Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 46
Filtrar
Mais filtros

Intervalo de ano de publicação
1.
Exp Cell Res ; 408(1): 112857, 2021 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-34600900

RESUMO

Toxic advanced glycation end products (toxic AGEs) derived from glycolaldehyde (AGE3) have been implicated in the development of diabetic vascular complications such as retinopathy characterised by excessive angiogenesis. Different receptor types, such as receptor for AGEs (RAGE), Toll like receptor-4 and scavenger receptors, are expressed in endothelial cells and contribute to AGE-elicited alteration of cell function. In the present study, we examined the involvement of AGE-related receptors on AGE-induced angiogenesis in endothelial cells. The effects of pharmacological inhibitors or receptor neutralizing antibodies on AGE3-induced tube formation were investigated using the in vitro Matrigel tube formation assay in b.End5 cells (mouse endothelial cells). AGE3-induced signalling pathways and receptor expression changes were analysed by Western blot analysis and flow cytometry, respectively. Both FPS-ZM1, a RAGE inhibitor, and fucoidan, a ligand for scavenger receptors, suppressed AGE3-induced tube formation. Cocktails of neutralizing antibodies against the scavenger receptors CD36, CD163 and LOX-1 prevented AGE3-induced tube formation. AGE3 activated mTOR signalling, resulting in facilitation of tube formation. Activation of the AGE-RAGE pathway also led to the upregulation of scavenger receptors. Taken together, our findings suggest that the scavenger receptors CD36, CD163 and LOX-1 in conjunction with the RAGE receptor work together to mediate toxic AGE-induced facilitation of angiogenesis.


Assuntos
Células Endoteliais/efeitos dos fármacos , Produtos Finais de Glicação Avançada/farmacologia , Neovascularização Patológica/metabolismo , Receptores Depuradores/metabolismo , Animais , Células Endoteliais/metabolismo , Produtos Finais de Glicação Avançada/metabolismo , Camundongos , Neovascularização Patológica/tratamento farmacológico , Receptor para Produtos Finais de Glicação Avançada/efeitos dos fármacos , Receptor para Produtos Finais de Glicação Avançada/metabolismo , Receptores Depuradores/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos , Regulação para Cima/efeitos dos fármacos
2.
J Integr Neurosci ; 20(2): 255-264, 2021 Jun 30.
Artigo em Inglês | MEDLINE | ID: mdl-34258924

RESUMO

The abnormal deposition of the extracellular amyloid-ß peptide is the typical pathological hallmark of Alzheimer's disease. Strategies to reduce the amyloid-ß deposition effectively alleviate the neuronal degeneration and cognitive deficits of Alzheimer's disease. Danggui-Shaoyao-San has been considered a useful therapeutic agent known for the treatment of Alzheimer's disease. However, the mechanism of Danggui-Shaoyao-San for the treatment of Alzheimer's disease remains unclear. We investigated Danggui-Shaoyao-San's effect on amyloidosis and neuronal degeneration in an APP/PS1 mouse model. We found Danggui-Shaoyao-San alleviated the cognitive deficits in APP/PS1 mice. Additionally, Danggui-Shaoyao-San ameliorated the neuronal degeneration in these mice. Danggui-Shaoyao-San reduced the amyloidosis and amyloid-ß1-42 deposition in APP/PS1 mouse brain and down-regulated the receptor for advanced glycation end products, and up-regulated the level of low-density lipoprotein receptor-related protein-1. However, the protein expression of the ß-amyloid precursor protein, ß-Secretase and presenilin-1 (PS1) in the amyloid-ß production pathway, and the expression of neprilysin and insulin-degrading enzyme in the amyloid-ß degradation pathway were not altered. Our findings collectively suggest that Danggui-Shaoyao-San could ameliorate the amyloidosis and neuronal degeneration of Alzheimer's disease, which may be associated with its up-regulation lipoprotein receptor-related protein-1 and down-regulation of the receptor for advanced glycation end products.


Assuntos
Doença de Alzheimer/tratamento farmacológico , Amiloidose/tratamento farmacológico , Disfunção Cognitiva/tratamento farmacológico , Medicamentos de Ervas Chinesas/farmacologia , Proteína-1 Relacionada a Receptor de Lipoproteína de Baixa Densidade/efeitos dos fármacos , Receptor para Produtos Finais de Glicação Avançada/efeitos dos fármacos , Animais , Modelos Animais de Doenças , Regulação para Baixo/efeitos dos fármacos , Camundongos , Camundongos Transgênicos , Regulação para Cima/efeitos dos fármacos
3.
J Cell Physiol ; 234(7): 10547-10553, 2019 07.
Artigo em Inglês | MEDLINE | ID: mdl-30480814

RESUMO

OBJECTIVE: To investigate the role of high mobility group box 1 (HMGB1) and receptor for advanced glycation end product (RAGE) in the lungs of hyperoxia-induced rats and the effect of N--acetlycystein (NAC). METHODS: A model of hyperoxic lung injury was established, rats in the NAC intervention, and control, hyperoxia group were given nebulized NAC aerosol, nebulized same volume of saline once a day for 7 consecutive days, respectively. Wet/dry ( W/ D) ratio of the lungs was determined to evaluate the edema of the lung tissues. Conventional hematoxylin-eosin (HE) staining was used to observe the pathological changes of lung tissues. Immunohistochemical staining was used to investigate the expression of HMGB1 and RAGE in the lung tissues. Quantitative reverse-transcription polymerase chain reaction and western blot analysis were used to measured the changes in the messenger RNA (mRNA) and protein expression of HMGB1 and RAGE, respectively. RESULTS: Weight gain of the rats in the hyperoxia group was significantly slower than that in the control group and intervention group (p < 0.05). HE staining results showed lung tissues in the hyperoxia group were severely damaged compared with control group. W/D ratio in hyperoxia group was significantly higher than that in control group and intervention group (p < 0.05). Protein and mRNA expression of HMGB1 and RAGE in the hyperoxia group were significantly higher than control group and intervention group (p < 0.05). CONCLUSION: HMGB1 and RAGE were involved in the pathogenesis of hyperoxia-induced lung injury, inhalation of NAC might alleviate hyperoxia-induced lung injury by regulating the expression of HMGB1 and RAGE.


Assuntos
Acetilcisteína/farmacologia , Proteína HMGB1/genética , Hiperóxia/tratamento farmacológico , Lesão Pulmonar/tratamento farmacológico , Receptor para Produtos Finais de Glicação Avançada/efeitos dos fármacos , Animais , Animais Recém-Nascidos , Modelos Animais de Doenças , Regulação da Expressão Gênica/efeitos dos fármacos , Humanos , Hiperóxia/complicações , Hiperóxia/genética , Hiperóxia/patologia , Pulmão/efeitos dos fármacos , Pulmão/patologia , Lesão Pulmonar/etiologia , Lesão Pulmonar/genética , Lesão Pulmonar/patologia , NF-kappa B/genética , Nebulizadores e Vaporizadores , Ratos , Transdução de Sinais/efeitos dos fármacos , Fator de Necrose Tumoral alfa/genética
4.
Respir Res ; 20(1): 27, 2019 Feb 06.
Artigo em Inglês | MEDLINE | ID: mdl-30728013

RESUMO

BACKGROUND: Animal production workers are persistently exposed to organic dust and can suffer from a variety of respiratory disease symptoms and annual decline in lung function. The role of high mobility group box-1 (HMGB1) in inflammatory airway diseases is emerging. Hence, we tested a hypothesis that organic dust exposure of airway epithelial cells induces nucleocytoplasmic translocation of HMGB1 and blocking this translocation dampens organic dust-induced lung inflammation. METHODS: Rats were exposed to either ambient air or swine barn (8 h/day for either 1, 5, or 20 days) and lung tissues were processed for immunohistochemistry. Swine barn dust was collected and organic dust extract (ODE) was prepared and sterilized. Human airway epithelial cell line (BEAS-2B) was exposed to either media or organic dust extract followed by treatment with media or ethyl pyruvate (EP) or anti-HMGB1 antibody. Immunoblotting, ELISA and other assays were performed at 0 (control), 6, 24 and 48 h. Data (as mean ± SEM) was analyzed using one or two-way ANOVA followed by Bonferroni's post hoc comparison test. A p value of less than 0.05 was considered significant. RESULTS: Compared to controls, barn exposed rats showed an increase in the expression of HMGB1 in the lungs. Compared to controls, ODE exposed BEAS-2B cells showed nucleocytoplasmic translocation of HMGB1, co-localization of HMGB1 and RAGE, reactive species and pro-inflammatory cytokine production. EP treatment reduced the ODE induced nucleocytoplasmic translocation of HMGB1, HMGB1 expression in the cytoplasmic fraction, GM-CSF and IL-1ß production and augmented the production of TGF-ß1 and IL-10. Anti-HMGB1 treatment reduced ODE-induced NF-κB p65 expression, IL-6, ROS and RNS but augmented TGF-ß1 and IL-10 levels. CONCLUSIONS: HMGB1-RAGE signaling is an attractive target to abrogate OD-induced lung inflammation.


Assuntos
Poeira , Proteína HMGB1/metabolismo , Pneumonia/tratamento farmacológico , Piruvatos/uso terapêutico , Receptor para Produtos Finais de Glicação Avançada/metabolismo , Doenças Respiratórias/tratamento farmacológico , Transdução de Sinais/efeitos dos fármacos , Transporte Ativo do Núcleo Celular/efeitos dos fármacos , Animais , Linhagem Celular , Citocinas/biossíntese , Proteína HMGB1/efeitos dos fármacos , Humanos , Imuno-Histoquímica , Pneumonia/etiologia , Ratos , Receptor para Produtos Finais de Glicação Avançada/efeitos dos fármacos , Doenças Respiratórias/etiologia , Suínos
5.
Crit Rev Food Sci Nutr ; 59(7): 1169-1177, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-29185795

RESUMO

OBJECTIVES: This study was aimed to review and collate effects of curcumin on generation of advanced glycation end products (AGEs) and AGEs induced detrimental agents. METHODS: Pubmed, Googlescholar, ScienceDirect, and Scopus databases were searched. Searching was not limited to specific publication period. Only English language original articles (in vitro, experimental and human) which had examined the effect of curcumin on AGEs formation and AGEs induced apoptosis, oxidative stress or inflammatory responses were included. To review effect of curcumin on AGEs formation, search terms were as following: ''curcumin" (title) and AGEs or pentosidine or methylglyoxal or carboxymethyllysine or glucosylation (title/abstract). Totally 104 articles were searched which 19 were selected for review. To review effect of curcumin on AGEs induced harmful agents, key words were as following: "curcumin" (title) and AGEs (title/abstract) and apoptosis or oxidative stress or DNA damage or cell injury or inflammatory or cell death or cell proliferation (title/abstract). Totally 126 articles were searched which 18 were found appropriate for review. RESULTS: Regarding curcumin and AGEs formation, ten eligible articles (1 human trial, 5 animal models and 4 in vitro) and with regarding curcumin and AGEs-induced complications, 17 articles (5 on apoptosis, 9 on oxidative stress, and 3 on inflammatory responses) were selected. Except one, all studies indicated that curcumin is able to prevent AGEs formation and AGEs-induced disturbances with different potential mechanisms. CONCLUSION: Curcumin can inhibit AGEs formation and AGEs-induced disturbances. More RCT researches are suggested to evaluate beneficial effect of curcumin regarding AGEs in different age-related chronic diseases, with specific attention to AGEs memberships.


Assuntos
Curcumina/farmacologia , Produtos Finais de Glicação Avançada/efeitos dos fármacos , Receptor para Produtos Finais de Glicação Avançada/efeitos dos fármacos , Animais , Antioxidantes , Apoptose/efeitos dos fármacos , Arginina/análogos & derivados , Arginina/farmacologia , Morte Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Dano ao DNA/efeitos dos fármacos , Bases de Dados Factuais , Humanos , Lisina/análogos & derivados , Lisina/farmacologia , NF-kappa B , Estresse Oxidativo/efeitos dos fármacos , Aldeído Pirúvico/farmacologia
6.
Med Sci Monit ; 25: 1113-1121, 2019 Feb 11.
Artigo em Inglês | MEDLINE | ID: mdl-30739905

RESUMO

BACKGROUND Diabetic retinopathy (DR) is a macrovascular complication that occurs in diabetic patients. Conventional treatments for the management of DR have many limitations. Thus, the present investigation evaluated the protective effect of fangchinoline against diabetic retinopathy (DR). MATERIAL AND METHODS DR was induced by streptozotocin (STZ; 60 mg/kg; i.p.) and rats were treated with fangchinoline 1, 3, and 10 mg/kg for 16 weeks. DR was confirmed by determining the concentration of advanced glycation end-products (AGEs) and morphology of retinal tissues. Parameters of oxidative stress and expression of inflammatory cytokines and receptor for advanced glycation end-products (RAGE) in the retinal tissue were determined by Western blot assay and reverse transcription polymerase chain reaction (RT-PCR). Moreover enzyme-linked immunosorbent assay (ELISA) was used to determine the apoptosis index and activity of nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB) in the retinal tissues. RESULTS Our study reveals that the concentration of glycosylated hemoglobin (HbA1c) and glucose in the plasma and AGEs in the retinal tissue were significantly reduced in the fangchinoline group compared to the DR group. Moreover, treatment with fangchinoline attenuated the altered retinal morphology and expression of inflammatory mediators and RAGE in the retinal tissues of DR rats. There was a significant (p<0.01) decrease in oxidative stress, activity of NF-κB, and apoptosis index in the fangchinoline group compared to the DR group of rats. CONCLUSIONS Our investigation shows that fangchinoline attenuates the apoptosis of retinal cells in STZ-induced diabetic retinopathy rats by inhibiting the RAGE/NF-κB pathway.


Assuntos
Benzilisoquinolinas/farmacologia , Retinopatia Diabética/tratamento farmacológico , Animais , Apoptose/efeitos dos fármacos , Benzilisoquinolinas/uso terapêutico , Citocinas , Diabetes Mellitus Experimental/induzido quimicamente , Diabetes Mellitus Experimental/tratamento farmacológico , Hemoglobinas Glicadas , Masculino , NF-kappa B/efeitos dos fármacos , Estresse Oxidativo , Ratos , Ratos Wistar , Receptor para Produtos Finais de Glicação Avançada/efeitos dos fármacos , Retina , Transdução de Sinais
7.
Med Sci Monit ; 25: 7813-7825, 2019 Oct 18.
Artigo em Inglês | MEDLINE | ID: mdl-31625533

RESUMO

BACKGROUND The aim of this study was to investigate the protective mechanism of neurovascular unit of Buyang Huanwu decoction (BYHWD) in an Alzheimer's disease (AD) cell model via RAGE/LRP1 pathway and find a reliable target for Alzheimer's disease treatment. MATERIAL AND METHODS Rat brain microvessel endothelial cells (BMECs) were cultured in 10% FBS and 1% penicillin/streptomycin. The AD model was established by administration of 24 µmol/L amyloid-ß peptides 25~35. Different concentrations of BYHWD (0.1 mg/mL, 1 mg/mL, and 10 mg/mL) were added as the drug intervention. The morphology of the cells was observed by light microscopy and the ultrastructure of the cells was observed by microscopy. The inflammatory factors IL-1ß, IL-6, TNF-alpha, and Aß25-35 were detected by ELISA. Flow cytometry was used to assess the apoptosis rate. The expressions of RAGE, LRP1, ICAM-1, VCAM-1, Apo J, Apo E, and NF-kappaBp65 were detected by Western blotting. RESULTS The structure of cells in BYHWDM and BYHWDH gradually recovered with increasing dose. BYHWD decreased the apoptotic rate of BMECs induced by Aß25-35. The cells treated with different concentrations of BYHWD had significant difference in terms of anti-apoptotic effect. The therapeutic effect of BYHWD on AD was via the RAGE/LRP1 and NF-kappaBp65 pathways. CONCLUSIONS BYHWD regulates Aß metabolism via the RAGE/LRP1 pathway, inhibits vascular endothelial inflammation induced by ICAM-1 and VCAM-1 via the NF-kappaBP65 pathway, and promotes morphological changes induced by Aß-induced brain microvascular endothelial cell damage.


Assuntos
Doença de Alzheimer/tratamento farmacológico , Medicamentos de Ervas Chinesas/farmacologia , Doença de Alzheimer/metabolismo , Peptídeos beta-Amiloides/metabolismo , Animais , Apoptose/efeitos dos fármacos , Encéfalo/metabolismo , Proliferação de Células/efeitos dos fármacos , Medicamentos de Ervas Chinesas/metabolismo , Células Endoteliais/metabolismo , Inflamação/tratamento farmacológico , Inflamação/metabolismo , Proteína Associada a Proteínas Relacionadas a Receptor de LDL/efeitos dos fármacos , Proteína Associada a Proteínas Relacionadas a Receptor de LDL/metabolismo , Modelos Biológicos , Cultura Primária de Células , Ratos , Receptor para Produtos Finais de Glicação Avançada/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos
8.
Biosci Biotechnol Biochem ; 82(1): 57-64, 2018 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-29191087

RESUMO

Alzheimer's disease (AD) is the most common cause of dementia among elderly population. Deranged ß-amyloid (Aß) trafficking across the blood-brain barrier is known to be a critical element in the pathogenesis of AD. In the vascular endothelial cells of hippocampus, Aß transport is mainly mediated by low-density lipoprotein-associated protein 1 (LRP1) and the receptor for advanced glycation end (RAGE) products; therefore, LRP1 and RAGE endothelial cells are potential therapeutic targets for AD. In this study, we explored the effects of Formononetin (FMN) on learning and memory improvement in APP/PS1 mice and the related mechanisms. We found that FMN significantly improved learning and memory ability by suppressing Aß production from APP processing, RAGE-dependent inflammatory signaling and promoted LRP1-dependent cerebral Aß clearance pathway. Moreover, FMN treatment alleviated ultrastructural changes in hippocampal vascular endothelial cells. In conclusion, we believe that FMN may be an efficacious and promising treatment for AD.


Assuntos
Doença de Alzheimer/tratamento farmacológico , Sistemas de Liberação de Medicamentos , Isoflavonas/uso terapêutico , Fármacos Neuroprotetores , Animais , Humanos , Proteína-1 Relacionada a Receptor de Lipoproteína de Baixa Densidade , Camundongos , Fármacos Neuroprotetores/uso terapêutico , Receptor para Produtos Finais de Glicação Avançada/efeitos dos fármacos , Receptores de LDL/efeitos dos fármacos , Proteínas Supressoras de Tumor/efeitos dos fármacos
9.
Microvasc Res ; 112: 30-36, 2017 07.
Artigo em Inglês | MEDLINE | ID: mdl-28209519

RESUMO

Crocetin is a major active constituent of Gardenia jasminoides J. Ellis, and can aid in the prevention of cardiovascular disease. The effect and possible mechanism of crocetin on the migration of vascular smooth muscle cells (VSMCs) induced by advanced glycosylation end products (AGEs) were investigated. VSMCs were pre-incubated with or without crocetin and exposed to AGEs subsequently. The invasion of the cells was investigated using a 24-well Cell Invasion Chamber. The anti-proliferative activity of crocetin was evaluated by MTT assay and VSMCs cell-cycle distribution was examined by flow cytometry. Cytokine TNF-α and IL-6 secreted by VSMCs and the amount of matrix metalloproteinase MMP-2 and MMP-9 in the culture supernatant were detected by ELISA. The expression level of RAGE (AGEs receptor), in cells was analyzed by western blot. The results demonstrated that AGEs increased about two-fold migration of VSMCs compared with control (OD=0.778±0.191 vs OD=0.413±0.214, P<0.01), and the proliferation increased by about 20% (OD=0.335±0.043 vs OD=0.281±0.037, P<0.01). Pre-treatment with crocetin (1.0µM) or RAGE antibody (10µg/ml) could inhibit the AGEs triggered migration of VSMCs obviously. Furthermore, both crocetin and RAGE antibody inhibited the increase of RAGE protein in VSMCs stimulated by AGEs. The levels of TNF-α and IL-6 decreased in the crocetin (1.0µM) pre-treated group compared to the AGEs (without pre-treated) group (37.60±3.08pg/ml vs 46.59±1.92pg/ml, 32.11±4.69pg/ml vs 49.99±8.84pg/ml, respectively). Crocetin (1.0µM) also reduced the value of MMP-2 and MMP-9 compared with the AGEs group (2.81±0.35ng/ml vs 6.40±0.85ng/ml, 2.69±0.25ng/ml vs 4.32±0.57ng/ml, respectively). In summary, crocetin inhibits the migration of VSMCs induced by AGEs through RAGE-dependent signaling pathway. And it is meaningful to diabetic vascular complications.


Assuntos
Carotenoides/farmacologia , Movimento Celular/efeitos dos fármacos , Angiopatias Diabéticas/prevenção & controle , Produtos Finais de Glicação Avançada/toxicidade , Músculo Liso Vascular/efeitos dos fármacos , Miócitos de Músculo Liso/efeitos dos fármacos , Animais , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Angiopatias Diabéticas/metabolismo , Angiopatias Diabéticas/patologia , Relação Dose-Resposta a Droga , Interleucina-6/metabolismo , Metaloproteinase 2 da Matriz/metabolismo , Metaloproteinase 9 da Matriz/metabolismo , Músculo Liso Vascular/metabolismo , Músculo Liso Vascular/patologia , Miócitos de Músculo Liso/metabolismo , Miócitos de Músculo Liso/patologia , Ratos , Receptor para Produtos Finais de Glicação Avançada/efeitos dos fármacos , Receptor para Produtos Finais de Glicação Avançada/metabolismo , Transdução de Sinais/efeitos dos fármacos , Fator de Necrose Tumoral alfa/metabolismo , Vitamina A/análogos & derivados
10.
Eur J Clin Invest ; 47(9): 675-683, 2017 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-28722189

RESUMO

BACKGROUND: Receptor for advanced glycation end products (RAGE) signalling plays a critical role in the pathogenesis of cardiovascular disease. Calcitriol modulates cardiac RAGE expression. This study explored the mechanisms underlying the effect of calcitriol on RAGE and soluble RAGE (sRAGE) expression in cardiomyocytes. MATERIALS AND METHODS: Western blot, ELISA, fluorometric assay and PCR analyses were used to evaluate the RAGE, sRAGE, endogenous secretory RAGE (esRAGE), Jun N-terminal kinase (JNK), and a disintegrin and metalloprotease 10 (ADAM10) expression and enzyme activity in HL-1 atrial myocytes without and with calcitriol (10 and 100 nM), nuclear factor-κB (NF-κB) inhibitor (50 µg/mL), or ADAM10 inhibitor (5 µM) incubation for 48 h. RESULTS: Calcitriol (10 nM) significantly reduced RAGE protein expression and increased sRAGE concentrations in HL-1 cardiomyocytes compared with control cells. These changes were associated with increased protein expression and enzyme activity of ADAM10 and higher mRNA expression of esRAGE. In the presence of ADAM10 inhibitor, however, the suppressive effect of calcitriol on RAGE was diminished. Methylglyoxal (500 µM for 10 min)-mediated JNK phosphorylation was attenuated in the presence of calcitriol (10 nM). Moreover, control and NF-κB inhibitor-treated HL-1 cells had similar RAGE and sRAGE expression, suggesting that calcitriol-mediated RAGE modulation was independent of NF-κB signalling. CONCLUSIONS: We showed that RAGE downregulation and increased sRAGE production by calcitriol were mediated through ADAM10 activation in cardiomyocytes. The results suggest that calcitriol has therapeutic potential in treating RAGE-mediated cardiovascular complications.


Assuntos
Proteína ADAM10/efeitos dos fármacos , Secretases da Proteína Precursora do Amiloide/efeitos dos fármacos , Calcitriol/farmacologia , Proteínas Quinases JNK Ativadas por Mitógeno/efeitos dos fármacos , Proteínas de Membrana/efeitos dos fármacos , Miócitos Cardíacos/efeitos dos fármacos , Receptor para Produtos Finais de Glicação Avançada/efeitos dos fármacos , Vitaminas/farmacologia , Proteína ADAM10/antagonistas & inibidores , Proteína ADAM10/metabolismo , Secretases da Proteína Precursora do Amiloide/antagonistas & inibidores , Secretases da Proteína Precursora do Amiloide/metabolismo , Animais , Western Blotting , Regulação para Baixo , Ensaio de Imunoadsorção Enzimática , Átrios do Coração/citologia , Proteínas Quinases JNK Ativadas por Mitógeno/metabolismo , Proteínas de Membrana/antagonistas & inibidores , Proteínas de Membrana/metabolismo , Camundongos , Miócitos Cardíacos/metabolismo , NF-kappa B/antagonistas & inibidores , Reação em Cadeia da Polimerase em Tempo Real , Receptor para Produtos Finais de Glicação Avançada/genética , Receptor para Produtos Finais de Glicação Avançada/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa
11.
Med Sci Monit ; 23: 6121-6131, 2017 Dec 26.
Artigo em Inglês | MEDLINE | ID: mdl-29278639

RESUMO

BACKGROUND Peroxisome proliferator-activated receptor-g (PPAR-g) exhibits anti-inflammatory and anti-diabetic properties, and is protective against cardiovascular diseases. This study aimed to determine the effects of a PPAR-g agonist pioglitazone on atherogenesis in an ApoE knockout mouse (ApoE-/-) diabetic mouse model and in a cultured vascular smooth muscle cells (VSMCs) model. MATERIAL AND METHODS Male ApoE-/- mice were rendered diabetic by 5 daily intraperitoneal injections of streptozotocin. Pioglitazone (20 mg/kg/d) or PPAR-γ inhibitor GW9662 (1 mg/kg/d) were administered for 12 weeks. At the end of treatment, mice were killed and the aortae were isolated. Oil Red O staining was used to evaluate atherosclerotic plaque area. H&E staining was used to evaluate the number of complicated plaques. Western blotting and immunohistochemistry were used to determine the expression of advanced glycation end-products (RAGE) and PPAR-γ. The effects of pioglitazone and GW9662 on RAGE and PPAR-g expression were examined in cultured primary mouse VSMCs in hyperglycemic conditions. RESULTS Administration of pioglitazone in diabetic ApoE-/- mice successfully reduced atherosclerotic plaque area and the number of complicated plaques. Moreover, pioglitazone inhibited RAGE and stimulated PPAR-γ protein expression in atherosclerotic plaques of diabetic ApoE-/- mice. In cultured VSMCs upon high-glucose challenge, pioglitazone downregulated RAGE mRNA and protein expression. Blockade of PPAR-γ activity by GW9662 remarkably attenuated the inhibitory actions of pioglitazone on atherogenesis, both in diabetic ApoE-/- mice and in cultured VSMCs, upon high-glucose challenge. CONCLUSIONS Pioglitazone has a therapeutic effect on atherosclerosis in diabetes, and inhibition of RAGE signaling plays a critical role in mediating the beneficial effects of pioglitazone.


Assuntos
Aterosclerose/tratamento farmacológico , Receptor para Produtos Finais de Glicação Avançada/efeitos dos fármacos , Tiazolidinedionas/farmacologia , Animais , Apolipoproteínas E/metabolismo , Aterosclerose/metabolismo , Células Cultivadas , Complicações do Diabetes/tratamento farmacológico , Diabetes Mellitus Experimental/metabolismo , Angiopatias Diabéticas/etiologia , Modelos Animais de Doenças , Masculino , Camundongos , Camundongos Knockout , Músculo Liso Vascular/metabolismo , PPAR gama/antagonistas & inibidores , Pioglitazona , Placa Aterosclerótica/complicações , Receptor para Produtos Finais de Glicação Avançada/metabolismo , Transdução de Sinais/genética , Tiazolidinedionas/metabolismo
12.
Biol Res ; 50(1): 23, 2017 Jun 21.
Artigo em Inglês | MEDLINE | ID: mdl-28637501

RESUMO

BACKGROUND: Airway remodeling is a key feature of asthma, characterized by increased proliferation of airway smooth muscle cells (ASMCs). S100A8 is a calcium-binding protein with a potential to regulate cell proliferation. Here, the effect of exogenous S100A8 protein on the proliferation of ASMCs induced by platelet-derived growth factor (PDGF) and the underlying molecular mechanism was investigated. METHODS: Rat ASMCs were cultured with or without a neutralizing antibody to the receptor for advanced glycation end-products (RAGE), a potential receptor for S100A8 protein. Purified recombinant rat S100A8 protein was then added into the cultured cells, and the proliferation of ASMCs induced by PDGF was detected by colorimetric-based WST-8 assay and ampedance-based xCELLigence proliferation assay. The expression levels of RAGE in ASMCs were analyzed using western blotting assay. RESULTS: Results showed that exogenous S100A8 inhibited the PDGF-induced proliferation of rat ASMCs in a dose-dependent manner with the maximal effect at 1 µg/ml in vitro. Furthermore, when ASMCs was pre-treated with anti-RAGE neutralizing antibody, the inhibitory effect of S100A8 on PDGF-induced proliferation was significantly suppressed. In addition, neither the treatment with S100A8 or PDGF alone nor the pre-treatment with rS100A8 followed by PDGF stimulation affected the expression levels of RAGE. CONCLUSIONS: Our study demonstrated that S100A8 inhibits PDGF-induced ASMCs proliferation in a manner dependent on membrane receptor RAGE.


Assuntos
Calgranulina A/administração & dosagem , Proliferação de Células/efeitos dos fármacos , Miócitos de Músculo Liso/efeitos dos fármacos , Fator de Crescimento Derivado de Plaquetas/agonistas , Receptor para Produtos Finais de Glicação Avançada/efeitos dos fármacos , Animais , Células Cultivadas , Ratos
13.
Nutr Metab Cardiovasc Dis ; 26(9): 797-807, 2016 09.
Artigo em Inglês | MEDLINE | ID: mdl-27212619

RESUMO

BACKGROUND AND AIMS: Advanced glycation end products (AGEs)-receptor RAGE interaction evokes oxidative stress and inflammatory reactions, thereby being involved in endothelial cell (EC) damage in diabetes. Sulforaphane is generated from glucoraphanin, a naturally occurring isothiocyanate found in widely consumed cruciferous vegetables, by myrosinase. Sulforaphane has been reported to protect against oxidative stress-mediated cell and tissue injury. However, effects of sulforaphane on AGEs-induced vascular damage remain unclear. METHODS AND RESULTS: In this study, we investigated whether and how sulforaphane could inhibit inflammation in AGEs-exposed human umbilical vein ECs (HUVECs) and AGEs-injected rat aorta. Sulforaphane treatment for 4 or 24 h dose-dependently inhibited the AGEs-induced increase in RAGE, monocyte chemoattractant protein-1 (MCP-1), intercellular adhesion molecule-1 (ICAM-1), and vascular cell adhesion molecular-1 (VCAM-1) gene expression in HUVECs. AGEs significantly stimulated MCP-1 production by, and THP-1 cell adhesion to, HUVECs, both of which were prevented by 1.6 µM sulforaphane. Sulforaphane significantly suppressed oxidative stress generation and NADPH oxidase activation evoked by AGEs in HUVECs. Furthermore, aortic RAGE, ICAM-1 and VCAM-1 expression in AGEs-injected rats were increased, which were suppressed by simultaneous infusion of sulforaphane. CONCLUSION: The present study demonstrated for the first time that sulforaphane could inhibit inflammation in AGEs-exposed HUVECs and AGEs-infused rat aorta partly by suppressing RAGE expression through its anti-oxidative properties. Inhibition of the AGEs-RAGE axis by sulforaphane might be a novel therapeutic target for vascular injury in diabetes.


Assuntos
Anti-Inflamatórios/farmacologia , Antioxidantes/farmacologia , Aorta/efeitos dos fármacos , Aortite/prevenção & controle , Células Endoteliais/efeitos dos fármacos , Produtos Finais de Glicação Avançada , Isotiocianatos/farmacologia , Animais , Aorta/metabolismo , Aortite/induzido quimicamente , Aortite/metabolismo , Moléculas de Adesão Celular/metabolismo , Linhagem Celular Tumoral , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Células Endoteliais/metabolismo , Células Endoteliais da Veia Umbilical Humana/efeitos dos fármacos , Células Endoteliais da Veia Umbilical Humana/metabolismo , Mediadores da Inflamação/metabolismo , Masculino , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Ratos Wistar , Receptor para Produtos Finais de Glicação Avançada/efeitos dos fármacos , Receptor para Produtos Finais de Glicação Avançada/genética , Receptor para Produtos Finais de Glicação Avançada/metabolismo , Sulfóxidos , Fatores de Tempo
14.
Am J Physiol Heart Circ Physiol ; 309(7): H1130-40, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-26254329

RESUMO

Mechanisms of restenosis in type 2 diabetes mellitus (T2DM) are incompletely elucidated, but advanced glycation end-product (AGE)-induced vascular remodeling likely contributes. We tested the hypothesis that AGE-related collagen cross-linking (ARCC) leads to increased downstream vascular resistance and altered in-stent hemodynamics, thereby promoting neointimal hyperplasia (NH) in T2DM. We proposed that decreasing ARCC with ALT-711 (Alagebrium) would mitigate this response. Abdominal aortic stents were implanted in Zucker lean (ZL), obese (ZO), and diabetic (ZD) rats. Blood flow, vessel diameter, and wall shear stress (WSS) were calculated after 21 days, and NH was quantified. Arterial segments (aorta, carotid, iliac, femoral, and arterioles) were harvested to detect ARCC and protein expression, including transforming growth factor-ß (TGF-ß) and receptor for AGEs (RAGE). Downstream resistance was elevated (60%), whereas flow and WSS were significantly decreased (44% and 56%) in ZD vs. ZL rats. NH was increased in ZO but not ZD rats. ALT-711 reduced ARCC and resistance (46%) in ZD rats while decreasing NH and producing similar in-stent WSS across groups. No consistent differences in RAGE or TGF-ß expression were observed in arterial segments. ALT-711 modified lectin-type oxidized LDL receptor 1 but not RAGE expression by cells on decellularized matrices. In conclusion, ALT-711 decreased ARCC, increased in-stent flow rate, and reduced NH in ZO and ZD rats through RAGE-independent pathways. The study supports an important role for AGE-induced remodeling within and downstream of stent implantation to promote enhanced NH in T2DM.


Assuntos
Aorta Abdominal/efeitos dos fármacos , Diabetes Mellitus/metabolismo , Oclusão de Enxerto Vascular/metabolismo , Neointima/metabolismo , Obesidade/metabolismo , Stents , Estresse Mecânico , Tiazóis/farmacologia , Resistência Vascular/efeitos dos fármacos , Animais , Aorta Abdominal/metabolismo , Colágeno/efeitos dos fármacos , Colágeno/metabolismo , Produtos Finais de Glicação Avançada/efeitos dos fármacos , Produtos Finais de Glicação Avançada/metabolismo , Masculino , Neointima/prevenção & controle , Ratos , Ratos Zucker , Receptor para Produtos Finais de Glicação Avançada/efeitos dos fármacos , Receptor para Produtos Finais de Glicação Avançada/metabolismo , Resistência ao Cisalhamento , Fator de Crescimento Transformador beta/efeitos dos fármacos , Fator de Crescimento Transformador beta/metabolismo
15.
Exp Dermatol ; 24(6): 418-23, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-25739980

RESUMO

Quercetin, glycosylated form of flavonoid compound, has potent antioxidant and anti-inflammatory properties. In this study, we have investigated the effects of quercetin on skin lesion, high-mobility group box (HMGB)1 cascade signalling and inflammation in atopic dermatitis (AD) mouse model. AD-like lesion was induced by the application of house dust mite extract to the dorsal skin of NC/Nga transgenic mouse. After AD induction, quercetin (50 mg/kg, p.o) was administered daily for 2 weeks. We evaluated dermatitis severity, histopathological changes and changes in protein expression by Western blotting for HMGB1, receptor for advanced glycation end products (RAGE), toll-like receptor (TLR)4, nuclear factor (NF)κB, nuclear factor erythroid-2-related factor (Nrf)2, kelch-like ECH-associated protein (Keap)1, extracellular signal-regulated kinase (ERK)1/2, cyclooxygenase (COX)2, tumor necrosis factor (TNF)α, interleukin (IL)-1ß, IL-2Rα and other inflammatory markers in the skin of AD mice. In addition, serum levels of T helper (Th) cytokines (interferon (IFN)γ, IL-4) were measured by enzyme-linked immunosorbent assay. Quercetin treatment attenuated the development of AD-like skin lesions. Histological analysis showed that quercetin inhibited hyperkeratosis, parakeratosis, acanthosis, mast cells and infiltration of inflammatory cells. Furthermore, quercetin treatment downregulated cytoplasmic HMGB1, RAGE, nuclear p-NFκB, p-ERK1/2, COX2, TNFα, IL-1ß, IL-2Rα, IFNγ and IL-4 and upregulated nuclear Nrf2. Our data demonstrated that the HMGB1/RAGE/NFκB signalling might play an important role in skin inflammation, and quercetin treatment could be a promising agent for AD by modulating the HMGB1/RAGE/NFκB signalling and induction of Nrf2 protein.


Assuntos
Dermatite Atópica/tratamento farmacológico , Proteína HMGB1/genética , NF-kappa B/fisiologia , Quercetina/farmacologia , Receptor para Produtos Finais de Glicação Avançada/fisiologia , Transdução de Sinais/efeitos dos fármacos , Translocação Genética/efeitos dos fármacos , Animais , Antioxidantes/farmacologia , Antioxidantes/uso terapêutico , Dermatite Atópica/etiologia , Dermatite Atópica/fisiopatologia , Dermatophagoides farinae/patogenicidade , Modelos Animais de Doenças , Feminino , Proteína HMGB1/efeitos dos fármacos , Proteína HMGB1/fisiologia , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Sistema de Sinalização das MAP Quinases/genética , Sistema de Sinalização das MAP Quinases/fisiologia , Camundongos , Camundongos Transgênicos , Fator 2 Relacionado a NF-E2/efeitos dos fármacos , Fator 2 Relacionado a NF-E2/genética , Fator 2 Relacionado a NF-E2/fisiologia , NF-kappa B/efeitos dos fármacos , NF-kappa B/genética , Quercetina/uso terapêutico , Receptor para Produtos Finais de Glicação Avançada/efeitos dos fármacos , Receptor para Produtos Finais de Glicação Avançada/genética , Índice de Gravidade de Doença , Transdução de Sinais/genética , Transdução de Sinais/fisiologia , Pele/efeitos dos fármacos , Pele/patologia , Receptor 4 Toll-Like/efeitos dos fármacos , Receptor 4 Toll-Like/genética , Receptor 4 Toll-Like/fisiologia , Translocação Genética/genética , Translocação Genética/fisiologia
16.
Horm Metab Res ; 47(9): 686-92, 2015 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-25611208

RESUMO

Advanced glycation end products (AGEs) and receptor RAGE play a role in diabetic nephropathy. We have previously shown that increased glucose uptake into proximal tubular cells via sodium-glucose cotransporter 2 (SGLT2) stimulates oxidative stress generation and RAGE expression, thereby exacerbating the AGE-induced apoptosis in this cell type. However, the protective role of SGLT2 inhibition against the AGE-RAGE-induced renal damage in diabetic animals remains unclear. In this study, we investigated the effects of empagliflozin, SGLT2 inhibitor on AGE-RAGE axis, inflammatory and fibrotic reactions, and tubular injury in the kidney of streptozotocin-induced diabetic rats.Administration of empagliflozin for 4 weeks significantly improved hyperglycemia and HbA1c, and decreased expression levels of AGEs, RAGE, 8-hydroxydeoxyguanosine (8-OHdG), and F4/80, markers of oxidative stress and macrophages, respectively, in the diabetic kidney. Although empagliflozin did not reduce albuminuria, it significantly decreased urinary excretion levels of 8-OHdG and L-fatty acid binding protein, a marker of tubular injury. Moreover, inflammatory and fibrotic gene expression such as monocyte chemoattractant protein-1, intercellular adhesion molecule-1, plasminogen activator inhibitor-1, transforming growth factor-ß, and connective tissue growth factor was enhanced in the diabetic kidney, all of which were prevented by empagliflozin. The present study suggests that empagliflozin could inhibit oxidative, inflammatory and fibrotic reactions in the kidney of diabetic rats partly via suppression of the AGE-RAGE axis. Blockade of the increased glucose uptake into renal proximal tubular cells by empagliflozin might be a novel therapeutic target for tubulointerstitial damage in diabetic nephropathy.


Assuntos
Compostos Benzidrílicos/farmacologia , Diabetes Mellitus Experimental/tratamento farmacológico , Nefropatias Diabéticas/tratamento farmacológico , Fibromialgia/prevenção & controle , Glucosídeos/farmacologia , Produtos Finais de Glicação Avançada/efeitos dos fármacos , Hipoglicemiantes/farmacologia , Inflamação/prevenção & controle , Estresse Oxidativo/efeitos dos fármacos , Receptor para Produtos Finais de Glicação Avançada/efeitos dos fármacos , Inibidores do Transportador 2 de Sódio-Glicose , Animais , Diabetes Mellitus Experimental/induzido quimicamente , Nefropatias Diabéticas/induzido quimicamente , Expressão Gênica/efeitos dos fármacos , Masculino , Ratos , Ratos Sprague-Dawley
17.
Genet Mol Res ; 14(2): 3984-94, 2015 Apr 27.
Artigo em Inglês | MEDLINE | ID: mdl-25966170

RESUMO

Fimbristylis ovata has been long used as a traditional medicine for chronic inflammatory diseases; however, there are no data regarding its anti-inflammatory properties. In this study, we investigated the effects of F. ovata extracts on the secretion of pro-inflammatory cytokines, cell adhesion molecule, and receptor for advanced glycation end-products (RAGE) in lipopolysaccharide-stimulated cells. F. ovata was extracted using the maceration method with 3 different solvents: ethanol, methanol, and water. The effect of F. ovata extracts on cell viability was evaluated using the MTT assay. Pro-inflammatory cytokines and cell adhesion molecules were investigated by reverse transcription-polymerase chain reaction and an enzyme-linked immunosorbent assay. Upon incubation with F. ovata extracts up to 100 mg/mL, cell viability was more than 80%. F. ovata extracts could inhibit interleukin-6 level and gene expression as well as the RAGE gene in the monocytic cell lineU937. Moreover, the results showed that vascular cell adhesion molecule 1 secretion and gene expression were decreased when lipopolysaccharide-activated brain endothelial cells (bEnd.3) were treated with F. ovata extracts. Therefore, the anti-inflammatory activity of F. ovata extracts may result from their inhibitory actions via the RAGE signaling pathway.


Assuntos
Moléculas de Adesão Celular/efeitos dos fármacos , Cyperaceae/química , Citocinas/efeitos dos fármacos , Extratos Vegetais/farmacologia , Receptor para Produtos Finais de Glicação Avançada/efeitos dos fármacos , Animais , Moléculas de Adesão Celular/genética , Linhagem Celular , Sobrevivência Celular/efeitos dos fármacos , Citocinas/genética , Expressão Gênica , Humanos , Molécula 1 de Adesão Intercelular/genética , Interleucina-6/genética , Lipopolissacarídeos/metabolismo , Camundongos , Receptor para Produtos Finais de Glicação Avançada/genética , Transdução de Sinais/genética , Molécula 1 de Adesão de Célula Vascular/genética
18.
Life Sci ; 338: 122410, 2024 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-38191050

RESUMO

AIM: Endothelial hyperpermeability is an early stage of endothelial dysfunction associated with the progression and development of atherosclerosis. 3'-Sialyllactose (3'-SL) is the most abundant compound in human milk oligosaccharides, and it has the potential to regulate endothelial dysfunction. This study investigated the beneficial effects of 3'-SL on lipopolysaccharide (LPS)-induced endothelial dysfunction in vitro and in vivo. MAIN METHODS: We established LPS-induced endothelial dysfunction models in both cultured bovine aortic endothelial cells (BAECs) and mouse models to determine the effects of 3'-SL. Western blotting, qRT-PCR analysis, immunofluorescence staining, and en face staining were employed to clarify underlying mechanisms. Superoxide production was measured by 2',7'-dichlorofluorescin diacetate, and dihydroethidium staining. KEY FINDINGS: LPS significantly decreased cell viability, whereas 3'-SL treatment mitigated these effects via inhibiting ERK1/2 activation. Mechanistically, 3'-SL ameliorated LPS-induced ROS accumulation leading to ERK1/2 activation-mediated STAT1 phosphorylation and subsequent inhibition of downstream transcriptional target genes, including VCAM-1, TNF-α, IL-1ß, and MCP-1. Interestingly, LPS-induced ERK1/2/STAT1 activation leads to the HMGB1 release from the nucleus into the extracellular space, where it binds to RAGE, while 3'-SL suppressed EC hyperpermeability by suppressing the HMGB1/RAGE axis. This interaction also led to VE-cadherin endothelial junction disassembly and endothelial cell monolayer disruption through ERK1/2/STAT1 modulation. In mouse endothelium, en face staining revealed that 3'-SL abolished LPS-stimulated ROS production and VCAM-1 overexpression. SIGNIFICANCE: Our findings suggest that 3'-SL inhibits LPS-induced endothelial hyperpermeability by suppressing superoxide-mediated ERK1/2/STAT1 activation and HMGB1/RAGE axis. Therefore, 3'-SL may be a potential therapeutic agent for preventing the progression of atherosclerosis.


Assuntos
Aterosclerose , Proteína HMGB1 , Oligossacarídeos , Animais , Bovinos , Camundongos , Aterosclerose/metabolismo , Proteína HMGB1/metabolismo , Células Endoteliais da Veia Umbilical Humana/metabolismo , Lipopolissacarídeos/toxicidade , Lipopolissacarídeos/metabolismo , Sistema de Sinalização das MAP Quinases , Oligossacarídeos/farmacologia , Espécies Reativas de Oxigênio/metabolismo , Fator de Transcrição STAT1/metabolismo , Superóxidos/metabolismo , Molécula 1 de Adesão de Célula Vascular/metabolismo , Receptor para Produtos Finais de Glicação Avançada/efeitos dos fármacos , Receptor para Produtos Finais de Glicação Avançada/metabolismo
19.
Arch Physiol Biochem ; 129(4): 831-846, 2023 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-33508970

RESUMO

The current in-vivo study was premeditated to uncover the protective role of ezetimibe (EZ) against advanced glycation endproducts (AGEs)-related pathologies in experimental diabetes. Our results showed that EZ markedly improved the altered biochemical markers of diabetes mellitus (DM) (FBG, HbA1c, insulin, microalbumin, and creatinine) and cardiovascular disease (in-vivo lipid/lipoprotein level and hepatic HMG-CoA reductase activity) along with diminished plasma carboxymethyl-lysine (CML) and renal fluorescent AGEs level. Gene expression study revealed that EZ significantly down-regulated the renal AGEs-receptor (RAGE), nuclear factor-κB (NFκB-2), transforming growth factor-ß (TGF-ß1), and matrix metalloproteinase-2 (MMP-2) mRNA expression, however, the neuropilin-1 (NRP-1) mRNA expression was up-regulated. In addition, EZ also maintained the redox status via decreasing the lipid peroxidation and protein-bound carbonyl content (CC) and increasing the activity of high-density lipoprotein (HDL)-associated-paraoxonase-1 (PON-1) and renal antioxidant enzymes as well as also protected renal histopathological features. We conclude that EZ exhibits antidiabetic and reno-protective properties in diabetic rats.


Assuntos
Diabetes Mellitus Experimental , Nefropatias Diabéticas , Animais , Ratos , Diabetes Mellitus Experimental/tratamento farmacológico , Diabetes Mellitus Experimental/metabolismo , Nefropatias Diabéticas/metabolismo , Ezetimiba/farmacologia , Ezetimiba/uso terapêutico , Produtos Finais de Glicação Avançada/genética , Produtos Finais de Glicação Avançada/metabolismo , Reação de Maillard , Metaloproteinase 2 da Matriz/metabolismo , NF-kappa B/metabolismo , Estresse Oxidativo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Receptor para Produtos Finais de Glicação Avançada/efeitos dos fármacos
20.
Hum Exp Toxicol ; 41: 9603271221127429, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36112883

RESUMO

BACKGROUND: As an aggressive human malignancy, esophageal squamous cell carcinoma (ESCC) is prevalent globally, especially in China. Verbascoside (VE) exerts anti-cancer effects in several human cancers. This work was to investigate the effects of VE on ESCC cells. METHODS: Esophageal squamous cell carcinoma cell proliferation, apoptosis, migration, and invasion were assessed by CCK-8, TUNEL, and Transwell assays. Gene and protein levels were detected by RT-qPCR and western blotting. CDC42 activity was evaluated by G-lisa assay. RESULTS: Verbascoside significantly inhibited cell proliferation, migration, and invasion and induced cell apoptosis in ESCC cells. Furthermore, it was found that VE markedly inhibited HMGB1 and RAGE expression in a dose-dependent manner. Besides, HMGB1/RAGE upregulation partially reversed the anti-cancer effects of VE on ESCC cells. VE repressed HMGB1/RAGE-induced CDC42 activation in ESCC cells. In addition, ML141-mediated CDC42 inactivation further enhanced the effect of VE on ESCC cell proliferation, apoptosis, migration, and invasion. CONCLUSIONS: Our findings indicated that VE has significant anti-tumor potential in ESCC by suppressing HMGB1/RAGE-dependent CDC42 activation.


Assuntos
Neoplasias Esofágicas , Carcinoma de Células Escamosas do Esôfago , Glucosídeos , Polifenóis , Proteína cdc42 de Ligação ao GTP , Linhagem Celular Tumoral , Movimento Celular , Neoplasias Esofágicas/tratamento farmacológico , Neoplasias Esofágicas/metabolismo , Carcinoma de Células Escamosas do Esôfago/tratamento farmacológico , Carcinoma de Células Escamosas do Esôfago/genética , Carcinoma de Células Escamosas do Esôfago/metabolismo , Glucosídeos/farmacologia , Glucosídeos/uso terapêutico , Proteína HMGB1/efeitos dos fármacos , Proteína HMGB1/metabolismo , Humanos , Invasividade Neoplásica , Fenótipo , Polifenóis/farmacologia , Polifenóis/uso terapêutico , Receptor para Produtos Finais de Glicação Avançada/efeitos dos fármacos , Receptor para Produtos Finais de Glicação Avançada/metabolismo , Sincalida/química , Proteína cdc42 de Ligação ao GTP/efeitos dos fármacos , Proteína cdc42 de Ligação ao GTP/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA