Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 63
Filtrar
Más filtros

Medicinas Complementárias
Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
J Ethnopharmacol ; 283: 114456, 2022 Jan 30.
Artículo en Inglés | MEDLINE | ID: mdl-34333105

RESUMEN

ETHNOPHARMACOLOGICAL RELEVANCE: Hepatocellular carcinoma (HCC) is a liver malignancy which lacks effective treatment and has a poor prognosis. ß-Elemene refers to a natural Curcuma wenyujin-derived single molecular entity, which exhibits various biological activities, and is especially well-known for it's antitumor properties. AIM OF THE RESEARCH: LncRNA HOTAIR, SP1, and PDK1 have displayed oncogenic roles in many tumors, participating in the initiation and progression of cancers by mediating multiple signaling pathways. However, there are only a few reports about their roles and mutual relationship in the growth of HCC cells. Therefore, this study aimed to investigate the expression of LncRNA HOTAIR, SP1, and PDK1 and their interaction with ß-Elemene in HCC cells. MATERIALS AND METHODS: MTT, a Colony formation assay, and flow cytometry were employed to evaluate the growth of HCC and LO2 cells under ß-Elemene. LncRNA HOTAIR, SP1 and PDK1 plasmids were transfected into HCC cells by a transient transfection assay, and the expression and interaction of LncRNA HOTAIR, SP1 and PDK1 were assessed via qRT-PCR and western blotting. RESULTS: ß-Elemene suppressed HCC cell growth through the downregulation of LncRNA HOTAIR, SP1 and PDK1. The results demonstrated a reciprocal interaction among LncRNA HOTAIR, SP1 and PDK1. Exogenous overexpression LncRNA HOTAIR or SP1 eliminated the suppressive effects of ß-Elemene on them, and both of which regulated PDK1 expression in HCC cells. Additionally, exogenously overexpressed SP1 or LncRNA HOTAIR prevented ß-Elemene inhibition of the protein-level expression of PDK1, whereas overexpressing PDK1 had no effect on SP1, though it still weakened the inhibition of cell growth and LncRNA HOTAIR expression by ß-Elemene. CONCLUSION: ß-Elemene suppresses HCC cell proliferation via through the regulation of LncRNA HOTAIR, SP1, PDK1 and their interaction.


Asunto(s)
Carcinoma Hepatocelular/tratamiento farmacológico , Neoplasias Hepáticas/tratamiento farmacológico , ARN Largo no Codificante/genética , Sesquiterpenos/farmacología , Antineoplásicos Fitogénicos/farmacología , Carcinoma Hepatocelular/genética , Línea Celular , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica , Células Hep G2 , Humanos , Neoplasias Hepáticas/genética , Piruvato Deshidrogenasa Quinasa Acetil-Transferidora/genética , Factor de Transcripción Sp1/genética
3.
Toxicol Appl Pharmacol ; 433: 115774, 2021 12 15.
Artículo en Inglés | MEDLINE | ID: mdl-34699867

RESUMEN

Supplements containing pharmacological concentrations of biotin are commercially available. The mechanisms by which biotin at pharmacological concentrations exerts its action have been the subject of multiple investigations, particularly for biotin's medicinal potential and wide use for cosmetic purposes. Several studies have reported that biotin supplementation increases cell proliferation; however, the mechanisms involved in this effect have not yet been characterized. In a previous study, we found that a biotin-supplemented diet increased spermatogonia proliferation. The present study was focused on investigating the molecular mechanisms involved in biotin-induced testis cell proliferation. Male BALB/cAnNHsd mice were fed a control or a biotin-supplemented diet (1.76 or 97.7 mg biotin/kg diet) for eight weeks. Compared with the control group, the biotin-supplemented mice presented augmented protein abundance of the c-kit-receptor and pERK1/2Tyr204 and pAKTSer473, the active forms of ERK/AKT proliferation signaling pathways. No changes were observed in the testis expression of the stem cell factor and in the serum levels of the follicle-stimulating hormone. Analysis of mRNA abundance found an increase in cyclins Ccnd3, Ccne1, Ccna2; Kinases Cdk4, Cdk2; and E2F; and Sp1 & Sp3 transcription factors. Decreased expression of cyclin-dependent kinase inhibitor 1a (p21) was observed but not of Cdkn2a inhibitor (p16). The results of the present study identifies, for the first time, the mechanisms associated with biotin supplementation-induced cell proliferation, which raises concerns about the effects of biotin on male reproductive health because of its capacity to cause hyperplasia, especially because this vitamin is available in large amounts without regulation.


Asunto(s)
Biotina/toxicidad , Proliferación Celular/efectos de los fármacos , Suplementos Dietéticos/toxicidad , Hormona Folículo Estimulante/sangre , Espermatogonias/efectos de los fármacos , Factor de Células Madre/metabolismo , Testículo/efectos de los fármacos , Animales , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Masculino , Ratones Endogámicos BALB C , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Fosforilación , Proteínas Proto-Oncogénicas c-akt/metabolismo , Proteínas Proto-Oncogénicas c-kit/genética , Proteínas Proto-Oncogénicas c-kit/metabolismo , Transducción de Señal , Factor de Transcripción Sp1/genética , Factor de Transcripción Sp1/metabolismo , Factor de Transcripción Sp3/genética , Factor de Transcripción Sp3/metabolismo , Espermatogonias/metabolismo , Espermatogonias/patología , Testículo/metabolismo , Testículo/patología
4.
Toxicol Appl Pharmacol ; 425: 115606, 2021 08 15.
Artículo en Inglés | MEDLINE | ID: mdl-34087332

RESUMEN

Triptolide (TP), a primary bioactive ingredient isolated from the traditional Chinese herbal medicine Tripterygium wilfordii Hook. F. (TWHF), has attracted great interest for its therapeutic biological activities in inflammation and autoimmune disease. However, its clinical use is limited by severe testicular toxicity, and the underlying mechanism has not been elucidated. Our preliminary evidence demonstrated that TP disrupted glucose metabolism and caused testicular toxicity. During spermatogenesis, Sertoli cells (SCs) provide lactate as an energy source to germ cells by glycolysis. The transcription factors GATA-binding protein 4 (GATA4) and specificity protein 1 (Sp1) can regulate glycolysis. Based on this evidence, we speculate that TP causes abnormal glycolysis in SCs by influencing the expression of the transcription factors GATA4 and Sp1. The mechanism of TP-induced testicular toxicity was investigated in vitro and in vivo. The data indicated that TP decreased glucose consumption, lactate production, and the mRNA levels of glycolysis-related transporters and enzymes. TP also downregulated the protein expression of the transcription factors GATA4 and Sp1, as well as the glycolytic enzyme phosphofructokinase platelet (PFKP). Phosphorylated GATA4 and nuclear GATA4 protein levels were reduced in a dose- and time-dependent manner after TP incubation. Similar effects were observed in shGata4-treated TM4 cells and BALB/c mice administered 0.4 mg/kg TP for 28 days, and glycolysis was also inhibited. Gata4 knockdown downregulated Sp1 and PFKP expression. Furthermore, the Sp1 inhibitor plicamycin inhibited PFKP protein levels in TM4 cells. In conclusion, TP inhibited GATA4-mediated glycolysis by suppressing Sp1-dependent PFKP expression in SCs and caused testicular toxicity.


Asunto(s)
Diterpenos/farmacología , Factor de Transcripción GATA4/metabolismo , Glucólisis/efectos de los fármacos , Fenantrenos/farmacología , Fosfofructoquinasa-1 Tipo C/metabolismo , Células de Sertoli/efectos de los fármacos , Factor de Transcripción Sp1/metabolismo , Animales , Línea Celular , Proliferación Celular , Supervivencia Celular/efectos de los fármacos , Regulación hacia Abajo , Compuestos Epoxi/farmacología , Factor de Transcripción GATA4/efectos de los fármacos , Factor de Transcripción GATA4/genética , Regulación de la Expresión Génica/efectos de los fármacos , Células HEK293 , Humanos , Masculino , Ratones , Ratones Endogámicos ICR , Fosfofructoquinasa-1 Tipo C/efectos de los fármacos , Fosfofructoquinasa-1 Tipo C/genética , Células de Sertoli/metabolismo , Transducción de Señal/efectos de los fármacos , Factor de Transcripción Sp1/efectos de los fármacos , Factor de Transcripción Sp1/genética
5.
Carcinogenesis ; 42(3): 344-356, 2021 04 17.
Artículo en Inglés | MEDLINE | ID: mdl-33146712

RESUMEN

Recently, we identified that the atypical protein kinase C isoform ι (PKCι) enhances the expression of Yes-associated protein 1 (YAP1) to promote the tumorigenesis of pancreatic adenocarcinoma harboring mutant KRAS (mu-KRAS). To advance our understanding about underlying mechanisms, we analyze the transcription of YAP1 in pancreatic cancer cells and reveal that transcription factor specificity protein 1 (Sp1) is upregulated by PKCι and subsequently binds to multiple sites in YAP1 promoter to drive the transactivation of YAP1 in pancreatic cancer cells carrying mu-KRAS. The bioinformatics analysis further substantiates that the expression of PKCι, Sp1 and YAP1 is correlated and associated with the stages and prognosis of pancreatic tumors. Moreover, our apoptotic detection data demonstrate that combination of PKCι and Sp1 inhibitors at subtoxic doses displays synergistic effects on inducing apoptosis and reversing the immunosuppression of pancreatic cancer cells, establishing the combination of PKCι and Sp1 inhibitors as a promising novel therapeutic approach, or an adjuvant strategy to potentiate the antitumor effects of other immunotherapeutic agents in pancreatic cancer treatment.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/genética , Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Isoenzimas/metabolismo , Neoplasias Pancreáticas/genética , Proteína Quinasa C/metabolismo , Factor de Transcripción Sp1/genética , Factores de Transcripción/genética , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Apoptosis/efectos de los fármacos , Apoptosis/genética , Apoptosis/inmunología , Carcinogénesis/efectos de los fármacos , Carcinogénesis/genética , Carcinogénesis/inmunología , Línea Celular Tumoral , Biología Computacional , Conjuntos de Datos como Asunto , Sinergismo Farmacológico , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica/inmunología , Humanos , Isoenzimas/antagonistas & inhibidores , Mutación , Páncreas/inmunología , Páncreas/patología , Neoplasias Pancreáticas/tratamiento farmacológico , Neoplasias Pancreáticas/inmunología , Neoplasias Pancreáticas/patología , Pronóstico , Regiones Promotoras Genéticas/genética , Proteína Quinasa C/antagonistas & inhibidores , Proteínas Proto-Oncogénicas p21(ras)/genética , RNA-Seq , Factor de Transcripción Sp1/antagonistas & inhibidores , Factor de Transcripción Sp1/metabolismo , Activación Transcripcional/efectos de los fármacos , Activación Transcripcional/inmunología , Escape del Tumor/efectos de los fármacos , Escape del Tumor/genética , Regulación hacia Arriba/efectos de los fármacos , Regulación hacia Arriba/inmunología , Proteínas Señalizadoras YAP
6.
Diab Vasc Dis Res ; 17(1): 1479164119878427, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-31726874

RESUMEN

Thiamine prevents high glucose-induced damage in microvasculature, and progression of retinopathy and nephropathy in diabetic animals. Impaired thiamine availability causes renal damage in diabetic patients. Two single-nucleotide polymorphisms in SLC19A3 locus encoding for thiamine transporter 2 are associated with absent/minimal diabetic retinopathy and nephropathy despite long-term type 1 diabetes. We investigated the involvement of thiamine transporter 1 and thiamine transporter 2, and their transcription factor specificity protein 1, in high glucose-induced damage and altered thiamine availability in cells of the inner blood-retinal barrier. Human endothelial cells, pericytes and Müller cells were exposed to hyperglycaemic-like conditions and/or thiamine deficiency/over-supplementation in single/co-cultures. Expression and localization of thiamine transporter 1, thiamine transporter 2 and transcription factor specificity protein 1 were evaluated together with intracellular thiamine concentration, transketolase activity and permeability to thiamine. The effects of thiamine depletion on cell function (viability, apoptosis and migration) were also addressed. Thiamine transporter 2 and transcription factor specificity protein 1 expression were modulated by hyperglycaemic-like conditions. Transketolase activity, intracellular thiamine and permeability to thiamine were decreased in cells cultured in thiamine deficiency, and in pericytes in hyperglycaemic-like conditions. Thiamine depletion reduced cell viability and proliferation, while thiamine over-supplementation compensated for thiamine transporter 2 reduction by restoring thiamine uptake and transketolase activity. High glucose and reduced thiamine determine impairment in thiamine transport inside retinal cells and through the inner blood-retinal barrier. Thiamine transporter 2 modulation in our cell models suggests its major role in thiamine transport in retinal cells and its involvement in high glucose-induced damage and impaired thiamine availability.


Asunto(s)
Retinopatía Diabética/metabolismo , Células Endoteliales/efectos de los fármacos , Células Ependimogliales/efectos de los fármacos , Glucosa/toxicidad , Proteínas de Transporte de Membrana/metabolismo , Pericitos/efectos de los fármacos , Vasos Retinianos/efectos de los fármacos , Tiamina/metabolismo , Apoptosis/efectos de los fármacos , Línea Celular , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Microambiente Celular , Técnicas de Cocultivo , Retinopatía Diabética/genética , Retinopatía Diabética/patología , Células Endoteliales/metabolismo , Células Endoteliales/patología , Células Ependimogliales/metabolismo , Células Ependimogliales/patología , Humanos , Proteínas de Transporte de Membrana/genética , Pericitos/metabolismo , Pericitos/patología , Vasos Retinianos/metabolismo , Vasos Retinianos/patología , Factor de Transcripción Sp1/genética , Factor de Transcripción Sp1/metabolismo , Transcetolasa/metabolismo
7.
J Hypertens ; 37(5): 964-971, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-30335670

RESUMEN

OBJECTIVE: Melatonin, an endogenous neurohormone secreted predominately by the pineal gland, has a variety of physiological functions. However, its protective role in atherosclerosis is not clear. In this study, we sought to investigate the potential effects of melatonin in modulating atherosclerotic plaque stability in apolipoprotein E knockout (ApoE) mice. METHOD AND RESULTS: Smooth muscle cells were treated with melatonin, which significantly increased mRNA and protein levels of a key intracellular enzyme essential for collagen maturation and secretion, prolyl-4-hydroxylase α1 (P4Hα1). Mechanistically, melatonin increased Akt phosphorylation and transcriptional activation of specificity protein 1 (Sp1), which bound with the P4Hα1 promoter and then induced P4Hα1 expression. Pretreatment with either Akt inhibitor LY294002 or Sp1 inhibitor mithramycin A (MTM) could inhibit melatonin-induced P4Hα1 expression. Finally, atherosclerotic lesions were induced by placing a perivascular collar on the right common carotid artery of ApoE mice, which were received with or without different doses of melatonin or MTM. High-dose melatonin enhanced atherosclerotic plaque stability in ApoE mice in vivo by inducing the expression of P4Hα1, which was reversed by MTM. CONCLUSION: We propose that melatonin supplementation may provide a novel and promising approach to atherosclerosis treatment.


Asunto(s)
Apolipoproteínas E/genética , Depresores del Sistema Nervioso Central/farmacología , Expresión Génica/efectos de los fármacos , Melatonina/farmacología , Miocitos del Músculo Liso/metabolismo , Procolágeno-Prolina Dioxigenasa/genética , Animales , Línea Celular , Depresores del Sistema Nervioso Central/administración & dosificación , Cromonas/farmacología , Inhibidores Enzimáticos/farmacología , Masculino , Melatonina/administración & dosificación , Ratones , Ratones Noqueados , Morfolinas/farmacología , Fosforilación/efectos de los fármacos , Placa Aterosclerótica/tratamiento farmacológico , Plicamicina/análogos & derivados , Plicamicina/farmacología , Proteínas Proto-Oncogénicas c-akt/metabolismo , ARN Mensajero/metabolismo , Factor de Transcripción Sp1/antagonistas & inhibidores , Factor de Transcripción Sp1/genética
8.
Anticancer Drugs ; 30(2): 153-158, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-30362980

RESUMEN

Colorectal cancer occurs throughout the world but is most common in developed countries. Cancer progression is believed to be driven by genetic mutations in this complex condition. Risk factors for developing colorectal cancer include a genetic family history, long-term ulcerative colitis, and colonic polyps. The use of baicalin has been reported to be clinically efficacious against colon tumors in Asian countries despite an unclear mechanism of action. Several cancers have been found to be biologically dependent on the specificity protein 1 (sp1) transcription factor family. We hypothesized that baicalin may exert its chemotherapeutic effects by sp1 downregulation. Using the SW480 human colorectal cancer cell line, we investigated the physiological properties of baicalin. Our experiments were designed toward clarifying three goals: (a) to determine the mRNA expression profile of transcription factors in colorectal cancer patients using a microarray-based analysis; (b) to determine the effects of baicalin on the sp1 transcription factor with western blotting and reporter cell assays; and (c) to contrast the effects of mithramycin-A (an sp1 transcription factor inhibitor) and baicalin using western blotting and reporter cell assays. Both baicalin and mithramycin-A downregulated sp1 expression, attenuated SW480 cell proliferation, and increased cell apoptosis. Baicalin inhibited sp1 expression and led to SW480 apoptosis, thus clarifying the effect of this traditional Chinese medicine compound in the treatment of colon cancer.


Asunto(s)
Antiinflamatorios no Esteroideos/farmacología , Apoptosis , Biomarcadores de Tumor/metabolismo , Neoplasias del Colon/patología , Flavonoides/farmacología , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Factor de Transcripción Sp1/antagonistas & inhibidores , Biomarcadores de Tumor/genética , Proliferación Celular , Neoplasias del Colon/tratamiento farmacológico , Neoplasias del Colon/genética , Neoplasias del Colon/metabolismo , Perfilación de la Expresión Génica , Humanos , Factor de Transcripción Sp1/genética , Factor de Transcripción Sp1/metabolismo , Células Tumorales Cultivadas
9.
Am J Chin Med ; 46(5): 1093-1110, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29976085

RESUMEN

The mechanism of ethanol-induced hepatotoxicity was complicated, accompanied by the over-expressions of the cytochrome P450 2E1 (CYP2E1), heat shock protein 70 (Hsp70) and the nuclear factor specificity protein 1 (SP1). Kaempferol (Kaem) could protect the ethanol-induced hepatotoxicity likely by inhibiting the CYP2E1 expression and activity. This study investigated the protective mechanism(s) of kaempferol on ethanol-induced toxicity by dynamic alteration of SP1, Hsp70 and CYP2E1 among the nucleus and different organelles in hepatocytes. After ethanol treatment alone and co-incubation hepatocytes with kaempferol, protein levels of CYP2E1, Hsp70, and SP1 were determined in vitro (western blotting and immunofluorescence). Hepatocytes' viability was assessed by terminal deoxynucleotidyl transferase-mediated dUTP nick end labeling (TUNEL) methods. Glutathione (GSH) levels were evaluated for ethanol-induced oxidative stress. In the ethanol-treated hepatocytes, kaempferol decreased protein levels of CYP2E1 in both microsome and mitochondria, cytosolic Hsp70 and SP1 in nuclear and cytosol, and the oxidative stress and increased the cell viability compared to those of ethanol group. Collectively, our findings propose that the protective mechanism of kaempferol is involved in the synchronous, early and persistent inhibitions of mitochondrial and microsomal CYP2E1, cytosolic Hsp70 and nuclear and cytosolic SP1 in mouse primary hepatocytes' injury induced by ethanol.


Asunto(s)
Citocromo P-450 CYP2E1/metabolismo , Etanol/efectos adversos , Proteínas HSP70 de Choque Térmico/metabolismo , Hepatocitos/efectos de los fármacos , Quempferoles/farmacología , Hepatopatías Alcohólicas/metabolismo , Hígado/efectos de los fármacos , Sustancias Protectoras/farmacología , Factor de Transcripción Sp1/metabolismo , Animales , Células Cultivadas , Citocromo P-450 CYP2E1/genética , Glutatión/metabolismo , Proteínas HSP70 de Choque Térmico/genética , Hepatocitos/metabolismo , Humanos , Hígado/lesiones , Hígado/metabolismo , Hepatopatías Alcohólicas/genética , Hepatopatías Alcohólicas/prevención & control , Masculino , Ratones , Estrés Oxidativo/efectos de los fármacos , Factor de Transcripción Sp1/genética
10.
Am J Chin Med ; 46(5): 1021-1044, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29986596

RESUMEN

An extract of Dendropanax morbifera branch exerts antioxidant, anti-inflammatory, antithrombotic, and anticancer activities. The purpose of this study was to investigate the effect of the extract in isoproterenol-induced cardiac hypertrophy. Phalloidin staining showed that treatment with the extract dramatically prevents isoproterenol-induced H9c2 cell enlargement and the expression of cardiac hypertrophic marker genes, including atrial natriuretic peptide (ANP) and B-type brain natriuretic peptide (BNP). Further, pretreatment with the extract decreased isoproterenol-induced GATA4 and Sp1 expression in H9c2 cells. Overexpression of Sp1 induced the expression of GATA4. The forced expression of Sp1 or its downstream target GATA4, as well as the co-transfection of Sp1 and GATA4 increased the expression of ANP, which was decreased by treatment with the extract. To further elucidate the regulation of the Sp1/GATA4-mediated expression of ANP, knockdown experiments were performed. Transfection with small interfering RNAs (siRNAs) for Sp1 or GATA4 decreased ANP expression. The extract did not further inhibit the expression of ANP reduced by the transfection of GATA4 siRNA. Sp1 knockdown did not affect the expression of ANP that was induced by the overexpression of GATA4; however, GATA4 knockdown abolished the expression of ANP that had been induced by Sp1 overexpression. The extract treatment also attenuated the isoproterenol-induced activation of p38 MAPK, ERK1/2, and JNK1. Hesperidin, catechin, 2,5-dihydroxybenzoic acid, and salicylic acid are the main phenolic compounds present in the extract as observed by high performance liquid chromatography. Hesperidin and 2,5-dihydroxybenzoic acid attenuated isoproterenol-induced cardiac hypertrophy. These findings suggest that the D. morbifera branch extract prevents cardiac hypertrophy by downregulating the activation of Sp1/GATA4 and MAPK signaling pathways.


Asunto(s)
Araliaceae/química , Cardiomegalia/metabolismo , Factor de Transcripción GATA4/metabolismo , Miocitos Cardíacos/efectos de los fármacos , Extractos Vegetales/farmacología , Factor de Transcripción Sp1/metabolismo , Animales , Cardiomegalia/tratamiento farmacológico , Cardiomegalia/genética , Línea Celular , Regulación hacia Abajo/efectos de los fármacos , Factor de Transcripción GATA4/genética , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Miocitos Cardíacos/metabolismo , Ratas , Transducción de Señal/efectos de los fármacos , Factor de Transcripción Sp1/genética
11.
Sci Rep ; 7(1): 5557, 2017 07 17.
Artículo en Inglés | MEDLINE | ID: mdl-28717181

RESUMEN

Deciphering gene regulatory networks requires identification of gene expression modules. We describe a novel bottom-up approach to identify gene modules regulated by cis-regulatory motifs from a human gene co-expression network. Target genes of a cis-regulatory motif were identified from the network via the motif's enrichment or biased distribution towards transcription start sites in the promoters of co-expressed genes. A gene sub-network containing the target genes was extracted and used to derive gene modules. The analysis revealed known and novel gene modules regulated by the NF-Y motif. The binding of NF-Y proteins to these modules' gene promoters were verified using ENCODE ChIP-Seq data. The analyses also identified 8,048 Sp1 motif target genes, interestingly many of which were not detected by ENCODE ChIP-Seq. These target genes assemble into house-keeping, tissues-specific developmental, and immune response modules. Integration of Sp1 modules with genomic and epigenomic data indicates epigenetic control of Sp1 targets' expression in a cell/tissue specific manner. Finally, known and novel target genes and modules regulated by the YY1, RFX1, IRF1, and 34 other motifs were also identified. The study described here provides a valuable resource to understand transcriptional regulation of various human developmental, disease, or immunity pathways.


Asunto(s)
Epigénesis Genética , Redes Reguladoras de Genes , Regiones Promotoras Genéticas , Factor de Unión a CCAAT/genética , Regulación de la Expresión Génica , Humanos , Factor 1 Regulador del Interferón/genética , Lisina/metabolismo , Metilación , Motivos de Nucleótidos , Análisis de Secuencia por Matrices de Oligonucleótidos , Factor Regulador X1/genética , Factor de Transcripción Sp1/genética , Factor de Transcripción YY1/genética
12.
Eur J Nutr ; 56(5): 1899-1909, 2017 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-27250629

RESUMEN

PURPOSE: In this study, we sought to investigate the effects of maternal betaine supplementation on the expression and regulation of GALK1 gene in the liver of neonatal piglets. METHODS: Sixteen sows of two groups were fed control or betaine-supplemented diets (3 g/kg), respectively, throughout the pregnancy. Newborn piglets were individually weighed immediately after birth, and one male piglet close to mean body weight from the same litter was selected and killed before suckling. Serum samples of newborn piglets were analyzed for biochemical indexes, hormone and amino acid levels. Liver samples were analyzed for GALK1 expression by real-time PCR and western blotting, while GALK1 regulational mechanism was analyzed by methylated DNA immunoprecipitation, chromatin immunoprecipitation and microRNAs expression. RESULTS: Betaine-exposed neonatal piglets had lower serum concentration of galactose, which was associated with significantly down-regulated hepatic GALK1 expression. The repression of GALK1 mRNA expression was associated with DNA hypermethylation and more enriched repression histone mark H3K27me3 on its promoter. Binding sites of SP1, GR and STAT3 were predicted on GALK1 promoter, and decreased SP1 protein content and lower SP1 binding to GALK1 promoter were detected in the liver of betaine-exposed piglets. Furthermore, the expression of miRNA-149 targeting GALK1 was up-regulated in the liver of betaine-exposed piglets, along with elevated miRNAs-processing enzymes Dicer and Ago2. CONCLUSIONS: Our results suggest that maternal dietary betaine supplementation during gestation suppresses GALK1 expression in the liver of neonatal piglets, which involves complex gene regulation mechanisms including DNA methylation, histone modification, miRNAs expression and SP1-mediated transcriptional modulation.


Asunto(s)
Betaína/administración & dosificación , Represión Epigenética , Galactoquinasa/genética , Factor de Transcripción Sp1/metabolismo , Aminoácidos/sangre , Animales , Animales Recién Nacidos , Betaína/sangre , Biomarcadores/sangre , Inmunoprecipitación de Cromatina , Metilación de ADN/efectos de los fármacos , Dieta , Suplementos Dietéticos , Femenino , Galactoquinasa/metabolismo , Galactosa/metabolismo , Regulación de la Expresión Génica , Insulina/sangre , Hígado/metabolismo , Masculino , MicroARNs/genética , MicroARNs/metabolismo , Tamaño de los Órganos/efectos de los fármacos , Regiones Promotoras Genéticas , Reproducibilidad de los Resultados , Factor de Transcripción Sp1/genética , Porcinos
13.
Acta Pharmacol Sin ; 37(12): 1623-1640, 2016 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-27694907

RESUMEN

AIM: Physcion is a major bioactive ingredient in the traditional Chinese medicine Radix et Rhizoma Rhei, which has an anthraquinone chemical structure and exhibits a variety of pharmacological activities including laxative, hepatoprotective, anti-inflammatory, anti-microbial and anti-proliferative effects. In this study we investigated the effect of physcion on human nasopharyngeal carcinoma in vitro and in vivo, as well as the mechanisms underlying the anti-tumor action. METHODS: The nasopharyngeal carcinoma cell line CNE2 was treated with physcion, and cell viability was detected using MTT and colony formation assays. Flow cytometry was used to assess the cell cycle arrest, mitochondrial membrane potential loss, apoptosis, autophagy and intracellular ROS generation. Apoptotic cell death was also confirmed by a TUNEL assay. The expression of target or marker molecules was determined using Western blotting. The activity of caspase-3, 8, and 9 was detected with an ELISA kit. A xenograft murine model was used to evaluate the in vivo anti-tumor action of physcion, the mice were administered physcion (10, 20 mg·kg-1·d-1, ip) for 30 d. RESULTS: Treatment with physcion (5, 10, and 20 µmol/L) dose-dependently suppressed the cell viability and colony formation in CNE2 cells. Physcion (10 and 20 µmol/L) dose-dependently blocked cell cycle progression at G1 phase and induced both caspase-dependent apoptosis and autophagy in CNE2 cells. Furthermore, physcion treatment induced excessive ROS generation in CNE2 cells, and subsequently disrupted the miR-27a/ZBTB10 axis, resulting in repression of the transcription factor Sp1 that was involved in physcion-induced apoptosis and autophagy. Moreover, physcion-induced autophagy acted as a pro-apoptotic factor, and possibly contributed to physcion-induced apoptosis. In the xenograft murine model, administration of physcion dose-dependently suppressed the tumor growth without affecting the body weight. Furthermore, the anti-tumor effects of physcion were correlated with downregulation of Sp1 and suppression of miR-27a in the tumor tissues. CONCLUSION: Physcion induces apoptosis and autophagy in human nasopharyngeal carcinoma by targeting Sp1, which was mediated by ROS/miR-27a/ZBTB10 signaling. The results suggest that physcion is a promising candidate for the treatment of human nasopharyngeal carcinoma.


Asunto(s)
Antineoplásicos Fitogénicos/farmacología , Apoptosis/efectos de los fármacos , Autofagia/efectos de los fármacos , Emodina/análogos & derivados , Neoplasias Nasofaríngeas/tratamiento farmacológico , Animales , Antineoplásicos Fitogénicos/uso terapéutico , Línea Celular Tumoral , Emodina/farmacología , Emodina/uso terapéutico , Xenoinjertos , Humanos , Ratones , Neoplasias Nasofaríngeas/metabolismo , Trasplante de Neoplasias , Especies Reactivas de Oxígeno/metabolismo , Factor de Transcripción Sp1/genética , Factor de Transcripción Sp1/metabolismo
14.
Biochem J ; 473(13): 1967-76, 2016 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-27143785

RESUMEN

Dystrophin Dp71, the smallest product encoded by the Duchenne muscular dystrophy gene, is ubiquitously expressed in all non-muscle cells. Although Dp71 is involved in various cellular processes, the mechanisms underlying its expression have been little studied. In hepatic cells, Dp71 expression is down-regulated by the xenobiotic ß-naphthoflavone. However, the effectors of this regulation remain unknown. In the present study we aimed at identifying DNA elements and transcription factors involved in Dp71 expression in hepatic cells. Relevant DNA elements on the Dp71 promoter were identified by comparing Dp71 5'-end flanking regions between species. The functionality of these elements was demonstrated by site-directed mutagenesis. Using EMSAs and ChIP, we showed that the Sp1 (specificity protein 1), Sp3 (specificity protein 3) and YY1 (Yin and Yang 1) transcription factors bind to the Dp71 promoter region. Knockdown of Sp1, Sp3 and YY1 in hepatic cells increased endogenous Dp71 expression, but reduced Dp71 promoter activity. In summary, Dp71 expression in hepatic cells is carried out, in part, by YY1-, Sp1- and Sp3-mediated transcription from the Dp71 promoter.


Asunto(s)
Distrofina/metabolismo , Hepatocitos/metabolismo , Factor de Transcripción Sp1/metabolismo , Factor de Transcripción Sp3/metabolismo , Factor de Transcripción YY1/metabolismo , Animales , Secuencia de Bases , Línea Celular , Inmunoprecipitación de Cromatina , Distrofina/genética , Humanos , Ratones , Datos de Secuencia Molecular , Mutagénesis Sitio-Dirigida , Regiones Promotoras Genéticas/genética , Unión Proteica , Interferencia de ARN , Reacción en Cadena en Tiempo Real de la Polimerasa , Factor de Transcripción Sp1/genética , Factor de Transcripción Sp3/genética , Transfección , Factor de Transcripción YY1/genética
15.
Am J Chin Med ; 44(1): 177-95, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-26916922

RESUMEN

High mortality and morbidity rates for hepatocellular carcinoma (HCC) in Taiwan primarily result from uncontrolled tumor metastasis. In our previous studies, we have reported that Dioscorea nipponica Makino extract (DNE) has anti-metastasis effects on human oral cancer cells. However, the effect of DNE on hepatoma metastasis have not been thoroughly investigated and remains poorly understood. To determine the effects of DNE on the migration and invasion in HCC cells we used a wound healing model, Boyden chamber assays, gelatin/casein zymography and Western blotting. Transcriptional levels of matrix metalloproteinase-9 (MMP-9) and urokinase-type plasminogen activator (u-PA) were detected by real-time PCR and promoter assays. In this study, DNE treatment significantly inhibited the migration/invasion capacities of Huh7 cell lines. The results of gelatin/casein zymography and Western blotting revealed that the activities and protein levels of the MMP-9 and u-PA were inhibited by DNE. Tests of the mRNA levels, real-time PCR, and promoter assays evaluated the inhibitory effects of DNE on u-PA expression in human hepatoma cells. A chromatin immunoprecipitation (ChIP) assay showed not only that DNE inhibits u-PA expression, but also the inhibitory effects were associated with the down-regulation of the transcription factors of NF-[Formula: see text]B and SP-1 signaling pathways. Western blot analysis also showed that DNE inhibits PI3K and phosphorylation of Akt. In conclusion, these results show that u-PA expression may be a potent therapeutic target in the DNE-mediated suppression of HCC invasion/migration. DNE may have potential use as a chemo-preventive agent against liver cancer metastasis.


Asunto(s)
Carcinoma Hepatocelular/genética , Carcinoma Hepatocelular/patología , Dioscorea/química , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/patología , FN-kappa B/antagonistas & inhibidores , Inhibidores de las Quinasa Fosfoinosítidos-3 , Extractos Vegetales/farmacología , Proteínas Proto-Oncogénicas c-akt/metabolismo , Factor de Transcripción Sp1/antagonistas & inhibidores , Transcripción Genética/efectos de los fármacos , Transcripción Genética/genética , Activador de Plasminógeno de Tipo Uroquinasa/antagonistas & inhibidores , Carcinoma Hepatocelular/tratamiento farmacológico , Carcinoma Hepatocelular/prevención & control , Línea Celular Tumoral , Movimiento Celular , Expresión Génica/efectos de los fármacos , Humanos , Neoplasias Hepáticas/tratamiento farmacológico , Neoplasias Hepáticas/prevención & control , Metaloproteinasa 9 de la Matriz/genética , Metaloproteinasa 9 de la Matriz/metabolismo , FN-kappa B/genética , Invasividad Neoplásica , Metástasis de la Neoplasia , Fosforilación/efectos de los fármacos , Fosforilación/genética , Fitoterapia , Extractos Vegetales/uso terapéutico , Factor de Transcripción Sp1/genética , Activador de Plasminógeno de Tipo Uroquinasa/genética , Activador de Plasminógeno de Tipo Uroquinasa/metabolismo
16.
Dev Dyn ; 245(3): 379-87, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26384464

RESUMEN

BACKGROUND: Mesp1 is critical for early cardiomyocyte differentiation and heart development. We previously observed down-regulation of Mesp1 expression in YY1-ablated mouse embryonic hearts. However, how Mesp1 expression is mediated by YY1 is not well understood. RESULTS: We excised YY1 in the murine embryos using Sox2-cre and found that Mesp1 was down-regulated in the embryonic day (E) 7.5 mutant embryos. Also, YY1 activated the 6 kb Mesp1 regulatory element fused to a luciferase reporter. We identified two putative YY1 binding sites in the proximal promoter region of Mesp1 gene, and found that mutation of these sites significantly reduced YY1-induced activation of the Mesp1 promoter. We also uncovered one cognitive site for SP1, one of the earliest binding partners of YY1 identified. Mutation of this SP1 site repressed SP1-induced activation of the Mesp1 promoter. Moreover, YY1 and SP1 synergistically activated the Mesp1 promoter. Consistently, while Lacz expression driven by the wild-type 6 kb regulatory element of Mesp1 gene was robust in E7.5 mouse embryos, the mutation of these binding sites in the context of this 6 kb sequence substantially reduced the LacZ expression during embryogenesis. CONCLUSIONS: YY1 and SP1 independently and cooperatively govern the Mesp1 expression during embryogenesis.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/biosíntesis , Embrión de Mamíferos/embriología , Regulación del Desarrollo de la Expresión Génica/fisiología , Corazón/embriología , Organogénesis/fisiología , Elementos de Respuesta/fisiología , Factor de Transcripción Sp1/metabolismo , Factor de Transcripción YY1/metabolismo , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Ratones , Factor de Transcripción Sp1/genética , Factor de Transcripción YY1/genética
17.
Ann Rheum Dis ; 75(3): 601-8, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25596157

RESUMEN

OBJECTIVE: MicroRNAs (miRNAs) are small endogenous, non-coding RNAs that act as post-transcriptional regulators. We analysed the in vivo effect of miRNA-124 (miR-124, the rat analogue of human miR-124a) on adjuvant-induced arthritis (AIA) in rats. METHODS: AIA was induced in Lewis rats by injecting incomplete Freund's adjuvant with heat-killed Mycobacterium tuberculosis. Precursor (pre)-miR-124 was injected into the right hind ankle on day 9. Morphological changes in the ankle joint were assessed by micro-CT and histopathology. Cytokine expression was examined by western blotting and real-time RT-PCR. The effect of miR-124 on predicted target messenger RNAs (mRNAs) was examined by luciferase reporter assays. The effect of pre-miR-124 or pre-miR-124a on the differentiation of human osteoclasts was examined by tartrate-resistant acid phosphatase staining. RESULTS: We found that miR-124 suppressed AIA in rats, as demonstrated by decreased synoviocyte proliferation, leucocyte infiltration and cartilage or bone destruction. Osteoclast counts and expression level of receptor activator of the nuclear factor κB ligand (RANKL), integrin ß1 (ITGB1) and nuclear factor of activated T cells cytoplasmic 1 (NFATc1) were reduced in AIA rats treated with pre-miR-124. Luciferase analysis showed that miR-124 directly targeted the 3'UTR of the rat NFATc1, ITGB1, specificity protein 1 and CCAAT/enhancer-binding protein α mRNAs. Pre-miR-124 also suppressed NFATc1 expression in RAW264.7 cells. Both miR-124 and miR-124a directly targeted the 3'-UTR of human NFATc1 mRNA, and both pre-miR-124 and pre-miR-124a suppressed the differentiation of human osteoclasts. CONCLUSIONS: We found that miR-124 ameliorated AIA by suppressing critical prerequisites for arthritis development, such as RANKL and NFATc1. Thus, miR-124a is a candidate for therapeutic use for human rheumatoid arthritis.


Asunto(s)
Artritis Experimental/genética , Diferenciación Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , MicroARNs/farmacología , Osteoclastos/efectos de los fármacos , ARN Mensajero/efectos de los fármacos , Animales , Proteína alfa Potenciadora de Unión a CCAAT/efectos de los fármacos , Proteína alfa Potenciadora de Unión a CCAAT/genética , Progresión de la Enfermedad , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Integrina beta1/efectos de los fármacos , Integrina beta1/genética , Ligando RANK/efectos de los fármacos , Ligando RANK/genética , ARN Mensajero/metabolismo , Ratas , Ratas Endogámicas Lew , Factor de Transcripción Sp1/efectos de los fármacos , Factor de Transcripción Sp1/genética , Membrana Sinovial/citología , Factores de Transcripción/efectos de los fármacos , Factores de Transcripción/genética
18.
J Exp Clin Cancer Res ; 34: 154, 2015 Dec 21.
Artículo en Inglés | MEDLINE | ID: mdl-26689593

RESUMEN

BACKGROUND: Lung cancer is the most common cause of cancer-related deaths worldwide. Natural phytochemicals from traditional medicinal plants such as solamargine have been shown to have anticancer properties. The prostaglandin E2 receptor EP4 is highly expressed in human cancer, however, the functional role of EP4 in the occurrence and progression of non small cell lung cancer (NSCLC) remained to be elucidated. METHODS: Cell viability was measured by MTT assays. Western blot was performed to measure the phosphorylation and protein expression of PI3-K downstream effector Akt, transcription factors SP1, p65, and EP4. Quantitative real-time PCR (qRT-PCR) was used to examine the mRNA levels of EP4 gene. Exogenous expression of SP1, p65, and EP4 genes was carried out by transient transfection assays. EP4 promoter activity was measured by Dual Luciferase Reporter Kit. RESULTS: We showed that solamargine inhibited the growth of lung cancer cells. Mechanistically, we found that solamargine decreased the phosphorylation of Akt, the protein, mRNA expression, and promoter activity of EP4. Moreover, solamargine inhibited protein expression of SP1 and NF-κB subunit p65, all of which were abrogated in cells transfected with exogenous expressed Akt. Intriguingly, exogenous expressed SP1 overcame the effect of solamargine on inhibition of p65 protein expression, and EP4 protein expression and promoter activity. Finally, exogenous expressed EP4 feedback reversed the effect of solamargine on phosphorylation of Akt and cell growth inhibition. CONCLUSION: Our results show that solamargine inhibits the growth of human lung cancer cells through inactivation of Akt signaling, followed by reduction of SP1 and p65 protein expression. This results in the inhibition of EP4 gene expression. The cross-talk between SP1 and p65, and the positive feedback regulatory loop of PI3-K/Akt signaling by EP4 contribute to the overall responses of solamargine in this process. This study unveils a novel mechanism by which solamargine inhibits growth of human lung cancer cells.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas/tratamiento farmacológico , Proteína Oncogénica v-akt/biosíntesis , Subtipo EP4 de Receptores de Prostaglandina E/biosíntesis , Alcaloides Solanáceos/administración & dosificación , Factor de Transcripción Sp1/biosíntesis , Factor de Transcripción ReIA/biosíntesis , Carcinoma de Pulmón de Células no Pequeñas/genética , Carcinoma de Pulmón de Células no Pequeñas/patología , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Proteína Oncogénica v-akt/genética , Fosfatidilinositol 3-Quinasas/biosíntesis , Fosfatidilinositol 3-Quinasas/genética , Fosforilación/efectos de los fármacos , Regiones Promotoras Genéticas , ARN Mensajero/biosíntesis , Subtipo EP4 de Receptores de Prostaglandina E/genética , Transducción de Señal/efectos de los fármacos , Factor de Transcripción Sp1/genética , Factor de Transcripción ReIA/genética
19.
Int J Mol Sci ; 16(1): 1806-20, 2015 Jan 14.
Artículo en Inglés | MEDLINE | ID: mdl-25594872

RESUMEN

Specificity protein 1 (SP1) is a ubiquitous transcription factor that plays an important role in controlling gene expression. Although important in mediating the function of various hormones, the role of SP1 in regulating milk fat formation remains unknown. To investigate the sequence and expression information, as well as its role in modulating lipid metabolism, we cloned SP1 gene from mammary gland of Xinong Saanen dairy goat. The full-length cDNA of the SP1 gene is 4376 bp including 103 bp of 5'UTR, 2358 bp of ORF (HM_236311) and 1915 bp of 3'UTR, which is predicted to encode a 786 amino acids polypeptide. Phylogenetic tree analysis showed that goat SP1 has the closest relationship with sheep, followed by bovines (bos taurus, odobenus and ceratotherium), pig, primates (pongo, gorilla, macaca and papio) and murine (rattus and mus), while the furthest relationship was with canis and otolemur. Expression was predominant in the lungs, small intestine, muscle, spleen, mammary gland and subcutaneous fat. There were no significant expression level differences between the mammary gland tissues collected at lactation and dry-off period. Overexpression of SP1 in goat mammary epithelial cells (GMECs) led to higher mRNA expression level of peroxisome proliferator-activated receptor-γ (PPARγ) and lower liver X receptor α (LXRα) mRNA level, both of which were crucial in regulating fatty acid metabolism, and correspondingly altered the expression of their downstream genes in GMECs. These results were further enhanced by the silencing of SP1. These findings suggest that SP1 may play an important role in fatty acid metabolism.


Asunto(s)
Células Epiteliales/metabolismo , Ácidos Grasos/metabolismo , Regulación de la Expresión Génica , Cabras/metabolismo , Glándulas Mamarias Humanas/metabolismo , Factor de Transcripción Sp1/metabolismo , Animales , ADN Complementario/genética , Ácidos Grasos/genética , Femenino , Cabras/genética , Humanos , Receptores X del Hígado , Glándulas Mamarias Humanas/citología , Modelos Moleculares , Receptores Nucleares Huérfanos/genética , PPAR gamma/genética , Filogenia , Factor de Transcripción Sp1/química , Factor de Transcripción Sp1/genética
20.
Arterioscler Thromb Vasc Biol ; 35(1): 71-8, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25359865

RESUMEN

OBJECTIVE: Homocysteine can accelerate the senescence of endothelial progenitor cells or endothelial cells (ECs) via telomerase inactivation and length shortening. However, the underlying mechanism is unclear. Here, we investigated whether homocysteine promotes endothelial senescence by reducing the expression and activity of human telomerase reverse transcriptase (hTERT) by DNA methylation to reduce ECs telomerase activity. APPROACH AND RESULTS: When compared with primary human umbilical vein endothelial cells grown under standard conditions, ECs with chronic homocysteine treatment showed accelerated upregulation of p16, p21, and p53, markers of cellular senescence, during 6 to 10 passages. Interestingly, homocysteine-stimulated but not angiotensin II-stimulated ECs senescence could be reversed by hypermethylation induced by folic acid or s-adenosylmethionine supplementation. Meanwhile, homocysteine promoted the shortening of telomere length specifically related to restoration of hTERT transcriptional expression and CCCTC-binding factor binding sites with hTERT promoter hypomethylation, as detected by quantitative real-time polymerase chain reaction, Western blot, methylation-specific polymerase chain reaction, and bisulfite sequencing assay. Electrophoretic mobility shift assay and chromatin immunoprecipitation results showed that homocysteine-reduced telomere activity and homocysteine-induced EC senescence might contribute to hTERT promoter demethylation by increasing CCCTC-binding factor repression and interfering in the SP1 binding to the demethylated hTERT promoter, which might relate with reduced of DNA methyltransferase 1. Furthermore, the CCCTC-binding factor-dependent mechanism of homocysteine-reduced hTERT expression via DNA demethylation was confirmed in aortic endothelia of mice with hyperhomocysteine levels. CONCLUSIONS: CCCTC-binding factor and SP1 cross talk may contribute to homocysteine-reduced hTERT DNA methylation and expression in endothelial senescence.


Asunto(s)
Senescencia Celular/efectos de los fármacos , Metilación de ADN/efectos de los fármacos , Homocisteína/farmacología , Células Endoteliales de la Vena Umbilical Humana/efectos de los fármacos , Telomerasa/metabolismo , Telómero/efectos de los fármacos , Angiotensina II/metabolismo , Animales , Sitios de Unión , Factor de Unión a CCCTC , Células Cultivadas , Inhibidor p16 de la Quinasa Dependiente de Ciclina/metabolismo , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/metabolismo , Modelos Animales de Enfermedad , Ácido Fólico/farmacología , Regulación Enzimológica de la Expresión Génica , Células Endoteliales de la Vena Umbilical Humana/enzimología , Humanos , Hiperhomocisteinemia/sangre , Hiperhomocisteinemia/enzimología , Hiperhomocisteinemia/genética , Masculino , Ratones Endogámicos C57BL , Regiones Promotoras Genéticas , Interferencia de ARN , Proteínas Represoras/genética , Proteínas Represoras/metabolismo , S-Adenosilmetionina/farmacología , Factor de Transcripción Sp1/genética , Factor de Transcripción Sp1/metabolismo , Telomerasa/genética , Telómero/metabolismo , Acortamiento del Telómero/efectos de los fármacos , Factores de Tiempo , Transfección , Proteína p53 Supresora de Tumor/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA