Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 7 de 7
Filtrar
1.
FASEB J ; 26(10): 3980-92, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-22713523

RESUMEN

Despite the key role in neuronal development of a deficit in the methyl donor folate, little is known on the underlying mechanisms. We therefore studied the consequences of folate deficiency on proliferation, differentiation, and plasticity of the rat H19-7 hippocampal cell line. Folate deficit reduced proliferation (17%) and sensitized cells to differentiation-associated apoptosis (+16%). Decreased production (-58%) of S-adenosylmethionine (the universal substrate for transmethylation reactions) and increased expression of histone deacetylases (HDAC4,6,7) would lead to epigenomic changes that may impair the differentiation process. Cell polarity, vesicular transport, and synaptic plasticity were dramatically affected, with poor neurite outgrowth (-57%). Cell treatment by an HDAC inhibitor (SAHA) led to a noticeable improvement of cell polarity and morphology, with longer processes. Increased homocysteine levels (+55%) consecutive to folate shortage produced homocysteinylation, evidenced by coimmunoprecipitations and mass spectrometry, and aggregation of motor proteins dynein and kinesin, along with functional alterations, as reflected by reduced interactions with partner proteins. Prominent homocysteinylation of key neuronal proteins and subsequent aggregation certainly constitute major adverse effects of folate deficiency, affecting normal development with possible long-lasting consequences.


Asunto(s)
Deficiencia de Ácido Fólico/metabolismo , Ácido Fólico/farmacología , Hipocampo/citología , Homocisteína/farmacología , Neuronas/citología , Neuronas/efectos de los fármacos , Animales , Western Blotting , Diferenciación Celular/efectos de los fármacos , Línea Celular , Movimiento Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Células Hep G2 , Humanos , Inmunohistoquímica , Neuronas/metabolismo , Unión Proteica , Ratas , Ratas Wistar , Reacción en Cadena en Tiempo Real de la Polimerasa , Vitamina B 12/farmacología
2.
Am J Pathol ; 176(1): 270-7, 2010 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-19948829

RESUMEN

Methyl donor deficiency (MDD) during pregnancy influences intrauterine development. Ghrelin is expressed in the stomach of fetuses and influences fetal growth, but MDD influence on gastric ghrelin is unknown. We examined the gastric ghrelin system in MDD-induced intrauterine growth retardation. By using specific markers and approaches (such as periodic acid-Schiff, bromodeoxyuridine, homocysteine, terminal deoxynucleotidyl transferase biotin-dUTP nick end labeling, immunostaining, reverse transcription-polymerase chain reaction), we studied the gastric oxyntic mucosa cellular organization and ghrelin gene expression in the mucosa in 20-day-old fetuses and weanling pups, and plasma ghrelin concentration in weanling rat pups of dams either normally fed or deprived of choline, folate, vitamin B6, and vitamin B12 during gestation and suckling periods. MDD fetuses weighed less than controls; the weight deficit reached 57% at weaning (P < 0.001). Both at the end of gestation and at weaning, they presented with an aberrant gastric oxyntic mucosa formation with loss of cell polarity, anarchic cell migration, abnormal progenitor differentiation, apoptosis, and signs of surface layer erosion. Ghrelin cells were abnormally located in the pit region of oxyntic glands. At weaning, plasma ghrelin levels were decreased (-28%; P < 0.001) despite unchanged mRNA expression in the stomach. This decrease was associated with lower body weight. Taken together, these data indicate that one mechanism through which MDD influences fetal programming is the remodeling of gastric cellular organization, leading to dysfunction of the ghrelin system and dramatic effects on growth.


Asunto(s)
Enfermedades Carenciales/embriología , Enfermedades Carenciales/fisiopatología , Desarrollo Fetal , Mucosa Gástrica/metabolismo , Mucosa Gástrica/patología , Ghrelina/metabolismo , Animales , Peso Corporal , Linaje de la Célula , Colina/metabolismo , Células Enteroendocrinas/metabolismo , Femenino , Ácido Fólico/metabolismo , Ghrelina/sangre , Hormona del Crecimiento/sangre , Homocisteína/sangre , Inmunohistoquímica , Embarazo , Ratas , Ratas Wistar , Vitamina B 12/metabolismo , Destete
4.
PLoS One ; 4(7): e6325, 2009 Jul 22.
Artículo en Inglés | MEDLINE | ID: mdl-19623264

RESUMEN

BACKGROUND: Oleosin is a plant protein localized to lipid droplets and endoplasmic reticulum of plant cells. Our idea was to use it to target functional secretory proteins of interest to the cytosolic side of the endoplasmic reticulum of mammalian cells, through expressing oleosin-containing chimeras. We have designed this approach to create cellular models deficient in vitamin B12 (cobalamin) because of the known problematics associated to the obtainment of effective vitamin B12 deficient cell models. This was achieved by the overexpression of transcobalamin inside cells through anchoring to oleosin. METHODOLOGY: chimera gene constructs including transcobalamin-oleosin (TC-O), green fluorescent protein-transcobalamin-oleosin (GFP-TC-O) and oleosin-transcobalamin (O-TC) were inserted into pAcSG2 and pCDNA3 vectors for expression in sf9 insect cells, Caco2 (colon carcinoma), NIE-115 (mouse neuroblastoma), HEK (human embryonic kidney), COS-7 (Green Monkey SV40-transfected kidney fibroblasts) and CHO (Chinese hamster ovary cells). The subcellular localization, the changes in vitamin B12 binding activity and the metabolic consequences were investigated in both Caco2 and NIE-115 cells. PRINCIPAL FINDINGS: vitamin B12 binding was dramatically higher in TC-O than that in O-TC and wild type (WT). The expression of GFP-TC-O was observed in all cell lines and found to be co-localized with an ER-targeted red fluorescent protein and calreticulin of the endoplasmic reticulum in Caco2 and COS-7 cells. The overexpression of TC-O led to B12 deficiency, evidenced by impaired conversion of cyano-cobalamin to ado-cobalamin and methyl-cobalamin, decreased methionine synthase activity and reduced S-adenosyl methionine to S-adenosyl homocysteine ratio, as well as increases in homocysteine and methylmalonic acid concentration. CONCLUSIONS/SIGNIFICANCE: the heterologous expression of TC-O in mammalian cells can be used as an effective strategy for investigating the cellular consequences of vitamin B12 deficiency. More generally, expression of oleosin-anchored proteins could be an interesting tool in cell engineering for studying proteins of pharmacological interest.


Asunto(s)
Retículo Endoplásmico/metabolismo , Proteínas de Plantas/metabolismo , Transcobalaminas/metabolismo , Vitamina B 12/metabolismo , Animales , Secuencia de Bases , Western Blotting , Línea Celular , Cartilla de ADN , ADN Complementario , Electroforesis en Gel de Poliacrilamida , Humanos , Microscopía Confocal
5.
Cell Physiol Biochem ; 20(6): 967-76, 2007.
Artículo en Inglés | MEDLINE | ID: mdl-17982279

RESUMEN

BACKGROUND: P-glycoprotein (Pgp), produced by multidrug resistance-1 gene (mdr-1), is a main mechanism developed by cancer cells to guard against anti-cancer drugs. Alterations of DNA methylation of the mdr-1 gene promoter are known to be linked to mdr-1 gene expression and are probably related to intracellular S-adenosyl-methionine. We here used HepG2 cells to determine the role of the methionine cycle (through the use of the Methionine-Synthase (MS) cofactor, cobalamin) on mdr-1 gene expression. METHODS: Semiquantitative RT-PCR of mdr-1 gene, cellular retention of rhodamine-123, and vinblastine cytotoxicity were carried out on cells cultivated with and without cobalamin. Methylation status of the mdr-1 gene promoter was determined by methylation-specific PCR. RESULTS: Addition of cobalamin to the cells led to an increase in MS activity, to a significant decrease in mdr-1 gene expression which is correlated to an increase in retention of the Pgp substrate Rhodamine 123. Furthermore, cobalamin potentiated cell sensitivity to vinblastine to the same range as that of the Pgp blocker verapamil and prevented methotrexate-induced up-regulation of mdr-1 gene expression. However, no modification in methylation of the mdr-1 gene promoter was observed. CONCLUSION: Cobalamin downregulates mdr-1 gene expression, as well as Pgp expression and function, and significantly increases cytotoxicity of vinblastine. The identification of this novel way of diminishing cellular resistance to the chemotherapeutic agent vinblastine holds promises of leading to better treatments for cancer patients.


Asunto(s)
Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/genética , Regulación hacia Abajo/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Vinblastina/farmacología , Vitamina B 12/farmacología , 5-Metiltetrahidrofolato-Homocisteína S-Metiltransferasa/metabolismo , Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/metabolismo , Muerte Celular/efectos de los fármacos , Línea Celular Tumoral , Metilación de ADN/efectos de los fármacos , Resistencia a Antineoplásicos/efectos de los fármacos , Sinergismo Farmacológico , Humanos , Metotrexato/farmacología , Regiones Promotoras Genéticas , Rodamina 123/metabolismo , Regulación hacia Arriba/efectos de los fármacos
6.
J Gene Med ; 7(11): 1439-49, 2005 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-16001392

RESUMEN

BACKGROUND: In vivo, tracheal gland serous cells highly express the cystic fibrosis transmembrane conductance regulator (cftr) gene. This gene is mutated in the lethal monogenic disease cystic fibrosis (CF). Clinical trials in which the human CFTR cDNA was delivered to the respiratory epithelia of CF patients have resulted in weak and transient gene expression. METHODS AND RESULTS: As CF is characterized by mucus inspissation, airway infection, and severe inflammation, we tested the hypothesis that inflammation and especially two cytokines involved in the Th1/Th2 inflammatory response, interleukin 4 (IL-4) and TNFalpha, could inhibit gene transfer efficiency using a model of human CF tracheal gland cells (CF-KM4) and Lipofectamine reagent as a transfection reagent. The specific secretory defects of CF-KM4 cells were corrected by Lipofectamine-mediated human CFTR gene transfer. However, this was altered when cells were pre-treated with IL-4 and TNFalpha. Inhibition of luciferase reporter gene expression by IL-4 and TNFalpha pre-treated CF-KM4 cells was measured by activity and real-time RT-PCR. Both cytokines induced similar and synergistic inhibition of transgene expression and activity. This cytokine-mediated inhibition could be prevented by anti-inflammatory agents such as glucocorticoids but not by non-steroidal (NSAI) agents. CONCLUSIONS: This data suggests that an inflammatory context generated by IL-4 and TNFalpha can inhibit human CFTR gene transfer in CF tracheal gland cells and that glucocorticoids may have a protecting action.


Asunto(s)
Regulador de Conductancia de Transmembrana de Fibrosis Quística , Fibrosis Quística , Técnicas de Transferencia de Gen , Interleucina-4/metabolismo , Lípidos/química , Tráquea/citología , Factor de Necrosis Tumoral alfa/metabolismo , Antiinflamatorios/metabolismo , Línea Celular , Fibrosis Quística/genética , Fibrosis Quística/inmunología , Fibrosis Quística/fisiopatología , Fibrosis Quística/terapia , Regulador de Conductancia de Transmembrana de Fibrosis Quística/genética , Regulador de Conductancia de Transmembrana de Fibrosis Quística/metabolismo , Dexametasona/metabolismo , Genes Reporteros , Humanos , Luciferasas/genética , Luciferasas/metabolismo , Proteínas Inhibidoras de Proteinasas Secretoras , Proteínas/metabolismo , Receptores Tipo I de Factores de Necrosis Tumoral/metabolismo , Receptores Tipo II del Factor de Necrosis Tumoral/metabolismo , Tráquea/metabolismo
7.
Cell Physiol Biochem ; 14(4-6): 407-14, 2004.
Artículo en Inglés | MEDLINE | ID: mdl-15319544

RESUMEN

BACKGROUND: Methionine synthase (MTR) synthesizes methionine from homocysteine, using cobalamin as a cofactor and 5-methyltetrahydrofolate as a cosubstrate. AIM: To determine the influence of homocystine (Hcy, dimer of homocysteine) in the presence of either cobalamin or methionine on the transcription and the activity of methionine synthase in Caco-2, a human adenocarcinoma cell line. METHODS: Methionine synthase activity and quantification of its mRNA by real-time RT-PCR were determined in cells cultivated under four differents conditions: Hcy with cobalamin (Hcy+ Cbl+), Hcy with methionine (Hcy+Met+), methionine with Cbl (Met+ Cbl+) and methionine only (Met+). RESULTS: Activity (nmol/h/mg protein) was maximal in cells cultivated in Hcy+Cbl+ (2.45 +/- 0.35), compared to cells cultivated in Hcy+Met+ (0.18 +/- 0.01, p<0.001), in Met+ Cbl+ (1.60 +/- 0.06, p<0.05), and in Met+ (0.40 +/- 0.05, p<0.001), suggesting an adaptation of the cells to requirement in synthesized methionine. The mRNA level of MTR in Hcy+ Cbl+ and Hcy+Met+ (2.82 +/-0.49 and 3.33 +/- 0.48 AU, respectively ) was about 2.5 / 3.0-fold higher than that in Met+ Cbl+ and in Met+ (1.00 +/-0.13 and 1.20 +/-0.20 AU, respectively, p<0.001). CONCLUSION: Methionine synthase expression of Caco-2 cell is under a transcriptional control influenced by Hcy.


Asunto(s)
5-Metiltetrahidrofolato-Homocisteína S-Metiltransferasa/genética , Adenocarcinoma/genética , Regulación Neoplásica de la Expresión Génica , Homocisteína/fisiología , Regulación hacia Arriba , 5-Metiltetrahidrofolato-Homocisteína S-Metiltransferasa/análisis , 5-Metiltetrahidrofolato-Homocisteína S-Metiltransferasa/biosíntesis , Adenocarcinoma/química , Adenocarcinoma/enzimología , Células CACO-2 , Homocisteína/farmacología , Humanos , Metionina/farmacología , Metionina/fisiología , ARN Mensajero/análisis , ARN Mensajero/metabolismo , Transcripción Genética/genética , Vitamina B 12/farmacología , Vitamina B 12/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA