Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
J Cell Biochem ; 120(6): 9291-9299, 2019 06.
Artigo em Inglês | MEDLINE | ID: mdl-30548681

RESUMO

Oxidized low-density lipoprotein (ox-LDL)-induced endothelial dysfunction is recognized as a driving force in the development of atherosclerosis (AS). Paeoniflorin (Pae), a typical traditional herbal medicine, possesses anti-inflammatory, antioxidative, antihyperglycaemic, and antiapoptotic properties. Our study aimed to investigate the effects of Pae on ox-LDL-induced injury of the human umbilical vein endothelial cells (HUVECs) and to explore its molecular mechanism. We found that ox-LDL stimulation inhibited cell viability, activated autophagy, and induced apoptosis and adhesion molecule expression in HUVECs. Pae rescued ox-LDL-induced viability reduction and enhanced the ox-LDL-induced autophagy activation in HUVECs. Pae inhibited ox-LDL-induced apoptosis and adhesion molecule expression by autophagy enhancement in HUVECs. In addition, inhibition of SIRT1 by EX-527 abolished the promoting effect of Pae on autophagy and restored the inhibitory effect of Pae on apoptosis and adhesion molecule expression in the presence of ox-LDL. In conclusion, Pae attenuated ox-LDL-induced apoptosis and adhesion molecule expression by autophagy enhancement via upregulation of SIRT1 in HUVECs, shedding light on the mechanism underlying the protective effect of Pae on ox-LDL-induced injury of HUVECs.


Assuntos
Vasos Sanguíneos/efeitos dos fármacos , Glucosídeos/farmacologia , Lipoproteínas LDL/genética , Monoterpenos/farmacologia , Sirtuína 1/genética , Antioxidantes/farmacologia , Apoptose/efeitos dos fármacos , Autofagia/efeitos dos fármacos , Vasos Sanguíneos/lesões , Carbazóis/farmacologia , Moléculas de Adesão Celular/genética , Sobrevivência Celular/efeitos dos fármacos , Regulação da Expressão Gênica/efeitos dos fármacos , Células Endoteliais da Veia Umbilical Humana , Humanos , Lipoproteínas LDL/antagonistas & inibidores , Estresse Oxidativo/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos
2.
Int Immunopharmacol ; 65: 373-381, 2018 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-30380512

RESUMO

Osthole, a naturally-derived coumarin, has been shown to exhibit pharmacological activities including anti-inflammatory, anti-oxidative and cardiovascular protective effects. However, the effect of osthole on oxidized low-density lipoprotein (ox-LDL)-induced endothelial injury and its underlying mechanism remain unknown. We found that osthole did not affect viability of human umbilical vein endothelial cells (HUVECs) but alleviated ox-LDL-induced cytotoxicity in HUVECs. Osthole repressed ox-LDL-induced release of tumor necrosis factor-α (TNF-α), interleukin (IL)-1ß, and IL-6 in HUVECs. Osthole reversed ox-LDL-induced elevation of reactive oxygen species (ROS) production and malondialdehyde (MDA) level, and reduction of superoxide dismutase (SOD) activity in HUVECs. Meanwhile, osthole attenuated ox-LDL-induced increase of mRNA expression and secretion of intercellular adhesion molecule 1 (ICAM-1) and vascular cell adhesion molecule-1 (VCAM-1) in HUVECs. Osthole increased nitric oxide (NO) production and endothelial nitric oxide synthase (eNOS) phosphorylation in ox-LDL-treated HUVECs. Furthermore, osthole inhibited ox-LDL-induced activation of the transforming growth factor-ß1 (TGF-ß1)/Smad pathway and activation of TGF-ß1/Smad pathway by TGF-ß1 attenuated the protective effects of osthole on HUVECs injury. These results suggested that osthole attenuated ox-LDL-induced HUVECs injury by inhibiting the TGF-ß1/Smad pathway, suggesting that osthole might be a promising therapeutic agent for the treatment of atherosclerosis.


Assuntos
Aterosclerose/tratamento farmacológico , Cumarínicos/farmacologia , Endotélio Vascular/fisiologia , Sobrevivência Celular , Células Endoteliais da Veia Umbilical Humana , Humanos , Mediadores da Inflamação/metabolismo , Molécula 1 de Adesão Intercelular/genética , Molécula 1 de Adesão Intercelular/metabolismo , Lipoproteínas LDL/metabolismo , Óxido Nítrico/metabolismo , Estresse Oxidativo , Espécies Reativas de Oxigênio/metabolismo , Transdução de Sinais , Proteínas Smad/metabolismo , Fator de Crescimento Transformador beta1/metabolismo , Molécula 1 de Adesão de Célula Vascular/genética , Molécula 1 de Adesão de Célula Vascular/metabolismo
3.
Biochem Biophys Res Commun ; 503(4): 3219-3224, 2018 09 18.
Artigo em Inglês | MEDLINE | ID: mdl-30146261

RESUMO

Myocardial ischemia/reperfusion (I/R) injury is a complex pathophysiological process related to the occurrence of myocardial infarction (MI). Oxidative stress is known to play a crucial role in the pathogenesis of I/R injury. Platycodin D (PD) is an active natural saponin that possesses strong anti-oxidant activity. The aim of the present study was to investigate the effect of PD on myocardial I/R injury. An in vitro hypoxia/reoxygenation (H/R) model was established in cardiomyocyte H9c2 cells. The results showed that PD improved the cell viability in H/R-stimulated H9c2 cells. The H/R-induced increase in the production of reactive oxygen species (ROS) and malondialdehyde (MDA), and decrease in the activities of superoxide dismutase (SOD) and catalase (CAT) were reversed by PD pretreatment. The histone-associated DNA fragment was increased by H/R stimulation, while decreased after PD treatment. Besides, PD pretreatment reduced the expressions of Bax and cleaved caspase-3, while induced Bcl-2 expression in H/R-induced H9c2 cells. Furthermore, PD was found to induce the activation of Akt/Nrf2/HO-1 pathway. The inhibitor of Akt, LY294002, attenuated the effects of PD on H/R-induced H9c2 cells. These findings indicated that PD exerted its protective effect via inducing the activation of Akt/Nrf2/HO-1 pathway. Our work provided new insights into the potential therapeutic role of PD in myocardial I/R injury.


Assuntos
Antioxidantes/farmacologia , Apoptose/efeitos dos fármacos , Cardiotônicos/farmacologia , Traumatismo por Reperfusão Miocárdica/tratamento farmacológico , Miócitos Cardíacos/efeitos dos fármacos , Estresse Oxidativo/efeitos dos fármacos , Saponinas/farmacologia , Triterpenos/farmacologia , Animais , Antioxidantes/química , Campanulaceae/química , Cardiotônicos/química , Hipóxia Celular/efeitos dos fármacos , Linhagem Celular , Sobrevivência Celular/efeitos dos fármacos , Traumatismo por Reperfusão Miocárdica/metabolismo , Miócitos Cardíacos/citologia , Miócitos Cardíacos/metabolismo , Ratos , Espécies Reativas de Oxigênio/metabolismo , Saponinas/química , Triterpenos/química
4.
Cardiovasc Pathol ; 31: 57-62, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28985493

RESUMO

Oxidized low-density lipoprotein (ox-LDL) has been reported to induce apoptosis of endothelial cells (ECs) and contribute to the progression of atherosclerosis. Kaempferol has been shown to possess antiatherosclerotic effect. The aim of the present study was to evaluate the effect of kaempferol on ox-LDL-induced apoptosis of human umbilical vein endothelial cells (HUVECs) and its possible molecular basis. The results showed that kaempferol alleviated ox-LDL-induced apoptosis. Kaempferol increased the ratio of LC3-II/I and beclin-1 level in ox-LDL-induced HUVECs. Moreover, the expression of p-Akt and p-mTOR was down-regulated after treatment with kaempferol in ox-LDL-treated HUVECs, which is similar to the effect of PI3K inhibitor (LY294002) or mTOR inhibitor [rapamycin (RAP)]. Besides, autophagy induced by kaempferol was enhanced by LY294002 or RAP, while kaempferol-induced autophagy was attenuated with insulin treatment, the activator of PI3K/Akt/mTOR pathway. Furthermore, insulin also abated the effect of kaempferol on cell viability and apoptosis in ox-LDL-induced HUVECs. The results indicated that kaempferol alleviated ox-LDL-induced cell apoptosis by up-regulation of autophagy via inhibiting PI3K/Akt/mTOR pathway in human ECs.


Assuntos
Apoptose/efeitos dos fármacos , Autofagia/efeitos dos fármacos , Células Endoteliais/efeitos dos fármacos , Quempferóis/farmacologia , Transdução de Sinais/efeitos dos fármacos , Aterosclerose/metabolismo , Células Cultivadas , Células Endoteliais/metabolismo , Células Endoteliais da Veia Umbilical Humana , Humanos , Lipoproteínas LDL/toxicidade , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Serina-Treonina Quinases TOR/metabolismo , Regulação para Cima
5.
Cardiovasc Ther ; 35(6)2017 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-28665537

RESUMO

INTRODUCTION: MiR-195 has been implicated in the development of abdominal aortic aneurysms (AAA). However, the underlying mechanisms have not been fully addressed. AIM: To explore the roles of miR-195 in regulating the proliferation and apoptosis of vascular smooth muscle cells (VSMCs), as well as its molecular basis in vitro. METHODS: qRT-PCR was used to determine the expression levels of miR-195 and Smad3 in aortic media specimens or VSMCs. Western blot was performed to detect the protein levels of Smad3, osteopontin (OPN), and collagen III in aortic media specimens and VSMCs. Luciferase reporter assay was applied to confirm the target of miR-195 in VSMCs. Proliferation and apoptosis of VSMCs were measured by MTT and flow cytometry, respectively. RESULTS: In comparison with the normal controls, the levels of miR-195, OPN, and collagen III were significantly increased in AAA tissue. Smad3 was validated to be a direct target of miR-195. miR-195 inhibited proliferation and induced apoptosis of VSMCs, which was abated by Smad3 overexpression. Expression of OPN and collagen III was improved in VSMCs after transfection with miR-195 mimics, while up-regulation of Smad3 reversed this effect. CONCLUSION: MiR-195 promotes media remodeling by targeting Smad3 in AAA progression. This study suggests that miR-195 contributes to the pathogenesis of AAA and reveals a new targeted therapy strategy for AAA patients.


Assuntos
Aneurisma da Aorta Abdominal/genética , MicroRNAs/genética , MicroRNAs/fisiologia , Proteína Smad3/genética , Apoptose , Caspase 3/biossíntese , Linhagem Celular , Proliferação de Células , Colágeno Tipo III/biossíntese , Marcação de Genes , Humanos , Técnicas In Vitro , Miócitos de Músculo Liso , Osteopontina/biossíntese , Regulação para Cima/genética
6.
J Cancer Res Ther ; 12(Supplement): 60-63, 2016 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-27721256

RESUMO

OBJECTIVES: The objective of this study was to investigate the expression pattern, functions, and possible mechanisms of RhoE/Rnd3, a novel member of the Rho GTPases family, in human esophageal squamous cell carcinoma (ESCC) cells by using molecular and cell-based experiments. MATERIALS AND METHODS: Quantitative polymerase chain reaction and Western blotting were carried out to determine the mRNA and protein expression of RhoE in ESCC cell lines, respectively. Both 3-(4,5-dimethylthiazol-2-yl)-2,5diphenyltetrazoliumbromide (MTT) and flow cytometry were applied to evaluate the effects of RhoE overexpression on ESCC cell growth and apoptosis. Furthermore, Western blotting was used to test the expression of epidermal growth factor receptor (EGFR) and phosphorylated-extracellular signal-regulated kinase (p-ERK) in ESCC cells after RhoE was forced and expressed. RESULTS: RhoE was downregulated in human ESCC tissues. Overexpression of RhoE inhibited cell growth as assessed by MTT assay and induced apoptosis. Importantly, we proved that RhoE could negatively regulate the protein expression of EGFR and p-ERK, suggesting that RhoE might inhibit ESCC progression through the EGFR/ERK pathway. CONCLUSION: Our data supported that RhoE could inhibit cell proliferation and promote apoptosis. Moreover, these tumor-suppressing effects might be acted through the negative regulation of EGFR/ERK signaling.


Assuntos
Carcinoma de Células Escamosas/genética , Carcinoma de Células Escamosas/metabolismo , Receptores ErbB/metabolismo , Neoplasias Esofágicas/genética , Neoplasias Esofágicas/metabolismo , Proteínas rho de Ligação ao GTP/genética , Proteínas rho de Ligação ao GTP/metabolismo , Apoptose/genética , Linhagem Celular Tumoral , Proliferação de Células , Regulação para Baixo , Carcinoma de Células Escamosas do Esôfago , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Regulação Neoplásica da Expressão Gênica , Humanos , RNA Mensageiro/genética , Transdução de Sinais
7.
Asian Pac J Trop Med ; 7(10): 772-5, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-25129458

RESUMO

OBJECTIVES: To study the effect of arsenic trioxide (As2O3) combined with ginsenosides Rg3 on inhibiting the NCI-H1299 lung cancer cells and subsistence in nude mice bearing hepatoma. METHODS: MTT method was used to measure the inhibition effect of As2O3 combined Rg3 on NCI-H1299 cells, and the proliferation inhibiting effect was observed via establishing the transplanted tumor model in vitro. A total of 40 tumor-bearing nude mice were randomly divided into normal saline group, As2O3, Rg3 and As2O3+Rg3 group. Transplantation tumor model of lung cancer in nude mice was constructed, followed by injection of certain concentrations of normal saline, As2O3, ginseng saponin Rg3 and As2O3+Rg3 every day. The survival duration and the tumors size of the mice were recorded and the Kaplan-Meier curve was made; microscopic observation of apoptosis of tumor cells in vivo was done using TUNEL staining. RESULTS: After 72 h of injection, inhibition rate of tumor cell in normal saline group, As2O3 group, Rg3 group and As2O3+Rg3 group was (5.66±0.31)%, (65.58±4.75)%, (44.69±3.32)% and (82.67±5.43)%, respectively. Inhibition rate of tumor cell in As2O3 group, Rg3 group and As2O3+Rg3 group was significantly higher than that of normal saline group (P<0.01); inhibition rate of tumor cells of As2O3+Rg3 group was significantly higher than that of the two groups given As2O3 or Rg3 alone (P<0.01). The tumor volume of As2O3 group, Rg3 group and As2O3+Rg3 group shrank to (65.38±3.25)%, (77.68±3.43)% and (42.65±3.55)% of the original, tumor volume of saline group was 1.21 times of the original size (P<0.01); Median survival of saline group, Rg3 group, As2O3 group were significantly shorter than that of As2O3+Rg3 group (P<0.01); co-ordinated intervention ability of As2O3+Rg3 on NCI-H1299 cell was significantly higher than that of As2O3 or Rg3, separately. CONCLUSIONS: As2O3 combined with Rg3 can significantly inhibit proliferation of NCI-H1299 cells in lung cancer, prolong survival of tumor-bearing nude mice, and promote tumor cell apoptosis, and have significant effect on lung cancer treatment.

8.
Med Oncol ; 31(8): 89, 2014 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-24972913

RESUMO

Researches have showed that interleukin family or receptors play a role in many human tumor progressions including esophageal carcinoma. In this study, we examined the expression of interleukin-8 receptor 2 (IL-8R2) and analyze the relationship between it and esophageal carcinoma clinical characteristics. IL-8R2 protein expression was confirmed by immunohistochemistry and immunofluorescence arrays and was analyzed further via Western blot and qRT-PCR analysis in frozen tissues. The correlation between their expression levels and clinical characteristics were evaluated by Mann-Whitney and Kruskal-Wallis test. Via Kaplan-Meier plots and Cox proportional hazard models, overall survival (OS) was analyzed. Compared with normal esophageal tissue, IL-8R2 protein was overexpressed significantly in esophageal cancer (p < 0.05) and was observed both in cytoplasm and nuclear. The lower expression of IL-8R2 protein was observed with higher p staging of esophageal cancer, and the significant association between them was confirmed (p = 0.000), and in advanced p T stage, the similar result was obtained (p = 0.015); however, compared with lymph node metastasis-negative group, it is no significant difference in positive group (p = 0.152). In a Kaplan-Meier analysis, compared with IL-8R2 low expression, IL-8R2 high expression identified a group of patients with the longest OS. Cox proportional hazard models revealed that IL-8R2 predicted long time to OS. The higher expression of IL-8R2 was found in early esophageal carcinoma, which may indicate that IL-8R2 plays an important role and is better prognostic factor in esophageal cancer development.


Assuntos
Adenocarcinoma/mortalidade , Adenocarcinoma/patologia , Biomarcadores Tumorais/metabolismo , Neoplasias Esofágicas/mortalidade , Neoplasias Esofágicas/patologia , Receptores de Interleucina-8B/metabolismo , Receptores de Interleucina-8/metabolismo , Adenocarcinoma/genética , Adenocarcinoma/metabolismo , Biomarcadores Tumorais/genética , Carcinoma de Células Escamosas/metabolismo , Carcinoma de Células Escamosas/patologia , Estudos de Casos e Controles , Neoplasias Esofágicas/genética , Neoplasias Esofágicas/metabolismo , Esôfago/metabolismo , Feminino , Humanos , Estimativa de Kaplan-Meier , Metástase Linfática/patologia , Masculino , Pessoa de Meia-Idade , Prognóstico , Receptores de Interleucina-8/genética , Receptores de Interleucina-8B/genética , Valores de Referência
9.
Asian Pac J Trop Med ; 7(4): 276-9, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24507675

RESUMO

OBJECTIVE: To investigate the effect of sevoflurane on tissue permeability of lung ischemia-reperfusion injury (LIRI) in rats. METHODS: A total of 45 wistar rats were randomly divided into 3 groups I, II, III. Modified Eppinger method was adopted to establish the rat lung ischemia-reperfusion injury model. Group I served as the control group, group III as ischemia reperfusion group, group III as sevoflurane ischemia-reperfusion group. Blood gas index, lung permeability index (LPI) change, lung tissue pathology change and lung water content were observed and compared between groups of rats at different time points. RESULTS: During ischemia reperfusion, all rats kept balance of the MAP during different time points, SPO2 of group II and III decreased significantly than I group (P<0.05); after reperfusion lung permeability index in Group II and III was higher than the control group significantly (P<0.05), 120 min after reperfusion LPI change and injury of group III was significantly lower than II group (P<0.05); interstitial and alveolar cavity effusion in of group III were lower than that of group II. CONCLUSIONS: Sevoflurane pretreatment can reduce the lung tissue permeability, and LIRI plays a protective role in LIRI.


Assuntos
Pulmão/irrigação sanguínea , Pulmão/efeitos dos fármacos , Éteres Metílicos/farmacologia , Substâncias Protetoras/farmacologia , Traumatismo por Reperfusão/metabolismo , Animais , Pulmão/metabolismo , Masculino , Permeabilidade/efeitos dos fármacos , Distribuição Aleatória , Ratos , Ratos Wistar , Traumatismo por Reperfusão/tratamento farmacológico , Sevoflurano
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...