Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 6 de 6
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
Front Endocrinol (Lausanne) ; 14: 1159657, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37334310

RESUMEN

Objective: (-)-Epigallocatechin-3-gallate (EGCG) has preventive effects on obesity-related precocious puberty, but its underlying mechanism remains unclear. The aim of this study was to integrate metabolomics and network pharmacology to reveal the mechanism of EGCG in the prevention of obesity-related precocious puberty. Materials and methods: A high-performance liquid chromatography-electrospray ionization ion-trap tandem mass spectrometry (LC-ESI-MS/MS) was used to analyze the impact of EGCG on serum metabolomics and associated metabolic pathways in a randomized controlled trial. Twelve weeks of EGCG capsules were given to obese girls in this trail. Additionally, the targets and pathways of EGCG in preventing obesity-related precocious puberty network pharmacology were predicted using network pharmacology. Finally, the mechanism of EGCG prevention of obesity-related precocious puberty was elucidated through integrated metabolomics and network pharmacology. Results: Serum metabolomics screened 234 endogenous differential metabolites, and network pharmacology identified a total of 153 common targets. These metabolites and targets mainly enrichment pathways involving endocrine-related pathways (estrogen signaling pathway, insulin resistance, and insulin secretion), and signal transduction (PI3K-Akt, MAPK, and Jak-STAT signaling pathways). The integrated metabolomics and network pharmacology indicated that AKT1, EGFR, ESR1, STAT3, IGF1, and MAPK1 may be key targets for EGCG in preventing obesity-related precocious puberty. Conclusion: EGCG may contribute to preventing obesity-related precocious puberty through targets such as AKT1, EGFR, ESR1, STAT3, IGF1, and MAPK1 and multiple signaling pathways, including the estrogen, PI3K-Akt, MAPK, and Jak-STAT pathways. This study provided a theoretical foundation for future research.


Asunto(s)
Farmacología en Red , Pubertad Precoz , Humanos , Femenino , Fosfatidilinositol 3-Quinasas , Proteínas Proto-Oncogénicas c-akt , Pubertad Precoz/tratamiento farmacológico , Pubertad Precoz/etiología , Espectrometría de Masas en Tándem , Metabolómica , Estrógenos , Receptores ErbB
2.
Biochimie ; 162: 107-115, 2019 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-30876970

RESUMEN

Triple-negative breast cancer (TNBC) is a heterogeneous group of breast cancer with one common feature: distinctly metastatic nature with higher rate of relapse and shorter survival compared with other subtypes of breast cancer. The epithelial to mesenchymal transition (EMT) is highly associated with cancer metastasis. Cyanidin-3-glucoside (C3G), the most abundant anthocyanin pigment enriched in fresh fruits and vegetables, showed ideal anti-oxidant property. C3G could also inhibit certain malignant behaviors of cancer cells, however, whether repression of EMT was involved in its anti-cancer especially TNBC effect remains unknown. Herein, we report that C3G decreases the migratory and invasive nature of TNBC lines MDA-MB-231 and BT-549. Mechanistically, C3G induces reversion of EMT characterized by phenotype modulation with increased epithelial marker E-ca and ZO-1, decreased mesenchymal marker Vimentin, N-ca and EMT-associated transcription factors Snail1, Snail2. NF-κB is pivotal for EMT and Sirt1 is a NF-κB inhibitor. We show that NF-κB is attenuated and Sirt1 is induced by C3G in TNBC, respectively. And later evidence demonstrates that abrogation of Sirt1 with small interfering RNA transfection abolished NF-κB inhibition and EMT reversion by C3G. Subsequently, we show that microRNA-138(miR-138) represses Sirt1 via mRNA translation inhibition and is inhibited by C3G. Moreover, miR-138 repression is involved in Sirt1 re-activation and migratory and invasive inhibition of TNBC by C3G. Taken together, we supplied more evidence to the anti-breast cancer mechanisms of C3G.


Asunto(s)
Antocianinas/farmacología , Transición Epitelial-Mesenquimal/efectos de los fármacos , Glucósidos/farmacología , Sirtuina 1/metabolismo , Neoplasias de la Mama Triple Negativas/enzimología , Antígenos CD/metabolismo , Cadherinas/metabolismo , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Femenino , Humanos , MicroARNs/metabolismo , FN-kappa B/metabolismo , Transducción de Señal/efectos de los fármacos , Factores de Transcripción de la Familia Snail , Vimentina/metabolismo , Proteína de la Zonula Occludens-1/metabolismo
3.
Environ Toxicol ; 31(9): 1068-79, 2016 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-25865073

RESUMEN

The environmental toxicant TCDD may elicit cytotoxic effects by inducing reactive oxygen species (ROS) generation. Autophagy is one of the first lines of defense against oxidative stress damage. Herein, we investigated whether autophagy played a regulatory role in TCDD-induced neurotoxicity. Here, we showed that TCDD exposure caused marked autophagy in SH-SY5Y cells, whose dose range was close to that inducing apoptosis. Electron microscopic and Western blot analyses revealed that TCDD induced autophagy at a starting dose of approximate 100 nM. Interestingly, 100-200 nM TCDD exposure resulted in obviously decreased cell viability and evident apoptotic phenotype. Furthermore, the levels of pro-apoptotic molecules, Bax and cleaved-PARP, increased significantly, whereas Bcl2 declined after exposed to 100 nM TCDD. In addition, the apoptosis was verified using flow cytometrical analysis. These data strongly suggested that TCDD induced both autophagy and apoptosis at a similar dose range in SH-SY5Y cells. Interestingly, pretreatment with ROS scavenger, N-acetyl-cysteine (NAC), could effectively block both TCDD-induced apoptosis and autophagy. More surprisingly, inhibition of autophagy with 3-methyladenine (3MA), remarkably augmented TCDD-induced apoptosis. The findings implicated that the onset of autophagy might serve as a protective mechanism to ameliorate ROS-triggered cytotoxic effects in human SH-SY5Y neuronal cells under TCDD exposure. © 2015 Wiley Periodicals, Inc. Environ Toxicol 31: 1068-1079, 2016.


Asunto(s)
Apoptosis/efectos de los fármacos , Autofagia/efectos de los fármacos , Dibenzodioxinas Policloradas/toxicidad , Sustancias Protectoras/farmacología , Acetilcisteína/farmacología , Adenina/análogos & derivados , Adenina/farmacología , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Humanos , Microscopía Electrónica de Transmisión , Microscopía Fluorescente , Estrés Oxidativo/efectos de los fármacos , Fenotipo , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Especies Reactivas de Oxígeno/metabolismo
4.
J Appl Toxicol ; 35(7): 851-60, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-25382668

RESUMEN

2, 3, 7, 8-tetrachlorodibenzo-p-dioxin (TCDD) is a ubiquitous environmental contaminant that could exert significant neurotoxicity in the human nervous system. Nevertheless, the molecular mechanism underlying TCDD-mediated neurotoxicity has not been clarified clearly. Herein, we investigated the potential role of TCDD in facilitating premature senescence in astrocytes and the underlying molecular mechanisms. Using the senescence-associated ß-galactosidase (SA-ß-Gal) assay, we demonstrated that TCDD exposure triggered significant premature senescence of astrocyte cells, which was accompanied by a marked activation of the Wingless and int (WNT)/ß-catenin signaling pathway. In addition, TCDD altered the expression of senescence marker proteins, such as p16, p21 and GFAP, which together have been reported to be upregulated in aging astrocytes, in both dose- and time-dependent manners. Further, TCDD led to cell-cycle arrest, F-actin reorganization and the accumulation of cellular reactive oxygen species (ROS). Moreover, the ROS scavenger N-acetylcysteine (NAC) markedly attenuated TCDD-induced ROS production, cellular oxidative damage and astrocyte senescence. Notably, the application of XAV939, an inhibitor of WNT/ß-catenin signaling pathway, ameliorated the effect of TCDD on cellular ß-catenin level, ROS production, cellular oxidative damage and premature senescence in astrocytes. In summary, our findings indicated that TCDD might induce astrocyte senescence via WNT/ß-catenin and ROS-dependent mechanisms.


Asunto(s)
Astrocitos/efectos de los fármacos , Senescencia Celular/efectos de los fármacos , Dioxinas/farmacología , Especies Reactivas de Oxígeno/metabolismo , Vía de Señalización Wnt/efectos de los fármacos , Animales , Western Blotting , Ciclo Celular/efectos de los fármacos , Daño del ADN/efectos de los fármacos , Dioxinas/toxicidad , Técnica del Anticuerpo Fluorescente , Ratas , Ratas Sprague-Dawley
5.
Neurotoxicology ; 44: 149-59, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-24932542

RESUMEN

Studies have shown that 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) induces apoptotic cell death in neuronal cells. However, whether this is the result of endoplasmic reticulum (ER) stress-mediated apoptosis remains unknown. In this study, we determined whether ER stress plays a role in the TCDD-induced apoptosis of pheochromocytoma (PC12) cells and primary neurons. PC12 cells were exposed to different TCDD concentrations (1, 10, 100, 200, or 500nM) for varying lengths of time (1, 3, 6, 12, or 24h). TCDD concentrations much higher than 10nM (100, 200, or 500nM) markedly increased glucose-regulated protein (GRP78) and C/EBP homologous protein (CHOP) levels, which are hallmarks of ER stress. We also evaluated the effects of TCDD on ER morphology in PC12 cells and primary neurons that were treated with different TCDD concentrations (1, 10, 50, or 200nM) for 24h. Ultrastructural ER alterations were observed with transmission electron microscopy in PC12 cells and primary neurons treated with high concentrations of TCDD. Furthermore, TCDD-induced ER stress significantly promoted the activation of the PKR-like ER kinase (PERK), a sensor for the unfolded protein response (UPR), and its downstream target eukaryotic translation initiation factor 2 α (eIF2α); in contrast, TCDD did not appear to affect inositol-requiring enzyme 1 (IRE1) and activating transcription factor 6 (ATF6), two other UPR sensors. Importantly, TCDD significantly inhibited eIF2α phosphorylation and triggered apoptosis in PC12 cells after 6-24h of treatment. Salubrinal, which activates the PERK-eIF2α pathway, significantly enhanced eIF2α phosphorylation in PC12 cells and attenuated the TCDD-induced cell death. In contrast, knocking down eIF2α using small interfering RNA markedly enhanced TCDD-induced cell death. Together, these results indicate that the PERK-eIF2α pathway plays an important role in TCDD-induced ER stress and apoptosis in PC12 cells.


Asunto(s)
Apoptosis/efectos de los fármacos , Estrés del Retículo Endoplásmico/efectos de los fármacos , Dibenzodioxinas Policloradas/toxicidad , eIF-2 Quinasa/metabolismo , Animales , Cinamatos/farmacología , Retículo Endoplásmico/enzimología , Retículo Endoplásmico/ultraestructura , Células PC12 , Ratas , Transducción de Señal , Tiourea/análogos & derivados , Tiourea/farmacología , Respuesta de Proteína Desplegada
6.
PLoS One ; 9(2): e89811, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24587053

RESUMEN

The widespread environmental pollutant 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) is a potent toxicant that causes significant neurotoxicity. However, the biological events that participate in this process remain largely elusive. In the present study, we demonstrated that TCDD exposure triggered apparent premature senescence in rat pheochromocytoma (PC12) and human neuroblastoma SH-SY5Y cells. Senescence-associated ß-galactosidase (SA-ß-Gal) assay revealed that TCDD induced senescence in PC12 neuronal cells at doses as low as 10 nM. TCDD led to F-actin reorganization and the appearance of an alternative senescence marker, γ-H2AX foci, both of which are important features of cellular senescence. In addition, TCDD exposure altered the expression of senescence marker proteins, such as p16, p21 and p-Rb, in both dose- and time-dependent manners. Furthermore, we demonstrated that TCDD promotes mitochondrial dysfunction and the accumulation of cellular reactive oxygen species (ROS) in PC12 cells, leading to the activation of signaling pathways that are involved in ROS metabolism and senescence. TCDD-induced ROS generation promoted significant oxidative DNA damage and lipid peroxidation. Notably, treatment with the ROS scavenger N-acetylcysteine (NAC) markedly attenuated TCDD-induced ROS production, cellular oxidative damage and neuronal senescence. Moreover, we found that TCDD induced a similar ROS-mediated senescence response in human neuroblastoma SH-SY5Y cells. In sum, these results demonstrate for the first time that TCDD induces premature senescence in neuronal cells by promoting intracellular ROS production, supporting the idea that accelerating the onset of neuronal senescence may be an important mechanism underlying TCDD-induced neurotoxic effects.


Asunto(s)
Senescencia Celular/efectos de los fármacos , Contaminantes Ambientales/toxicidad , Tejido Nervioso/efectos de los fármacos , Dibenzodioxinas Policloradas/toxicidad , Especies Reactivas de Oxígeno/metabolismo , Actinas/metabolismo , Análisis de Varianza , Animales , Biomarcadores/metabolismo , Western Blotting , Daño del ADN/efectos de los fármacos , Cartilla de ADN/genética , Relación Dosis-Respuesta a Droga , Contaminantes Ambientales/metabolismo , Fluorescencia , Histonas/metabolismo , Humanos , Peroxidación de Lípido/efectos de los fármacos , Tejido Nervioso/citología , Células PC12 , Fosfoproteínas/metabolismo , Dibenzodioxinas Policloradas/metabolismo , Ratas , Reacción en Cadena en Tiempo Real de la Polimerasa , Factores de Tiempo , beta-Galactosidasa/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA