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










Base de dados
Intervalo de ano de publicação
1.
Biochim Biophys Acta Mol Cell Biol Lipids ; 1869(3): 159453, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-38244675

RESUMO

Vascular smooth muscle cell (VSMC) senescence promotes atherosclerosis via lipid-mediated mitochondrial dysfunction and oxidative stress. However, the mechanisms of mitochondrial dysfunction and VSMC senescence in atherosclerosis have not been established. Here, we investigated the mechanisms whereby signaling pathways regulated by SRT1720 enhance or regulate mitochondrial functions in atherosclerotic VSMCs to suppress atherosclerosis. Initially, we examined the effect of SRT1720 on oleic acid (OA)-induced atherosclerosis. Atherosclerotic VSMCs exhibited elevated expressions of BODIPY and ADRP (adipose differentiation-related protein) and associated intracellular lipid droplet markers. In addition, the expression of collagen I was upregulated by OA, while the expressions of elastin and α-SMA were downregulated. mtDNA copy numbers, an ATP detection assay, transmission electron microscopy (TEM) imaging of mitochondria, mitochondria membrane potentials (assessed using JC-1 probe), and levels of mitochondrial oxidative phosphorylation (OXPHOS) were used to examine the effects of SRT1720 on OA-induced mitochondrial dysfunction. SRT1720 reduced mtDNA damage and accelerated mitochondria repair in VSMCs with OA-induced mitochondria dysfunction. In addition, mitochondrial reactive oxygen species (mtROS) levels were downregulated by SRT1720 in OA-treated VSMCs. Importantly, SRT1720 significantly increased SIRT1 and PGC-1α expression levels, but VSMCs senescence, inflammatory response, and atherosclerosis phenotypes were not recovered by treating cells with EX527 and SR-18292 before SRT1720. Mechanistically, the upregulations of SIRT1 and PGC-1α deacetylation by SRT1720 restored mitochondrial function, and consequently suppressed VSMC senescence and atherosclerosis-associated proteins and phenotypes. Collectively, this study indicates that SRT1720 can attenuate OA-induced atherosclerosis associated with VSMC senescence and mitochondrial dysfunction via SIRT1-mediated deacetylation of the PGC-1α pathway.


Assuntos
Aterosclerose , Compostos Heterocíclicos de 4 ou mais Anéis , Doenças Mitocondriais , Aterosclerose/genética , Aterosclerose/metabolismo , DNA Mitocondrial/metabolismo , Mitocôndrias/metabolismo , Doenças Mitocondriais/metabolismo , Sirtuína 1/genética , Sirtuína 1/metabolismo
2.
Biochem Pharmacol ; 218: 115892, 2023 12.
Artigo em Inglês | MEDLINE | ID: mdl-37890594

RESUMO

Cellular senescence is caused by diverse stimuli and contributes to cardiovascular diseases. Several studies have indicated that PPARγ acts as a key mediator of lipid metabolism and shown that it has a protective effect on vascular biology. Nevertheless, the mechanism responsible for the anti-aging effects of PPARγ has not been fully elucidated in vascular smooth muscle cell (VSMC). Furthermore, although mTOR complex 2 (mTORC2) is known to be involved in cellular senescence and autophagy, relatively few studies have investigated its effects as compared with mTOR complex 1 (mTORC1). Therefore, we focused on mTORC2 function and investigated the relationship between PPARγ and mTORC2, and the anti-aging mechanism in VSMC. We found PPARγ activation dose-dependently mitigated the hydrogen peroxide (H2O2)-induced senescence. Treatment of fisetin induced the translocation of PPARγ from cytosol to nuclear and inhibited VSMC senescence. Moreover, activated PPARγ increased PTEN transcription, leading to inhibition of the mTORC2 signaling pathway. We determined mTORC2 activation contributed to senescence by suppressing the FoxO3a-autophagy signaling pathway, and dual knockdown of mTORC1 and mTORC2 decreased cellular senescence and increased autophagy activation more than respective single knockdown. Finally, fisetin acted as a PPARγ activator and inhibited VSMC senescence through the mTORC2-FoxO3a-autophagy signaling pathway. These results demonstrate PPARγ is associated with cellular senescence and that fisetin has an anti-aging effect via PPARγ activation and mTORC2 inhibition in VSMC. These results demonstrate that the mTORC2 signaling pathway regulates autophagy and cellular senescence, which suggests mTORC2 should be considered a significant target for preventing cellular senescence and age-related diseases.


Assuntos
Músculo Liso Vascular , PPAR gama , Músculo Liso Vascular/metabolismo , PPAR gama/genética , PPAR gama/metabolismo , Peróxido de Hidrogênio/metabolismo , Transdução de Sinais , Senescência Celular , Alvo Mecanístico do Complexo 1 de Rapamicina/metabolismo , Alvo Mecanístico do Complexo 2 de Rapamicina/metabolismo , Autofagia
3.
Arch Gerontol Geriatr ; 108: 104927, 2023 05.
Artigo em Inglês | MEDLINE | ID: mdl-36645971

RESUMO

Reactive oxygen species (ROS) are a key risk factor of cellular senescence and age-related diseases, and protein kinase C (PKC) has been shown to activate NADPH oxidases (NOXs), which generate ROS. Although PKC activation induces oxidative stress, leading to the cellular dysfunction in various cell types, the correlation between PKC and senescence has not been reported in vascular smooth muscle cell (VSMC). Several studies have indicated cellular senescence is accompanied by phosphatase and tensin homolog (PTEN) loss and that an interaction exists between PTEN and PKC. Therefore, we aimed to determine whether PTEN and PKC are associated with VSMC senescence and to investigate the mechanism involved. We found hydrogen peroxide (H2O2) decreased PTEN expression and increased PKCδ phosphorylation. Moreover, H2O2 upregulated the NOX1 subunits, p22phox and p47phox, and induced VSMC senescence via p53-p21 signaling pathway. We identified PKCδ activation contributed to VSMC senescence through activation of NOX1 and ROS production. However, fisetin inhibited cellular senescence induced by the PTEN-PKCδ-NOX1-ROS signaling pathway, and this anti-aging effect was attributed to reduced ROS production caused by suppressing NOX1 activation. These results suggest that the PTEN-PCKδ signaling pathway is directly related to senescence via NOX1 activation and that the downregulation of PKCδ by flavonoids provides a potential means of treating age-associated diseases.


Assuntos
Peróxido de Hidrogênio , Músculo Liso Vascular , Humanos , Espécies Reativas de Oxigênio/metabolismo , Espécies Reativas de Oxigênio/farmacologia , Músculo Liso Vascular/metabolismo , NADPH Oxidase 1/metabolismo , NADPH Oxidase 1/farmacologia , Peróxido de Hidrogênio/metabolismo , Peróxido de Hidrogênio/farmacologia , Células Cultivadas , Senescência Celular , PTEN Fosfo-Hidrolase/metabolismo , PTEN Fosfo-Hidrolase/farmacologia
4.
Mech Ageing Dev ; 206: 111708, 2022 09.
Artigo em Inglês | MEDLINE | ID: mdl-35863470

RESUMO

The senescence of vascular smooth muscle cells (VSMCs) is an important cause of cardiovascular disease such as atherosclerosis and hypertension. These senescence may be triggered by many factors, such as oxidative stress, inflammation, DNA damage, and senescence-associated secretory phenotypes (SASPs). Mitochondrial oxidative stress induces cellular senescence, but the mechanisms by which mitochondrial reactive oxygen species (mtROS) regulates cellular senescence are still largely unknown. Here, we investigated the mechanism responsible for the anti-aging effect of metformin by examining links between VSMC senescence and mtROS in in vitro and in vivo. Metformin was found to increase p-AMPK (Ser485), but to decrease senescence-associated phenotypes and protein levels of senescence markers during ADR-induced VSMC senescence. Importantly, metformin decreased mtROS by inducing the deacetylation of superoxide dismutase 2 (SOD2) by increasing SIRT3 expression. Moreover, AMPK depletion reduced the expression of SIRT3 and increased the expression of acetylated SOD2 despite metformin treatment, suggesting AMPK activation by metformin is required to protect against mitochondrial oxidative stress by SIRT3. This study provides mechanistic evidence that metformin acts as an anti-aging agent and alleviates VSMC senescence by upregulating mitochondrial antioxidant induced p-AMPK (Ser485)-dependent SIRT3 expression, which suggests metformin has therapeutic potential for the treatment of age-associated vascular disease.


Assuntos
Metformina , Sirtuína 3 , Proteínas Quinases Ativadas por AMP/metabolismo , Senescência Celular , Metformina/farmacologia , Oxidantes/farmacologia , Estresse Oxidativo , Fosforilação , Espécies Reativas de Oxigênio/metabolismo , Sirtuína 3/metabolismo
5.
Exp Gerontol ; 156: 111598, 2021 12.
Artigo em Inglês | MEDLINE | ID: mdl-34695518

RESUMO

Cellular senescence is caused by a wide range of intracellular and extracellular stimuli and influences physiological functions, leading to the progression of age-related diseases. Many studies have shown that cellular senescence is related to phosphatase and tension homolog deleted on chromosome ten (PTEN) loss and mammalian target of rapamycin (mTOR) activation. Although it has been reported that mTOR complex 1 (mTORC1) is major anti-aging target in several cell types, the functions and mechanisms of mTOR complex 2 (mTORC2) during aging have not been elucidated in vascular smooth muscle cells (VSMCs). Therefore, the aim of this study was to reveal the relationship between PTEN and mTORC2 during VSMC senescence. We found adriamycin-induced VSMC senescence was accompanied by reduced PTEN protein expression and upregulation of the mTORC2-Akt (Ser 473) pathway and that fisetin treatment reduced VSMC senescence by increasing PTEN and decreasing mTORC2 protein levels. Furthermore, PTEN played a primary role in the anti-aging effect of fisetin, and fisetin-activated PTEN directly regulated the mTORC2-Akt (Ser 473) signaling pathway, and attenuated senescence phenotypes such as senescence-associated ß-galactosidase (SA-ß-gal) and the p53-p21 signaling pathway in VSMCs. In mouse aortas, fisetin delayed aging by regulating the PTEN-mTORC2-Akt (Ser473) signaling pathway. These results suggest PTEN and mTORC2 are associated with cellular senescence in VSMCs and that the mTORC2-Akt (Ser 473) signaling pathway be considered a new target for preventing senescence-related diseases.


Assuntos
Flavonóis/farmacologia , Músculo Liso Vascular , PTEN Fosfo-Hidrolase , Animais , Senescência Celular , Alvo Mecanístico do Complexo 2 de Rapamicina/metabolismo , Camundongos , PTEN Fosfo-Hidrolase/metabolismo , Fosforilação , Proteínas Proto-Oncogênicas c-akt/metabolismo
6.
Exp Gerontol ; 150: 111394, 2021 07 15.
Artigo em Inglês | MEDLINE | ID: mdl-33965557

RESUMO

Skin aging is a complex process and involves extrinsic and intrinsic processes with distinct characteristics. Understanding skin aging requires knowledge of the senescence of human dermal fibroblasts (HDFs) and the biological mechanisms involved in this process. However, the molecular mechanism responsible for the aging of HDFs is still not clear. Therefore, we investigated mechanisms of autophagy, inflammation, and cellular senescence by Western blotting, immunofluorescence, real-time PCR, and senescence-associated ß-galactosidase (SA-ß-gal) staining in senescent HDFs. We found SRT1720 inhibited the inductions of inflammatory cytokines and cellular senescence by deacetylating acetyl-NF-κB levels and enhancing levels of autophagy-associated proteins and SIRT1 in senescent HDFs. However, the NF-κB activator prostratin attenuated signals associated with autophagy, such as those of LC3-II and Beclin-1, but increased inflammatory cytokine levels and cellular senescence. Notably, the expression levels of SIRT1 and autophagy-associated proteins were higher in aged mice administered SRT1720 than in old mice, and SRT1720 also decreased levels of acetyl-NF-κB, inflammatory cytokines, and senescence markers, which was in accord with in vitro results. These findings support that SRT1720 acts as an anti-aging agent and inhibits the inductions of inflammatory cytokines and senescence by regulating the SIRT1/acetyl-NF-κB signaling pathway and activating autophagy in senescent HDFs.


Assuntos
NF-kappa B , Sirtuína 1 , Animais , Autofagia , Células Cultivadas , Senescência Celular , Citocinas , Fibroblastos , Humanos , Camundongos
7.
PLoS One ; 15(9): e0239976, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32997729

RESUMO

Cellular senescence is associated with inflammation and the senescence-associated secretory phenotype (SASP) of secreted proteins. Vascular smooth muscle cell (VSMC) expressing the SASP contributes to chronic vascular inflammation, loss of vascular function, and the developments of age-related diseases. Although VSMC senescence is well recognized, the mechanism of VSMC senescence and inflammation has not been established. In this study, we aimed to determine whether prednisolone (PD) attenuates adriamycin (ADR)-induced VSMC senescence and inflammation through the SIRT1-AMPK signaling pathway. We found that PD inhibited ADR-induced VSMC senescence and inflammation response by decreasing p-NF-κB expression through the SIRT1-AMPK signaling pathway. In addition, Western blotting revealed PD not only increased SIRT1 expression but also increased the phosphorylation of AMPK at Ser485 in ADR-treated VSMC. Furthermore, siRNA-mediated downregulation or pharmacological inhibitions of SIRT1 or AMPK significantly augmented ADR-induced inflammatory response and senescence in VSMC despite PD treatment. In contrast, the overexpression of SIRT1 or constitutively active AMPKα (CA-AMPKα) attenuated cellular senescence and p-NF-κB expression. Taken together, the inhibition of p-NF-κB by PD through the SIRT1 and p-AMPK (Ser485) pathway suppressed VSMC senescence and inflammation. Collectively, our results suggest that anti-aging effects of PD are caused by reduced VSMC senescence and inflammation due to reciprocal regulation of the SIRT1/p-AMPK (Ser485) signaling pathway.


Assuntos
Senescência Celular/efeitos dos fármacos , Doxorrubicina/farmacologia , Prednisolona/farmacologia , Transdução de Sinais/efeitos dos fármacos , Proteínas Quinases Ativadas por AMP/antagonistas & inibidores , Proteínas Quinases Ativadas por AMP/genética , Proteínas Quinases Ativadas por AMP/metabolismo , Animais , Aorta/efeitos dos fármacos , Aorta/metabolismo , Regulação para Baixo/efeitos dos fármacos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Músculo Liso Vascular/citologia , Músculo Liso Vascular/efeitos dos fármacos , Músculo Liso Vascular/metabolismo , NF-kappa B/antagonistas & inibidores , NF-kappa B/metabolismo , Fosforilação/efeitos dos fármacos , Interferência de RNA , RNA Interferente Pequeno/metabolismo , Ratos , Ratos Sprague-Dawley , Sirtuína 1/antagonistas & inibidores , Sirtuína 1/genética , Sirtuína 1/metabolismo , Proteína Supressora de Tumor p53/metabolismo
8.
Mech Ageing Dev ; 190: 111314, 2020 09.
Artigo em Inglês | MEDLINE | ID: mdl-32679123

RESUMO

Calcium (Ca2+) homeostasis is disrupted during aging in several cell types and this disruption leads to autophagy impairment. The mechanisms regarding Ca2+, senescence, and autophagy need to be elucidated. Therefore, we hypothesized that cellular senescence can be improved by regulating Ca2+ level and autophagy activity. We identified that hydrogen peroxide (H2O2)-induced senescence was accompanied by Ca2+ elevation, impairment of autophagic flux and increase of mammalian target of rapamycin (mTOR) phosphorylation in VSMCs. The treatment of nifedipine dose-dependently suppressed H2O2-induced senescence by reducing Ca2+ entry, autophagy impairment and mTOR signaling, and this suppression was found to be related to senescence-associated ß-galactosidase (SA-ß-gal) activity and the expressions of senescence marker protein 30 (SMP30), p53, and p21. Furthermore, H2O2-induced autophagy impairment also accelerated senescence and accumulations of ubiquitinated proteins. AMPK inhibition or transfection with AMPK siRNA showed that the anti-senescence effect of nifedipine involved AMPK activation. These results suggest nifedipine-inducted AMPK activation suppresses VSMC senescence by regulating autophagic flux and Ca2+ levels.


Assuntos
Proteínas Quinases Ativadas por AMP/metabolismo , Autofagia , Sinalização do Cálcio , Senescência Celular , Miócitos de Músculo Liso/metabolismo , Nifedipino/farmacologia , Serina-Treonina Quinases TOR/metabolismo , Animais , Autofagia/efeitos dos fármacos , Autofagia/fisiologia , Cálcio/metabolismo , Bloqueadores dos Canais de Cálcio/farmacologia , Sinalização do Cálcio/efeitos dos fármacos , Sinalização do Cálcio/fisiologia , Proteínas de Ligação ao Cálcio/metabolismo , Hidrolases de Éster Carboxílico/metabolismo , Células Cultivadas , Senescência Celular/efeitos dos fármacos , Senescência Celular/fisiologia , Músculo Liso Vascular , Fosforilação , Ratos
9.
Int J Mol Sci ; 21(11)2020 Jun 06.
Artigo em Inglês | MEDLINE | ID: mdl-32517197

RESUMO

It has been suggested that methylglyoxal (MGO), a glycolytic metabolite, has more detrimental effects on endothelial dysfunction than glucose itself. Recent reports showed that high glucose and MGO induced endoplasmic reticulum (ER) stress and myocyte apoptosis in ischemic heart disease was inhibited by apelin. The goal of the study is to investigate the molecular mechanism by which MGO induces endothelial dysfunction via the regulation of ER stress in endothelial cells, and to examine whether apelin-13, a cytoprotective polypeptide ligand, protects MGO-induced aortic endothelial dysfunction. MGO-induced ER stress and apoptosis were determined by immunoblotting and MTT assay in HUVECs. Aortic endothelial dysfunction was addressed by en face immunostaining and acetylcholine-induced vasodilation analysis with aortic rings from mice treated with MGO in the presence or absence of apelin ex vivo. TUDCA, an inhibitor of ER stress, inhibited MGO-induced apoptosis and reduction of cell viability, suggesting that MGO signaling to endothelial apoptosis is mediated via ER stress, which leads to activation of unfolded protein responses (UPR). In addition, MGO-induced UPR and aortic endothelial dysfunction were significantly diminished by apelin-13. Finally, this study showed that apelin-13 protects MGO-induced UPR and endothelial apoptosis through the AMPK pathway. Apelin-13 reduces MGO-induced UPR and endothelial dysfunction via regulating the AMPK activating pathway, suggesting the therapeutic potential of apelin-13 in diabetic cardiovascular complications.


Assuntos
Endotélio/efeitos dos fármacos , Endotélio/metabolismo , Peptídeos e Proteínas de Sinalização Intercelular/farmacologia , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Aldeído Pirúvico/farmacologia , Resposta a Proteínas não Dobradas/efeitos dos fármacos , Apoptose/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Células Cultivadas , Relação Dose-Resposta a Droga , Estresse do Retículo Endoplasmático/efeitos dos fármacos , Endotélio/fisiopatologia , Células Endoteliais da Veia Umbilical Humana/metabolismo , Humanos , Estresse Oxidativo/efeitos dos fármacos , Espécies Reativas de Oxigênio/metabolismo
10.
FEBS Open Bio ; 10(7): 1316-1325, 2020 07.
Artigo em Inglês | MEDLINE | ID: mdl-32421926

RESUMO

Aging is a major risk factor for hypertension and atherosclerosis, and vascular smooth muscle cell (VSMC) senescence can promote aging-related vascular diseases. Sirtuin-1 (SIRT1) and AMP-activated protein kinase (AMPK) were previously reported to modulate vascular senescence; however, its effects have not been well characterized. To determine the nature of the interaction between SIRT1 and AMPK in VSMC senescence, we investigated the effects of SRT1720 on its downstream targets of SIRT1 and the phosphorylation of AMPKα at Ser485. During Adriamycin-induced VSMC senescence, SRT1720 increased the activity of SIRT1 and AMPKα phosphorylation at Ser485 via the cAMP-protein kinase A (PKA) pathway. Telomere length and telomerase reverse transcriptase expression were increased by SIRT1 activation with SRT1720. Taken together, these data show that activation of the SIRT1/cAMP-PKA/p-AMPKα (Ser485) pathway may be an effective antisenescence mechanism for VSMCs.


Assuntos
Proteínas Quinases Ativadas por AMP/antagonistas & inibidores , Proteínas Quinases Dependentes de AMP Cíclico/antagonistas & inibidores , Compostos Heterocíclicos de 4 ou mais Anéis/farmacologia , Músculo Liso Vascular/efeitos dos fármacos , Serina/antagonistas & inibidores , Sirtuína 1/metabolismo , Proteínas Quinases Ativadas por AMP/metabolismo , Animais , Células Cultivadas , Senescência Celular/efeitos dos fármacos , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Músculo Liso Vascular/metabolismo , Fosforilação/efeitos dos fármacos , Ratos , Serina/metabolismo
11.
Korean J Physiol Pharmacol ; 24(1): 69-79, 2020 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-31908576

RESUMO

Aging is one of the risk factors for the development of cardiovascular diseases. During the progression of cellular senescence, cells enter a state of irreversible growth arrest and display resistance to apoptosis. As a flavonoid, quercetin induces apoptosis in various cells. Accordingly, we investigated the relationship between quercetin-induced apoptosis and the inhibition of cellular senescence, and determined the mechanism of oxidative stress-induced vascular smooth muscle cell (VSMC) senescence. In cultured VSMCs, hydrogen peroxide (H2O2) dose-dependently induced senescence, which was associated with increased numbers of senescence-associated ß-galactosidase-positive cells, decreased expression of SMP30, and activation of p53-p21 and p16 pathways. Along with senescence, expression of the anti-apoptotic protein Bcl-2 was observed to increase and the levels of proteins related to the apoptosis pathway were observed to decrease. Quercetin induced apoptosis through the activation of AMP-activated protein kinase. This action led to the alleviation of oxidative stress-induced VSMC senescence. Furthermore, the inhibition of AMPK activation with compound C and siRNA inhibited apoptosis and aggravated VSMC senescence by reversing p53-p21 and p16 pathways. These results suggest that senescent VSMCs are resistant to apoptosis and quercetin-induced apoptosis attenuated the oxidative stress-induced senescence through activation of AMPK. Therefore, induction of apoptosis by polyphenols such as quercetin may be worthy of attention for its anti-aging effects.

14.
Vascul Pharmacol ; 102: 21-28, 2018 03.
Artigo em Inglês | MEDLINE | ID: mdl-29337033

RESUMO

Gliclazide, a sulfonylurea that is widely used to treat type II-diabetes, specifically blocks KATP channels and recombinant smooth muscle (SUR2B/Kir6.1) KATP channels with high potency. Furthermore, it exerts antioxidant properties and inhibits tumor cell proliferation. In this study, we investigated the inhibitory effect of gliclazide on vascular smooth muscle cell (VSMC) proliferation and tried to identify the underlying signaling pathway. We first investigated the effect of gliclazide-induced AMP-activated protein kinase (AMPK) activation on the proliferation of VSMCs. Gliclazide induced phosphorylation of AMPK in a dose- and time-dependent manner and inhibited VSMC proliferation following stimulation by platelet-derived growth factor (PDGF). However, KATP channel openers and Kir6.1 siRNA prevented gliclazide-mediated inhibition of VSMC proliferation. Gliclazide also increased the levels of Ca2+/calmodulin-dependent protein kinase kinase ß (CaMKKß), an upstream kinase of AMPK. These findings suggested that the effects of KATP channels on AMPK activity were mediated by the regulation of intracellular Ca2+ levels. Oral administration of 2mg/kg gliclazide resulted in the activation of CaMKKß and AMPK in vivo, suggesting that gliclazide suppressed VSMC proliferation via the CaMKKß-AMPK signaling pathway. Taken together, our observations indicated that gliclazide-induced AMPK activation may act to prevent diabetes-associated atherosclerosis.


Assuntos
Proteínas Quinases Ativadas por AMP/metabolismo , Quinase da Proteína Quinase Dependente de Cálcio-Calmodulina/metabolismo , Proliferação de Células/efeitos dos fármacos , Gliclazida/farmacologia , Canais KATP/antagonistas & inibidores , Músculo Liso Vascular/efeitos dos fármacos , Miócitos de Músculo Liso/efeitos dos fármacos , Bloqueadores dos Canais de Potássio/farmacologia , Receptores de Sulfonilureias/antagonistas & inibidores , Animais , Cálcio/metabolismo , Células Cultivadas , Relação Dose-Resposta a Droga , Ativação Enzimática , Canais KATP/genética , Canais KATP/metabolismo , Masculino , Camundongos Endogâmicos C57BL , Músculo Liso Vascular/enzimologia , Músculo Liso Vascular/patologia , Miócitos de Músculo Liso/enzimologia , Miócitos de Músculo Liso/patologia , Fosforilação , Interferência de RNA , Ratos Sprague-Dawley , Transdução de Sinais/efeitos dos fármacos , Receptores de Sulfonilureias/genética , Receptores de Sulfonilureias/metabolismo , Fatores de Tempo , Transfecção
15.
J Med Food ; 21(2): 146-153, 2018 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-29035613

RESUMO

The vascular tone plays an important role in blood pressure and flow. It is influenced by the contraction of vascular smooth muscle cells (VSMCs), which in turn is regulated by the balance between the myosin light chain kinase (MLCK) and the phosphorylated myosin light chain (p-MLC). Quercetin is a common flavonoid which is found in many fruits and red wine. Although quercetin has been widely reported to be involved in cell proliferation, migration, and apoptosis in VSMCs, it has not yet been demonstrated whether quercetin is related to vasocontraction, a function regulated by the AMP-activated protein kinase (AMPK) signaling pathway. Accordingly, the aim of this study is to investigate the molecular mechanism through which the quercetin-activated LKB1-AMPK signaling pathway regulates the contraction of VSMCs. In cultured VSMCs, quercetin activated AMPK in a dose- and time-dependent manner. Quercetin inhibited the phenylephrine (PE)-induced expression of MLCK and p-MLC through the LKB1-AMPK signaling pathway and decreased the mRNA level of MLCK. Adenovirus-AMPK DN α1 and AMPK DN α2-transduced VSMCs displayed higher p-MLC expression. Moreover, quercetin inhibited the PE-mediated contraction in rat aorta. These data suggest that the quercetin-activated LKB1-AMPK signaling pathway regulates VSMC contraction by inhibiting MLCK and p-MLC; hence, it may be a therapeutic intervention for the treatment of cardiovascular disorders such as atherosclerosis and hypertension.


Assuntos
Proteínas Quinases Ativadas por AMP/metabolismo , Músculo Liso Vascular/efeitos dos fármacos , Proteínas Serina-Treonina Quinases/metabolismo , Quercetina/farmacologia , Vasoconstrição/efeitos dos fármacos , Quinases Proteína-Quinases Ativadas por AMP , Proteínas Quinases Ativadas por AMP/genética , Animais , Aorta/efeitos dos fármacos , Aorta/enzimologia , Aorta/fisiologia , Masculino , Músculo Liso Vascular/enzimologia , Músculo Liso Vascular/fisiologia , Miócitos de Músculo Liso/efeitos dos fármacos , Miócitos de Músculo Liso/enzimologia , Miócitos de Músculo Liso/metabolismo , Fosforilação , Proteínas Serina-Treonina Quinases/genética , Ratos , Ratos Sprague-Dawley
16.
Exp Gerontol ; 109: 51-58, 2018 08.
Artigo em Inglês | MEDLINE | ID: mdl-28797827

RESUMO

Cellular senescence is related to aging and extremely stable proliferative arrest with active metabolism. Senescent cells can activate mammalian target of rapamycin (mTOR) pathway, which plays a crucial role in the regulation of cell metabolism, cellular growth, and autophagy in senescence-associated cardiovascular diseases. Therefore, we examined whether mTOR pathway could induce cellular senescence by inhibition of autophagy in vascular smooth muscle cells (VSMCs). We found that adriamycin-induced VSMC senescence is accompanied by increased activity of mTOR, a major controller of cell growth and a negative regulator of autophagy. VSMC senescence induced by activation of mTOR pathway led to reduced levels of signal-associated autophagy proteins, and inhibition of mTOR pathway resulted in a drastic decrease in the number of senescence-associated ß-galactosidase (SA-ß-gal)-stained cells and increased levels of signal-associated autophagy proteins. Autophagic inhibition potentiated adriamycin-induced mTOR pathway activation as well as increase in the number of SA-ß-gal-stained VSMCs. Results of further experiments showed that mTOR pathway inhibition regulates adriamycin-induced expression of senescence markers (p53/p21/p16), which plays an important role in different aspects of cellular aging. Taken together, these results support the idea that intervention to modulate the interaction between mTOR pathway and autophagy could be a potential strategy for longevity.


Assuntos
Autofagia/fisiologia , Senescência Celular/efeitos dos fármacos , Inibidor p16 de Quinase Dependente de Ciclina/análise , Inibidor de Quinase Dependente de Ciclina p21/análise , Doxorrubicina/toxicidade , Músculo Liso Vascular/efeitos dos fármacos , Miócitos de Músculo Liso/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos , Serina-Treonina Quinases TOR/antagonistas & inibidores , Proteína Supressora de Tumor p53/análise , Animais , Músculo Liso Vascular/citologia , Miócitos de Músculo Liso/fisiologia , Ratos , Ratos Sprague-Dawley , Serina-Treonina Quinases TOR/fisiologia
17.
Arch Pharm Res ; 40(8): 943-951, 2017 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-28770536

RESUMO

Minoxidil is widely used for treatment of androgenic alopecia. Commercial products containing minoxidil are usually in solution form. Repeated applications of minoxidil solution can lead to adverse effects such as skin irritation and horniness. The aims of this study were to prepare lecithin-based microparticle in minoxidil solution for enhancement of minoxidil topical delivery and skin protection and evaluate the ability of lecithin on in vitro delivery, in vivo hair growth, and skin trouble improvement compared to commercial minoxidil solution. In in vitro skin permeation study, minoxidil solution containing lecithin microparticle showed higher skin penetration rate and higher retention of drug inside the skin compared to minoxidil solution without lecithin. After topical application of minoxidil solutions with or without lecithin to C57BL/6 mice, minoxidil 5% solution containing lecithin microparticle showed hair re-growth as efficient as commercial product of minoxidil 5% solution. It also significantly improved skin troubles while commercial product presented horny substance and crust formation. Therefore, the lecithin-based microparticle in minoxidil 5% solution has good ability to promote hair growth without adverse effects.


Assuntos
Sistemas de Liberação de Medicamentos , Cabelo/efeitos dos fármacos , Lecitinas/química , Minoxidil/administração & dosagem , Administração Cutânea , Alopecia/tratamento farmacológico , Animais , Portadores de Fármacos/química , Feminino , Cabelo/crescimento & desenvolvimento , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Microesferas , Minoxidil/farmacocinética , Minoxidil/farmacologia , Soluções Farmacêuticas , Ratos , Ratos Sprague-Dawley , Absorção Cutânea , Vasodilatadores/administração & dosagem , Vasodilatadores/farmacocinética , Vasodilatadores/farmacologia
18.
Biochem Biophys Res Commun ; 490(4): 1369-1374, 2017 09 02.
Artigo em Inglês | MEDLINE | ID: mdl-28694192

RESUMO

In healthy condition, vascular smooth muscle cells (VSMCs) are not directly exposed to shear stresses, because they are shielded by endothelial cell (EC) layer that lines blood vessels. After injury to EC layer caused by rupture of atherosclerotic lesions or invasive techniques such as angioplasty, VSMCs are directly exposed to blood flow which modulate molecular signaling and function. In endothelium, exposure to fluid shear stress has been reported to induce AMP-activated protein kinase (AMPK) phosphorylation and nitric oxide (NO) production. However, the influence of laminar shear stress on exposed VSMC is not defined. In this study, we investigated whether laminar shear stress regulates AMPK phosphorylation in VSMC and tried to identify underlying signaling pathway. NO production was increased by shear stress. The expression of NOS isoforms was increased 1 h after exposure to shear stress, and AMPK phosphorylation started to increase after 2 h. AMPK and LKB1, the upstream kinases of AMPK, phosphorylation were decreased by the non-selective NOS inhibitor l-NAME and the selective iNOS inhibitor aminoguanidine despite exposure to shear stress. On the other hand, compound C, a specific AMPK inhibitor, did not affect the expression of NOS isoforms. In addition, PDGF-induced VSMC proliferation was decreased by shear stress and restored by l-NAME. These findings suggest that shear stress upregulated AMPK phosphorylation in VSMC via NOS expression may be a beneficial route to prevent pathogenesis in the vascular system.


Assuntos
Proteínas Quinases Ativadas por AMP/genética , Células Endoteliais/metabolismo , Mecanotransdução Celular , Óxido Nítrico Sintase Tipo II/genética , Óxido Nítrico/biossíntese , Proteínas Serina-Treonina Quinases/genética , Quinases Proteína-Quinases Ativadas por AMP , Proteínas Quinases Ativadas por AMP/antagonistas & inibidores , Proteínas Quinases Ativadas por AMP/metabolismo , Animais , Aorta Torácica/citologia , Aorta Torácica/metabolismo , Proliferação de Células , Células Endoteliais/citologia , Regulação da Expressão Gênica , Guanidinas/farmacologia , Hemorreologia , Masculino , Músculo Liso Vascular/citologia , Músculo Liso Vascular/metabolismo , NG-Nitroarginina Metil Éster/farmacologia , Óxido Nítrico Sintase Tipo II/antagonistas & inibidores , Óxido Nítrico Sintase Tipo II/metabolismo , Fosforilação/efeitos dos fármacos , Cultura Primária de Células , Inibidores de Proteínas Quinases/farmacologia , Proteínas Serina-Treonina Quinases/metabolismo , Ratos , Ratos Sprague-Dawley , Estresse Mecânico
19.
Biochem Biophys Res Commun ; 480(4): 622-628, 2016 Nov 25.
Artigo em Inglês | MEDLINE | ID: mdl-27794481

RESUMO

Previous epidemiological studies have shown that methylglyoxal (MGO) levels are highly regulated in diabetic cardiovascular diseases. We have also previously reported that MGO mediates ER stress and apoptosis in cardiomyocytes. Furthermore, activated protein C (APC) has recently been shown to play a protective role against ER stress, as well as a cardioprotective role against ischemia and reperfusion injury by augmenting the AMP-activated protein kinase (AMPK) signaling pathway. Therefore, we hypothesized that APC protects against MGO-induced cardiomyocyte apoptosis through the inhibition of ER stress. Our results showed that APC inhibited MGO-induced cardiomyocyte apoptosis and ER stress-related gene expression. Additionally, APC inhibited MGO-induced Ca2+ mobilization and the generation of reactive oxygen species. In contrast, inhibitors of AMPK signaling abolished the cytoprotective effects of APC. Collectively, these data depict a pivotal role for AMPK signaling in inhibiting ER stress responses via the activation of APC during MGO-induced cardiomyocyte apoptosis. Thus, APC may be a potential novel therapeutic target for the management of diabetic cardiovascular complications such as diabetic cardiomyopathy.


Assuntos
Proteínas Quinases Ativadas por AMP/metabolismo , Apoptose/fisiologia , Estresse do Retículo Endoplasmático/fisiologia , Miócitos Cardíacos/fisiologia , Proteína C/farmacologia , Aldeído Pirúvico/farmacologia , Apoptose/efeitos dos fármacos , Cardiotônicos/farmacologia , Linhagem Celular , Relação Dose-Resposta a Droga , Estresse do Retículo Endoplasmático/efeitos dos fármacos , Humanos , Miócitos Cardíacos/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/fisiologia
20.
J Pharmacol Sci ; 132(1): 78-85, 2016 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-27665370

RESUMO

The prevalence rate of cardiovascular disease is higher for males than females, and estradiol (E2) induces AMP-activated protein kinase (AMPK) activation, which is known to regulate proliferation of VSMC. We identified the estrogenic properties of nordihydroguaiaretic acid (NDGA, a lignan phytoestrogen) that inhibit VSMC proliferation and explored the underlying mechanisms. Both the phosphorylation and expression of LKB1 were increased by NDGA. In addition, NDGA significantly attenuated angiotensin II (Ang II)-induced VSMC proliferation. To elucidate the estrogenic effects, we confirmed that NDGA increased estrogen receptor α (ERα) expression, similar to treatment with E2 and estriol (E3). Furthermore, tamoxifen and ERα siRNA obstructed the effects of NDGA including ERα expression, AMPK phosphorylation and both LKB1 phosphorylation and expression. VSMC proliferation was restored by tamoxifen and ERα siRNA. LKB1 siRNA also reversed the NDGA-mediated inhibition of VSMC proliferation. The estrogenic activity of NDGA induced LKB1 translocation from nucleus to cytosol, and tamoxifen obstructed LKB1 translocation. The absence of LKB1 completely abolished the increase of ERα expression induced by NDGA. Taken together, the beneficial effects of estrogenic compound (E2 and NDGA) on inhibition of VSMC proliferation are mediated by interaction between LKB1 and ERα, suggesting a potential mechanism for females having less cardiovascular disease.


Assuntos
Estradiol/farmacologia , Receptor alfa de Estrogênio/metabolismo , Estrogênios/farmacologia , Masoprocol/farmacologia , Miócitos de Músculo Liso/efeitos dos fármacos , Proteínas Serina-Treonina Quinases/metabolismo , Quinases Proteína-Quinases Ativadas por AMP , Proteínas Quinases Ativadas por AMP/metabolismo , Angiotensina II/farmacologia , Animais , Aorta Torácica/citologia , Ciclo Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Di-Hidrotestosterona/farmacologia , Estriol/farmacologia , Receptor alfa de Estrogênio/genética , Músculo Liso Vascular/citologia , Miócitos de Músculo Liso/metabolismo , Miócitos de Músculo Liso/fisiologia , Fosforilação/efeitos dos fármacos , Proteínas Serina-Treonina Quinases/genética , RNA Interferente Pequeno/genética , Ratos Sprague-Dawley
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...