kinase)-signaling pathway to induce vasoconstriction. CTRP1 promoted AT1R plasma membrane trafficking through phosphorylation of AKT and AKT substrate of 160 kDa (AS160). In addition, the administration of an AT1R blocker, losartan, recovered the hypertensive phenotype of CTRP1 TG (transgenic) mice. CONCLUSIONS: For the first time, we provide evidence that CTRP1 contributes to the regulation of BP homeostasis by preventing dehydration-induced hypotension.


Assuntos
Adipocinas/metabolismo , Pressão Sanguínea , Desidratação/metabolismo , Hipotensão/metabolismo , Adipocinas/genética , Bloqueadores do Receptor Tipo 1 de Angiotensina II/uso terapêutico , Animais , Linhagem Celular , Células Cultivadas , Desidratação/complicações , Desidratação/fisiopatologia , Feminino , Glucocorticoides/metabolismo , Humanos , Hipotensão/tratamento farmacológico , Hipotensão/etiologia , Hipotensão/fisiopatologia , Losartan/uso terapêutico , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Músculo Esquelético/metabolismo , Transporte Proteico , Proteínas Proto-Oncogênicas c-akt/metabolismo , Receptor Tipo 1 de Angiotensina/metabolismo , Vasoconstrição , Quinases Associadas a rho/metabolismo
3.
Pediatr Res ; 80(1): 119-27, 2016 07.
Artigo em Inglês | MEDLINE | ID: mdl-26959484

RESUMO

BACKGROUND: Pulmonary arterial hypertension (PAH) progressively leads to increases in pulmonary vasoconstriction. Modafinil plays a role in vasorelaxation and blocking KCa3.1 channel with a result of elevating intracellular cyclic adenosine monophosphate (cAMP) levels. The purpose of this study is to evaluate the effects on modafinil in monocrotaline (MCT)-induced PAH rat. METHODS: The rats were separated into three groups: the control group, the monocrotaline (M) group (MCT 60 mg/kg), and the modafinil (MD) group (MCT 60 mg/kg + modafinil). RESULTS: Reduced right ventricular pressure (RVP) was observed in the MD group. Right ventricular hypertrophy was improved in the MD group. Reduced number of intra-acinar pulmonary arteries and medial wall thickness were noted in the MD group. After the administration of modafinil, protein expressions of endothelin-1 (ET-1), endothelin receptor A (ERA) and KCa3.1 channel were significantly reduced. Modafinil suppressed pulmonary artery smooth muscle cell (PASMC) proliferation via cAMP and KCa3.1 channel. Additionally, we confirmed protein expressions such as Bcl-2-associated X, vascular endothelial growth factor, tumor necrosis factor-α, and interleukin-6 were reduced in the MD group. CONCLUSION: Modafinil improved PAH by vasorelaxation and a decrease in medial thickening via ET-1, ERA, and KCa3.1 down regulation. This is a meaningful study of a modafinil in PAH model.


Assuntos
Compostos Benzidrílicos/farmacologia , Hipertensão Pulmonar/induzido quimicamente , Hipertensão Pulmonar/tratamento farmacológico , Canais de Potássio Ativados por Cálcio de Condutância Intermediária/genética , Animais , Peso Corporal , AMP Cíclico/metabolismo , Modelos Animais de Doenças , Endotelina-1/metabolismo , Regulação da Expressão Gênica , Ventrículos do Coração/patologia , Humanos , Masculino , Modafinila , Monocrotalina , Miócitos de Músculo Liso/patologia , Pressão , Artéria Pulmonar/patologia , Ratos , Ratos Sprague-Dawley , Receptor de Endotelina A/metabolismo , Vasoconstrição
4.
Biochem Biophys Res Commun ; 468(4): 883-8, 2015 Dec 25.
Artigo em Inglês | MEDLINE | ID: mdl-26592662

RESUMO

Fabry disease is an X-linked lysosomal storage disorder that is caused by a deficiency of α-galactosidase A. The disease ultimately manifests as multiple organ dysfunctions owing to excessive accumulation of globotriaosylceramide (Gb3). Among the several complications of Fabry disease, ascending thoracic aortic aneurysm is relatively common, which is classically associated with connective tissue disorders characterized by abnormal defects or deficiencies in structural proteins such as collagen and elastin. Although an elevated Gb3 level is regarded as a prerequisite for the manifestations of Fabry disease, only this excess accumulation cannot explain the pathophysiology of these complications. Recently, an increased plasma level of lyso-Gb3 was suggested as a new biomarker in Fabry disease. Therefore, the aim of this study was to assess the effects of lyso-Gb3 on the pathogenesis of thoracic ascending aortic aneurysms in Fabry disease, with a particular focus on the responses related to aortic remodeling by fibroblasts. We found that lyso-Gb3 inhibited the growth of fibroblasts, as well as their differentiation into myofibroblasts, and collagen expression. Moreover, all of these compromised responses could be attributed to the effects of lyso-Gb3 on downregulation of KCa3.1 channel expression, and these impairments could be rescued when activating the KCa3.1 channel or increasing intracellular Ca(2+) concentration. This study provides new evidence that lyso-Gb3 inhibits the differentiation into myofibroblasts and collagen synthesis of fibroblasts owing to decreased Ca(2+) levels by KCa3.1 channel dysfunction. These findings suggest that the KCa3.1 channel can serve as a new target to attenuate and prevent development of ascending thoracic aortic aneurysm in Fabry disease.


Assuntos
Cálcio/metabolismo , Colágeno/biossíntese , Fibroblastos/citologia , Fibroblastos/fisiologia , Glicolipídeos/metabolismo , Canais de Potássio Ativados por Cálcio de Condutância Intermediária/metabolismo , Esfingolipídeos/metabolismo , Animais , Diferenciação Celular/efeitos dos fármacos , Diferenciação Celular/fisiologia , Proliferação de Células/efeitos dos fármacos , Proliferação de Células/fisiologia , Regulação para Baixo/efeitos dos fármacos , Regulação para Baixo/fisiologia , Fibroblastos/efeitos dos fármacos , Regulação da Expressão Gênica/efeitos dos fármacos , Regulação da Expressão Gênica/fisiologia , Glicolipídeos/administração & dosagem , Camundongos , Células NIH 3T3 , Esfingolipídeos/administração & dosagem
5.
Arterioscler Thromb Vasc Biol ; 34(1): 81-9, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-24158513

RESUMO

OBJECTIVE: Globotriaosylceramide (Gb3) induces KCa3.1 downregulation in Fabry disease (FD). We investigated whether Gb3 induces KCa3.1 endocytosis and degradation. APPROACH AND RESULTS: KCa3.1, especially plasma membrane-localized KCa3.1, was downregulated in both Gb3-treated mouse aortic endothelial cells (MAECs) and human umbilical vein endothelial cells. Gb3-induced KCa3.1 downregulation was prevented by lysosomal inhibitors but not by a proteosomal inhibitor. Endoplasmic reticulum stress-inducing agents did not induce KCa3.1 downregulation. Gb3 upregulated the protein levels of early endosome antigen 1 and lysosomal-associated membrane protein 2 in MAECs. Compared with MAECs from age-matched wild-type mice, those from aged α-galactosidase A (Gla)-knockout mice, an animal model of FD, showed downregulated KCa3.1 expression and upregulated early endosome antigen 1 and lysosomal-associated membrane protein 2 expression. In contrast, no significant difference was found in early endosome antigen 1 and lysosomal-associated membrane protein 2 expression between young Gla-knockout and wild-type MAECs. In aged Gla-knockout MAECs, clathrin was translocated close to the cell border and clathrin knockdown recovered KCa3.1 expression. Rab5, an effector of early endosome antigen 1, was upregulated, and Rab5 knockdown restored KCa3.1 expression, the current, and endothelium-dependent relaxation. CONCLUSIONS: -Gb3 accelerates the endocytosis and lysosomal degradation of endothelial KCa3.1 via a clathrin-dependent process, leading to endothelial dysfunction in FD.


Assuntos
Células Endoteliais/enzimologia , Endotélio Vascular/enzimologia , Doença de Fabry/enzimologia , Canais de Potássio Ativados por Cálcio de Condutância Intermediária/metabolismo , Lisossomos/metabolismo , Triexosilceramidas/metabolismo , Animais , Células Cultivadas , Clatrina/genética , Clatrina/metabolismo , Modelos Animais de Doenças , Endocitose , Endotélio Vascular/fisiopatologia , Doença de Fabry/genética , Doença de Fabry/fisiopatologia , Células Endoteliais da Veia Umbilical Humana/enzimologia , Humanos , Proteína 2 de Membrana Associada ao Lisossomo/metabolismo , Camundongos , Camundongos Knockout , Transporte Proteico , Proteólise , Interferência de RNA , Transfecção , Vasodilatação , Proteínas de Transporte Vesicular/metabolismo , alfa-Galactosidase/genética , alfa-Galactosidase/metabolismo , Proteínas rab5 de Ligação ao GTP/genética , Proteínas rab5 de Ligação ao GTP/metabolismo
6.
Korean J Physiol Pharmacol ; 19(1): 35-42, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25605995

RESUMO

In cardiovascular disorders, understanding of endothelial cell (EC) function is essential to elucidate the disease mechanism. Although the mouse model has many advantages for in vivo and in vitro research, efficient procedures for the isolation and propagation of primary mouse EC have been problematic. We describe a high yield process for isolation and in vitro culture of primary EC from mouse arteries (aorta, braches of superior mesenteric artery, and cerebral arteries from the circle of Willis). Mouse arteries were carefully dissected without damage under a light microscope, and small pieces of the vessels were transferred on/in a Matrigel matrix enriched with endothelial growth supplement. Primary cells that proliferated in Matrigel were propagated in advanced DMEM with fetal calf serum or platelet-derived serum, EC growth supplement, and heparin. To improve the purity of the cell culture, we applied shearing stress and anti-fibroblast antibody. EC were characterized by a monolayer cobble stone appearance, positive staining with acetylated low density lipoprotein labeled with 1,1'-dioctadecyl-3,3,3',3'-tetramethyl-indocarbocyanine perchlorate, RT-PCR using primers for von-Willebrand factor, and determination of the protein level endothelial nitric oxide synthase. Our simple, efficient method would facilitate in vitro functional investigations of EC from mouse vessels.

7.
Gastroenterology ; 145(1): 232-241, 2013 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-23542070

RESUMO

BACKGROUND & AIMS: The cyclic adenosine monophosphate (cAMP) and Ca(2+) signaling pathways synergize to regulate many physiological functions. However, little is known about the mechanisms by which these pathways interact. We investigated the synergy between these signaling pathways in mouse pancreatic and salivary gland ducts. METHODS: We created mice with disruptions in genes encoding the solute carrier family 26, member 6 (Slc26a6(-/-) mice) and inositol 1,4,5-triphosphate (InsP3) receptor-binding protein released with InsP3 (Irbit(-/-)) mice. We investigated fluid secretion by sealed pancreatic ducts and the function of Slc26a6 and the cystic fibrosis transmembrane conductance regulator (CFTR) in HeLa cells and in ducts isolated from mouse pancreatic and salivary glands. Slc26a6 activity was assayed by measuring intracellular pH, and CFTR activity was assayed by measuring Cl(-) current. Protein interactions were determined by immunoprecipitation analyses. RESULTS: Irbit mediated the synergistic activation of CFTR and Slc26a6 by Ca(2+) and cAMP. In resting cells, Irbit was sequestered by InsP3 receptors (IP3Rs) in the endoplasmic reticulum. Stimulation of Gs-coupled receptors led to phosphorylation of IP3Rs, which increased their affinity for InsP3 and reduced their affinity for Irbit. Subsequent weak stimulation of Gq-coupled receptors, which led to production of low levels of IP3, caused dissociation of Irbit from IP3Rs and allowed translocation of Irbit to CFTR and Slc26a6 in the plasma membrane. These processes stimulated epithelial secretion of electrolytes and fluid. These pathways were not observed in pancreatic and salivary glands from Irbit(-/-) or Slc26a6(-/-) mice, or in salivary gland ducts expressing mutant forms of IP3Rs that could not undergo protein kinase A-mediated phosphorylation. CONCLUSIONS: Irbit promotes synergy between the Ca(2+) and cAMP signaling pathways in cultured cells and in pancreatic and salivary ducts from mice. Defects in this pathway could be involved in cystic fibrosis, pancreatitis, or Sjögren syndrome.


Assuntos
Adenosil-Homocisteinase/fisiologia , Cálcio/metabolismo , AMP Cíclico/fisiologia , Transdução de Sinais/fisiologia , Animais , Antiporters/metabolismo , Transporte Biológico , Proteínas Quinases Dependentes de AMP Cíclico/fisiologia , Regulador de Condutância Transmembrana em Fibrose Cística/fisiologia , Epitélio/metabolismo , Inositol 1,4,5-Trifosfato/biossíntese , Receptores de Inositol 1,4,5-Trifosfato/fisiologia , Camundongos , Ductos Pancreáticos/metabolismo , Fosforilação , Ductos Salivares/metabolismo , Transportadores de Sulfato
8.
Korean J Physiol Pharmacol ; 17(3): 181-7, 2013 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-23776393

RESUMO

Reactive oxygen species (ROS) are generated in various cells, including vascular smooth muscle and endothelial cells, and regulate ion channel functions. KCa3.1 plays an important role in endothelial functions. However, the effects of superoxide and hydrogen peroxide radicals on the expression of this ion channel in the endothelium remain unclear. In this study, we examined the effects of ROS donors on KCa3.1 expression and the K(+) current in primary cultured human umbilical vein endothelial cells (HUVECs). The hydrogen peroxide donor, tert-butyl hydroperoxide (TBHP), upregulated KCa3.1 expression, while the superoxide donors, xanthine/xanthine oxidase mixture (X/XO) and lysopho-sphatidylcholine (LPC), downregulated its expression, in a concentration-dependent manner. These ROS donor effects were prevented by antioxidants or superoxide dismustase. Phosphorylated extracellular signal-regulated kinase (pERK) was upregulated by TBHP and downregulated by X/XO. In addition, repressor element-1-silencing transcription factor (REST) was downregulated by TBHP, and upregulated by X/XO. Furthermore, KCa3.1 current, which was activated by clamping cells with 1 µM Ca(2+) and applying the KCa3.1 activator 1-ethyl-2-benzimidazolinone, was further augmented by TBHP, and inhibited by X/XO. These effects were prevented by antioxidants. The results suggest that hydrogen peroxide increases KCa3.1 expression by upregulating pERK and downregulating REST, and augments the K(+) current. On the other hand, superoxide reduces KCa3.1 expression by downregulating pERK and upregulating REST, and inhibits the K(+) current. ROS thereby play a key role in both physiological and pathological processes in endothelial cells by regulating KCa3.1 and endothelial function.

9.
Pharmacol Res ; 66(1): 51-9, 2012 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-22414869

RESUMO

Modafinil has been used as a psychostimulant for the treatment of narcolepsy. However, its primary mechanism of action remains elusive. Therefore, we examined the effects of modafinil on K(Ca)3.1 channels and vascular smooth muscle contraction. K(Ca)3.1 currents and channel activity were measured using a voltage-clamp technique and inside-out patches in mouse embryonic fibroblast cell line, NIH-3T3 fibroblasts. Intracellular adenosine 3',5'-cyclic monophosphate (cAMP) concentration was measured, and the phosphorylation of K(Ca)3.1 channel protein was examined using western blotting in NIH-3T3 fibroblasts and/or primary cultured mouse aortic smooth muscle cells (SMCs). Muscle contractions were recorded from mouse aorta and rat pulmonary artery by using a myograph developed in-house. Modafinil was found to inhibit K(Ca)3.1 currents in a concentration-dependent manner, and the half-maximal inhibition (IC(50)) of modafinil for the current inhibition was 6.8 ± 0.7 nM. The protein kinase A (PKA) activator forskolin also inhibited K(Ca)3.1 currents. The inhibitory effects of modafinil and forskolin on K(Ca)3.1 currents were blocked by the PKA inhibitors PKI(14-22) or H-89. In addition, modafinil relaxed blood vessels (mouse aorta and rat pulmonary artery) in a concentration-dependent manner. Modafinil increased cAMP concentrations in NIH-3T3 fibroblasts or primary cultured mouse aortic SMCs and phosphorylated K(Ca)3.1 channel protein in NIH-3T3 fibroblasts. However, open probability and single-channel current amplitudes of K(Ca)3.1 channels were not changed by modafinil. From these results, we conclude that modafinil inhibits K(Ca)3.1 channels and vascular smooth muscle contraction by cAMP-dependent phosphorylation, suggesting that modafinil can be used as a cAMP-dependent K(Ca)3.1 channel blocker and vasodilator.


Assuntos
Compostos Benzidrílicos/farmacologia , AMP Cíclico/metabolismo , Canais de Potássio Ativados por Cálcio de Condutância Intermediária/antagonistas & inibidores , Músculo Liso Vascular/efeitos dos fármacos , Miócitos de Músculo Liso/efeitos dos fármacos , Bloqueadores dos Canais de Potássio/farmacologia , Sistemas do Segundo Mensageiro/efeitos dos fármacos , Vasoconstrição/efeitos dos fármacos , Vasodilatadores/farmacologia , Animais , Proteínas de Transporte/farmacologia , Colforsina/farmacologia , Proteínas Quinases Dependentes de AMP Cíclico/antagonistas & inibidores , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Relação Dose-Resposta a Droga , Ativação Enzimática , Ativadores de Enzimas/farmacologia , Humanos , Iloprosta/farmacologia , Canais de Potássio Ativados por Cálcio de Condutância Intermediária/metabolismo , Isoquinolinas/farmacologia , Masculino , Potenciais da Membrana , Camundongos , Modafinila , Músculo Liso Vascular/metabolismo , Miócitos de Músculo Liso/metabolismo , Células NIH 3T3 , Técnicas de Patch-Clamp , Fragmentos de Peptídeos/farmacologia , Fosforilação , Inibidores de Proteínas Quinases/farmacologia , Ratos , Sulfonamidas/farmacologia , Fatores de Tempo , Regulação para Cima
10.
Biomed Pharmacother ; 144: 112372, 2021 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-34794237

RESUMO

Small- and intermediate-conductance Ca2+-activated K+ channels, KCa2.3 and KCa3.1, are involved in cellular signaling processes associated with inflammation and fibrosis. KCa2.3 and KCa3.1 are upregulated by proinflammatory cytokines and profibrotic growth factors. Cyclic AMP, which downregulates KCa2.3 and KCa3.1, is elevated by modafinil in cells; accordingly, we investigated whether modafinil exerts anti-inflammatory and anti-fibrotic responses via KCa2.3- and KCa3.1-mediated pathways in high-fat diet (HFD)- or thioacetamide-induced liver disease models in mice. Modafinil was administered orally in the form of a racemate, (R)-isomer, or (S)-isomer. We also determined whether the treatment targeted the profibrotic activity of hepatic stellate cells using immortalized human hepatic stellate cells (LX-2 cells). Modafinil improved HFD- or thioacetamide-induced changes compared to the control, leading to a reduced inflammatory response, collagen deposition, and α-smooth muscle actin expression both in vivo and in vitro. However, modafinil did not relieve HFD-induced steatosis. There were no significant differences in the effects of the (R)- and (S)-isomers of modafinil. KCa2.3 and KCa3.1 were upregulated and catalase was downregulated in liver tissues from thioacetamide- or HFD-induced liver disease models or in TGF-ß-treated LX-2 cells. TGF-ß-induced upregulation of KCa2.3, KCa3.1, collagen, and α-smooth muscle actin and downregulation of catalase were reversed by modafinil, polyethylene glycol catalase, N-acetylcysteine, siRNA against KCa2.3 or KCa3.1, and Epac inhibitors. Our investigation revealed that modafinil attenuated inflammatory and fibrotic progression via KCa2.3- and KCa3.1-mediated pathways in nonalcoholic hepatitis, suggesting that inhibiting KCa2.3- and KCa3.1-mediated signaling may serve as a novel therapeutic approach for inflammatory and fibrotic liver diseases.


Assuntos
Anti-Inflamatórios não Esteroides/uso terapêutico , Cirrose Hepática/tratamento farmacológico , Modafinila/uso terapêutico , Hepatopatia Gordurosa não Alcoólica/tratamento farmacológico , Actinas/biossíntese , Animais , Anti-Inflamatórios não Esteroides/farmacologia , Linhagem Celular , Colágeno/metabolismo , Dieta Hiperlipídica , Fígado Gorduroso/tratamento farmacológico , Células Estreladas do Fígado , Humanos , Canais de Potássio Ativados por Cálcio de Condutância Intermediária/efeitos dos fármacos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Canais de Potássio Ativados por Cálcio de Condutância Baixa/efeitos dos fármacos , Estereoisomerismo , Tioacetamida/toxicidade
11.
Cell Physiol Biochem ; 25(2-3): 233-40, 2010.
Artigo em Inglês | MEDLINE | ID: mdl-20110684

RESUMO

We examined the mechanism through which lysophosphatidylcholine (LPC) induces endothelial nitric oxide (eNOS) downregulation. Human umbilical vein endothelial cells (HUVECs) were treated with LPC (50-150 microM) for 0.5-2 h or the reactive oxygen species (ROS) donors, xanthine/xanthine oxidase (X/XO), 1,4-hydroquinone (HQ) or tert-butylhydroperoxide (TBHP) for 2 h. Protein levels of eNOS, superoxide dismutase1 (SOD1), catalase, and phospho-extracellular signal regulated kinase 1/2 (pERK 1/2) were assessed using immunoblotting. LPC treatment reduced SOD1 levels but increased catalase levels. The superoxide donors X/XO and HQ showed similar effects. The hydroperoxide donor TBHP increased SOD1 levels but did not change catalase levels. LPC concentration- and time-dependently decreased eNOS levels, but this effect was blocked by antioxidants and SOD and potentiated by the SOD1 inhibitor, ammonium tetrathiomolybdate. LPC and X/XO inhibited ERK1/2 phosphorylation, whereas TBHP stimulated phosphorylation. Taken together, these data indicate that LPC induces superoxide overload in HUVECs via SOD1 inhibition and downregulates phospho-ERK1/2 and eNOS levels.


Assuntos
Células Endoteliais/metabolismo , Lisofosfatidilcolinas/farmacologia , Óxido Nítrico Sintase Tipo III/metabolismo , Superóxidos/metabolismo , Antioxidantes/farmacologia , Catalase/metabolismo , Regulação para Baixo , Células Endoteliais/efeitos dos fármacos , Inibidores Enzimáticos/farmacologia , Humanos , Hidroquinonas/farmacologia , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Molibdênio/farmacologia , Fosforilação , Superóxido Dismutase/metabolismo , terc-Butil Hidroperóxido/farmacologia
12.
Oxid Med Cell Longev ; 2019: 5820839, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31871552

RESUMO

Altered redox state modulates the expression levels of endothelial KCa2.3 and KCa3.1 (KCas) in normal pregnancy (NP) and preeclampsia (PE), thereby regulating vascular contractility. The mechanisms underlying KCas endocytosis and transportation remain unknown. We investigated the regulation of KCas expression in plasma membrane (PM) during NP and PE. Cultured human uterine artery endothelial cells were incubated in serum from normal nonpregnant women and women with NP or PE, or in oxidized LDL-, or lysophosphatidylcholine- (LPC-) containing a medium for 24 hours. NP serum elevated PM levels of KCas and reduced caveolin-1 and clathrin levels. PE serum, oxidized LDL, or LPC reduced PM levels of KCas and elevated caveolin-1, clathrin, Rab5c, and early endosome antigen-1 (EEA1) levels. Reduced KCas levels by PE serum or LPC were reversed by inhibition of caveolin-1, clathrin, or EEA1. Catalase and glutathione peroxidase 1 (GPX1) knockdown elevated PM-localized KCas levels and reduced caveolin-1 and clathrin levels. Elevated KCa2.3 levels upon catalase and GPX1 knockdown were reversed by PEG-catalase treatment. An H2O2 donor reduced clathrin and Rab5c. In contrast, elevated clathrin, caveolin-1, or colocalization of caveolin-1 with KCa3.1 by PE serum or LPC was reversed by NADPH oxidase inhibitors or antioxidants. A superoxide donor xanthine+xanthine oxidase elevated caveolin-1 or Rab5c levels. We concluded that KCas are endocytosed in a caveola- or a clathrin-dependent manner and transported in a Rab5c- and EEA1-dependent manner during pregnancy. The endocytosis and transportation processes may slow down via H2O2-mediated pathways in NP and may be accelerated via superoxide-mediated pathways in PE.


Assuntos
Células Endoteliais/metabolismo , Canais de Potássio Ativados por Cálcio de Condutância Intermediária/metabolismo , Pré-Eclâmpsia/metabolismo , Canais de Potássio Ativados por Cálcio de Condutância Baixa/metabolismo , Animais , Biotinilação , Catalase/genética , Catalase/metabolismo , Eletrofisiologia , Feminino , Glutationa Peroxidase/genética , Glutationa Peroxidase/metabolismo , Humanos , Immunoblotting , Imunoprecipitação , Camundongos Endogâmicos C57BL , Camundongos Knockout , Gravidez , Glutationa Peroxidase GPX1
13.
Antioxid Redox Signal ; 30(4): 505-519, 2019 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-29334762

RESUMO

AIMS: Altered redox state has been related to the development of normal pregnancy (NP) and preeclampsia (PE). Endothelial KCa2.3 and KCa3.1 (KCas) play an important role in vasodilation, and KCas levels are affected by oxidative stress. We investigated the mechanisms of oxidative stress-mediated KCas expression modulation during NP and PE. RESULTS: Human uterine microvascular endothelial cells were incubated in serum from normal nonpregnant women (n = 13) and women with NP (n = 24) or PE (n = 15), or in vascular endothelial growth factor (VEGF), oxidized low-density lipoprotein (ox-LDL), progesterone, or estradiol-17ß (E2)-containing medium for 24 h. NP serum elevated H2O2 levels via reducing catalase and glutathione peroxidase 1 levels, thereby enhancing KCas levels via a H2O2/fyn/extracellular signal-regulated kinase (ERK)-mediated pathway. VEGF enhanced H2O2 and KCas levels and KCa3.1 currents. KCas were upregulated and KCas activation-induced endothelium-dependent relaxation (EDR) was augmented in vessels from pregnant mice and rats. Whereas PE serum, ox-LDL, progesterone, or soluble fms-like tyrosine kinase 1 (sFlt-1) elevated superoxide levels via elevating NADPH oxidase 2 (NOX2) and NOX4 levels and reducing superoxide dismutase (SOD) 1 levels, thereby downregulating KCas. sFlt-1 inhibited EDR. PE serum- or progesterone-induced alterations in levels of KCas were reversed by polyethylene glycol-SOD, NOX inhibition, or E2. Innovation and Conclusions: This is the first study of how endothelial KCas levels are modulated during NP and PE. KCas were upregulated by soluble serum factors such as VEGF via H2O2 generation in NP, and were downregulated by serum factors such as progesterone and ox-LDL via superoxide generation in PE, which may contribute to hemodynamic adaptations in NP or to the development of PE.


Assuntos
Células Endoteliais/metabolismo , Pré-Eclâmpsia/metabolismo , Canais de Potássio Ativados por Cálcio de Condutância Baixa/metabolismo , Animais , Células Cultivadas , Feminino , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Oxirredução , Gravidez , Ratos , Ratos Sprague-Dawley
14.
J Mol Cell Cardiol ; 44(1): 151-9, 2008 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-17976639

RESUMO

Inhibition of Na(+),K(+)-ATPase has been implicated in the pathogenesis of hypertension via its effect on smooth muscle reactivity and myocardial contractility. We recently demonstrated that translationally controlled tumor protein (TCTP) interacts with the 3rd cytoplasmic domain of Na(+),K(+)-ATPase alpha(1)-subunit and acts as its cytoplasmic repressor. Therefore, we hypothesized that repression of Na(+),K(+)-ATPase by overexpressed TCTP might underlie the development of hypertension. In the present study, we confirmed that transgenic mice overexpressing TCTP developed systemic arterial hypertension at about 6 weeks after birth. Vascular smooth muscle of TCTP-overexpressing transgenic mice also displayed augmented contractile response to vasoconstrictors and attenuated relaxation response to vasodilators. These responses seem to be caused by reduced Na(+),K(+)-ATPase activity and increased intracellular calcium, suggesting that inhibition of Na(+),K(+)-ATPase by overexpression of TCTP is involved in the pathogenesis of hypertension. This study provides a new link between alteration of sodium pump activity and hypertension in vivo, and suggests that TCTP might be a therapeutic target for the treatment of hypertension.


Assuntos
Biomarcadores Tumorais/genética , Biomarcadores Tumorais/metabolismo , Hipertensão/enzimologia , Hipertensão/fisiopatologia , ATPase Trocadora de Sódio-Potássio/antagonistas & inibidores , Animais , Pressão Sanguínea , Sinalização do Cálcio , Galinhas , Expressão Gênica , Células HeLa , Humanos , Hipertensão/diagnóstico por imagem , Técnicas In Vitro , Espaço Intracelular/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Contração Muscular , Relaxamento Muscular , Músculo Liso Vascular/enzimologia , ATPase Trocadora de Sódio-Potássio/metabolismo , Proteína Tumoral 1 Controlada por Tradução , Ultrassonografia
15.
Eur J Pharmacol ; 582(1-3): 35-41, 2008 Mar 17.
Artigo em Inglês | MEDLINE | ID: mdl-18237728

RESUMO

The effect of the selective inhibitor of Na(+)/Ca(2+) exchanger (NCX), KB-R7943, on large-conductance Ca(2+)-activated K(+) (BK(Ca)) channels was examined in cultured human umbilical vein endothelial cells (HUVECs) and freshly isolated mouse aortic smooth muscle cells (MASMCs). In voltage-clamped cells, KB-R7943 reversibly activated BK(Ca) currents in HUVECs and MASMCs. The EC(50) of KB-R7943 for BK(Ca) current activation in HUVECs was determined to be 6.78+/-0.7 microM. In inside-out and outside-out patches, KB-R7943 markedly increased BK(Ca) channel activity and slightly decreased single channel current amplitudes. In inside-out patches, KB-R7943 shifted the relationship between [Ca(2+)](i) and open probability (P(o)) to the left; the [Ca(2+)](i) required to evoke half-maximal activation changed from 1220+/-68 nM (in the absence of KB-R7943) to 620+/-199 nM (in the presence of 10 microM KB-R7943). In addition, KB-R7943 shifted the relationship between membrane potential and P(o) to the left; the membrane potential to evoke half-maximal activation changed from 76.86+/-1.09 mV (in the absence of KB-R7943) to 49.62+/-2.55 mV (in the presence of 10 microM KB-R7943). In conclusion, KB-R7943 was found to act as a potent BK(Ca) channel activator, which increases the sensitivity of BK(Ca) channels to cytosolic free Ca(2+) and membrane potential, and thereby BK(Ca) channel activity. These results should be considered when KB-R7943 is used as NCX blocker.


Assuntos
Antiarrítmicos/farmacologia , Células Endoteliais/efeitos dos fármacos , Canais de Potássio Ativados por Cálcio de Condutância Alta/agonistas , Músculo Liso Vascular/efeitos dos fármacos , Trocador de Sódio e Cálcio/antagonistas & inibidores , Tioureia/análogos & derivados , Animais , Cálcio/metabolismo , Células Cultivadas , Células Endoteliais/fisiologia , Humanos , Canais de Potássio Ativados por Cálcio de Condutância Alta/fisiologia , Potenciais da Membrana/efeitos dos fármacos , Camundongos , Músculo Liso Vascular/fisiologia , Miócitos de Músculo Liso/efeitos dos fármacos , Miócitos de Músculo Liso/fisiologia , Técnicas de Patch-Clamp , Tioureia/farmacologia , Veias Umbilicais/citologia
16.
Eur J Obstet Gynecol Reprod Biol ; 133(1): 47-52, 2007 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-16949193

RESUMO

OBJECTIVES: Human paraoxonase-1 (PON-1) is thought to play a role in preeclampsia and atherosclerosis, mainly through a reduction in low-density lipoprotein oxidation. Oxidized low-density lipoprotein (LDL) is very important in endothelial dysfunction of preeclampsia. The aim of the present study was to investigate the association between PON1 gene polymorphism and preeclampsia and to determine concentrations of serum lipid in preeclampsia patients. We aimed also to evaluate serum oxidized LDL levels in normal and preeclampsia patients. STUDY DESIGN: We performed the present study in 57 control women and 32 preeclampsia patients. PON-1 genotypes were determined by polymerase chain reaction amplification and restriction fragment length polymorphism. Serum triglyceride, cholesterol, high-density lipoprotein cholesterol, and low-density lipoprotein cholesterol levels were measured. We also measured serum levels of oxidized LDL by ELISA method. RESULTS: There was no significant difference in PON1 genotype frequencies between the control and preeclampsia patients. The levels of serum cholesterol and high-density lipoprotein were significantly lower in preeclampsia patients compared with that of the control women (p=0.05, and p<0.01, respectively). The serum levels of oxidized LDL in preeclampsia patients were significantly higher than those in the control women (p=0.001). CONCLUSIONS: These findings support the importance of the atherogenic lipid profile and oxidized LDL that is enhanced in preeclampsia, and these findings may be significant contributors to endothelial dysfunction.


Assuntos
Arildialquilfosfatase/genética , Lipídeos/sangue , Lipoproteínas LDL/sangue , Polimorfismo Genético , Pré-Eclâmpsia/sangue , Pré-Eclâmpsia/genética , Biomarcadores , Feminino , Genótipo , Humanos , Lipoproteínas LDL/metabolismo , Oxirredução , Gravidez
17.
Aging Cell ; 15(5): 801-10, 2016 10.
Artigo em Inglês | MEDLINE | ID: mdl-27363720

RESUMO

Endothelial oxidative stress develops with aging and reactive oxygen species impair endothelium-dependent relaxation (EDR) by decreasing nitric oxide (NO) availability. Endothelial KCa 3.1, which contributes to EDR, is upregulated by H2 O2 . We investigated whether KCa 3.1 upregulation compensates for diminished EDR to NO during aging-related oxidative stress. Previous studies identified that the levels of ceramide synthase 5 (CerS5), sphingosine, and sphingosine 1-phosphate were increased in aged wild-type and CerS2 mice. In primary mouse aortic endothelial cells (MAECs) from aged wild-type and CerS2 null mice, superoxide dismutase (SOD) was upregulated, and catalase and glutathione peroxidase 1 (GPX1) were downregulated, when compared to MAECs from young and age-matched wild-type mice. Increased H2 O2 levels induced Fyn and extracellular signal-regulated kinases (ERKs) phosphorylation and KCa 3.1 upregulation. Catalase/GPX1 double knockout (catalase(-/-) /GPX1(-/-) ) upregulated KCa 3.1 in MAECs. NO production was decreased in aged wild-type, CerS2 null, and catalase(-/-) /GPX1(-/-) MAECs. However, KCa 3.1 activation-induced, N(G) -nitro-l-arginine-, and indomethacin-resistant EDR was increased without a change in acetylcholine-induced EDR in aortic rings from aged wild-type, CerS2 null, and catalase(-/-) /GPX1(-/-) mice. CerS5 transfection or exogenous application of sphingosine or sphingosine 1-phosphate induced similar changes in levels of the antioxidant enzymes and upregulated KCa 3.1. Our findings suggest that, during aging-related oxidative stress, SOD upregulation and downregulation of catalase and GPX1, which occur upon altering the sphingolipid composition or acyl chain length, generate H2 O2 and thereby upregulate KCa 3.1 expression and function via a H2 O2 /Fyn-mediated pathway. Altogether, enhanced KCa 3.1 activity may compensate for decreased NO signaling during vascular aging.


Assuntos
Envelhecimento/fisiologia , Endotélio Vascular/fisiologia , Canais de Potássio Ativados por Cálcio de Condutância Intermediária/metabolismo , Estresse Oxidativo/efeitos dos fármacos , Regulação para Cima , Vasodilatação , Envelhecimento/efeitos dos fármacos , Animais , Antioxidantes/metabolismo , Aorta/patologia , Células Endoteliais/metabolismo , Células Endoteliais/patologia , Endotélio Vascular/efeitos dos fármacos , Ativação Enzimática/efeitos dos fármacos , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Peróxido de Hidrogênio/metabolismo , Indometacina/farmacologia , Ativação do Canal Iônico/efeitos dos fármacos , Camundongos Knockout , Modelos Biológicos , Nitroarginina/farmacologia , Oxirredutases/deficiência , Oxirredutases/metabolismo , Proteínas Proto-Oncogênicas c-fyn/metabolismo , Esfingolipídeos/metabolismo , Regulação para Cima/efeitos dos fármacos , Vasodilatação/efeitos dos fármacos
18.
Endothelium ; 12(5-6): 263-9, 2005.
Artigo em Inglês | MEDLINE | ID: mdl-16410226

RESUMO

The authors have studied the effect of sphingosine-1-phosphate (S1P) on Ca2+ release from intracellular stores in cultured human umbilical vein endothelial cells (HUVECs). In the presence of extracellular Ca2+, S1P increased intracellular Ca2+ concentration ([Ca2+]i) and this increase was partially inhibited by La3+ (1 microM), indicating that S1P induces Ca2+ influx from extracellular pool and Ca2+ release from intracellular stores. S1P increased [Ca2+]i concentration dependently in Ca2+-free extracellular solution. The Hill coefficient (1.7) and EC50 (420 nM) was obtained from the concentration-response relationship. When caffeine depleted Ca2+ store in the presence of ryanodine, S1P did not induce intracellular Ca2+ release. Furthermore, the Ca2+-induced Ca2+ release inhibitors ruthenium red or dantrolene completely inhibited S1P-induced intracellular Ca2+ release. S1P-induced intracellular Ca2+ release was inhibited by the phospholipase C (PLC) inhibitors neomycin and U73312, or the inositol 1,4,5-triphosphate (IP3)-gated Ca2+ channel blocker aminoethoxybiphenyl borane (2-APB). In contrast, S1P-induced intracellular Ca2+ release was not inhibited by the mitochondrial Ca2+ uptake inhibitor CCCP or the mitochondrial Ca2+ release inhibitor cyclosporin A. These results show that S1P mobilizes Ca2+ from intracellular stores primarily via Ca2+-induced and IP3-induced Ca2+ release and this Ca2+ mobilization is independent of mitochondrial Ca2+ stores.


Assuntos
Sinalização do Cálcio/efeitos dos fármacos , Cálcio/metabolismo , Células Endoteliais/metabolismo , Lisofosfolipídeos/farmacologia , Mitocôndrias/metabolismo , Esfingosina/análogos & derivados , Veias Umbilicais/metabolismo , Células Cultivadas , Células Endoteliais/citologia , Humanos , Esfingosina/farmacologia , Veias Umbilicais/citologia
19.
Mol Cells ; 20(3): 435-41, 2005 Dec 31.
Artigo em Inglês | MEDLINE | ID: mdl-16404161

RESUMO

Classical transient receptor potential channels (TRPCs) are thought to be candidates for the nonselective cation channels (NSCCs) involved in pacemaker activity and its neuromodulation in murine stomach smooth muscle. We aimed to determine the role of TRPC4 in the formation of NSCCs and in the generation of slow waves. At a holding potential of -60 mV, 50 mM carbachol (CCh) induced INSCC of amplitude [500.8+/-161.8 pA (n=8)] at -60 mV in mouse gastric smooth muscle cells. We investigated the effects of commercially available antibodies to TRPC4 on recombinant TRPC4 expressed in HEK cells and CCh-induced NSCCs in gastric smooth muscle cells. TRPC4 currents in HEK cells were reduced from 1525.6+/-414.4 pA (n=8) to 146.4+/-83.3 pA (n=10) by anti-TRPC4 antibody and INSCC amplitudes were reduced from 230.9+/-36.3 pA (n=15) to 49.8+/-11.8 pA (n=9). Furthermore, INSCC in the gastric smooth muscle cells of TRPC4 knockout mice was only 34.4+/-10.4 pA (n=8) at -60 mV. However, slow waves were still present in the knockout mice. Our data suggest that TRPC4 is an essential component of the NSCC activated by muscarinic stimulation in the murine stomach.


Assuntos
Cátions/metabolismo , Mucosa Gástrica/metabolismo , Agonistas Muscarínicos/farmacologia , Miócitos de Músculo Liso/metabolismo , Canais de Cátion TRPC/fisiologia , Animais , Carbacol/farmacologia , Células Cultivadas , Agonistas Colinérgicos/farmacologia , Condutividade Elétrica , Heterozigoto , Homozigoto , Rim/metabolismo , Camundongos , Camundongos Knockout , Miócitos de Músculo Liso/citologia , Receptores Muscarínicos/metabolismo , Canais de Cátion TRPC/genética
20.
Eur J Obstet Gynecol Reprod Biol ; 119(1): 42-6, 2005 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-15734083

RESUMO

OBJECTIVE: The purpose of this study was to determine whether genetic variability in oxidative stress-related enzymes contributes to individual preeclampsia susceptibility differences. STUDY DESIGN: Polymorphisms in the cytochrome P450 (CYP)1A1 (MspI), CYP1A1(Ile/Val), glutathione S-transferase (GST)M1, GSTT1, myeloperoxidase (MPO), and manganese superoxide dismutase (MnSOD) genes were evaluated by polymerase chain reaction (PCR) or PCR-restriction fragment length polymorphism (RFLP) in 214 healthy controls with an uncomplicated obstetric history, and in 121 preeclampsia patients. Chi2 analyses were used to statistically evaluate differences. RESULTS: No significant differences in the CYP1A1(MspI) or CYP1A1(Ile/Val) genotypes were observed between the healthy controls and the preeclampsia patients (chi2 = 1.43, P = 0.49 versus chi2 = 1.54, P = 0.46). The GSTM1 homozygous null type and GSTT1 homozygous null type were no differences in the patients and controls (chi2 = 0.01, P = 0.92 versus chi(2) = 0.31, P = 0.57), and no significant differences in the polymorphisms of the MPO and MnSOD genotypes were found between the patients and controls (chi2 = 2.00, P = 0.37 versus chi2 = 0.07, P = 0.96). CONCLUSION: Polymorphisms in the oxidative stress-related genes (CYP1A1, GSTM1, GSTT1, MPO, MnSOD) do not seem to be risk factors for preeclampsia.


Assuntos
Citocromo P-450 CYP1A1/genética , Glutationa Transferase/genética , Peroxidase/genética , Pré-Eclâmpsia/genética , Superóxido Dismutase/genética , Adulto , Feminino , Predisposição Genética para Doença , Humanos , Estresse Oxidativo/genética , Polimorfismo Genético , Gravidez , Fatores de Risco
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA