Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Eur J Nutr ; 53(3): 711-22, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24468940

RESUMEN

Nutrition and lifestyle, particularly over-nutrition and lack of exercise, promote the progression and pathogenesis of obesity and metabolic diseases. Nutrition is likely the most important environmental factor that modulates the expression of genes involved in metabolic pathways and a variety of phenotypes associated with obesity and diabetes. During pregnancy, diet is a major factor that influences the organ developmental plasticity of the foetus. Experimental evidence shows that nutritional factors, including energy, fatty acids, protein, micronutrients, and folate, affect various aspects of metabolic programming. Different epigenetic mechanisms that are elicited by bioactive factors in early critical developmental ages affect the susceptibility to several diseases in adulthood. The beneficial effects promoted by exercise training are well recognised, and physical exercise may be considered one of the more prominent non-pharmacological tools that can be used to attenuate metabolic programming and to consequently ameliorate the illness provoked by metabolic diseases and reduce the prevalence of obesity, type 2 diabetes, and cardiovascular diseases. Literature on the different outcomes of unbalanced diets and the beneficial effects of some bioactive molecules during gestation and lactation on the metabolic health of offspring, as well as the potential mechanisms underlying these effects, was reviewed. The importance of the combined effects of functional nutrition and exercise as reprogramming tools of metabolic programming is discussed in depth. Finally, this review provides recommendations to healthcare providers that may aid in the control of early programming in an attempt to optimise the health of the mother and child.


Asunto(s)
Medicina Basada en la Evidencia , Hiperfagia/fisiopatología , Conducta Materna , Fenómenos Fisiologicos Nutricionales Maternos , Intercambio Materno-Fetal , Síndrome Metabólico/etiología , Conducta Sedentaria , Animales , Desarrollo Infantil , Susceptibilidad a Enfermedades , Epigénesis Genética , Ejercicio Físico , Femenino , Desarrollo Fetal , Humanos , Hiperfagia/dietoterapia , Hiperfagia/metabolismo , Lactante , Recién Nacido , Lactancia/metabolismo , Masculino , Síndrome Metabólico/metabolismo , Síndrome Metabólico/prevención & control , Actividad Motora , Embarazo
2.
Tunis Med ; 89(6): 565-8, 2011 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-21681722

RESUMEN

AIMS: To investigate the frequencies of C677T polymorphism in MTHFR gene and G80A polymorphism in RFC gene in obese and no obese Tunisian children and to assess their relation with homocysteine (tHcy), folate and vitamin B12 levels. METHODS: We have studied 31 obese compared to 22 no obese children. tHcy was assessed by fluorescence-immunoassay ; folate and vitamin B12 by radioimmunoassay. C677T and G80A mutations were detected using pyrosequencing. RESULTS: There were no differences in tHcy levels between obese and no obese, (10,34 ± 4,86µmoll/l vs11,00 ± 4,26µmoll/l). We found no difference for the allelic frequencies of the C677T polymorphism (29.03 % vs 30.95 %) and of the G80A polymorphism (64.52 % vs 59.52 %). Mean levels of tHcy, folic acid and vitamin B12 were not significantly different according to MTHFR and RFC genotypes. CONCLUSION: We demonstrated no difference in tHcy, folates, vitamin B12 levels and allelic frequencies of C677T and G80A polymorphisms in MTHFR and RFC genes between obese and no obese Tunisian children. These two polymorphisms don't seem to have any impact on homocysteine, folate and vitamin B12 status in the two populations.


Asunto(s)
Homocisteína/sangre , Metilenotetrahidrofolato Reductasa (NADPH2)/genética , Mutación , Obesidad/sangre , Obesidad/genética , Polimorfismo Genético , Proteína Portadora de Folato Reducido/genética , Adolescente , Niño , Preescolar , Femenino , Humanos , Masculino , Túnez
3.
Med Princ Pract ; 18(2): 81-4, 2009.
Artículo en Inglés | MEDLINE | ID: mdl-19204424

RESUMEN

OBJECTIVE: To develop a method for the simultaneous measurement of 5-methylcytosine (5-metC) and 2'-deoxyuridine monophosphate (dU). MATERIALS AND METHODS: Genomic DNA was extracted from the HepG2 cell line grown in experimental complete medium or in folate-depleted medium. Samples were treated with RNAse A and RNAse T1 to avoid any RNA contamination. High-performance liquid chromatography (HPLC)/electrospray ionization mass spectrometric (ESI-MS) method was used to separate nucleotides after enzymatic hydrolysis of DNA with nuclease P1, phosphodiesterase I and alkaline phosphatase. RESULTS: Using this sensitive new methodology, we were able to quantify simultaneously the concentration of DNA-5-metC and DNA-uracil in DNA. The linear correlation coefficient (R(2)) between the MS signal and the concentration of 5-metC in a range of 0.5-5 microM or dU in a range of 10-100 microM was 0.9954 and 0.9999, respectively. The coefficient of variation was 16.94 and 14.77%, respectively. The applicability of this assay is demonstrated by detection of a decrease in 5-metC% and elevation of dU/thymidylate (dT) into genomic DNA extracted from the HepG2 cell line grown in a folate-depleted medium. CONCLUSION: Our results confirm that the HPLC/ESI-MS method reported earlier for measuring 5-metC allows measurement of uracil misincorporation into DNA.


Asunto(s)
5-Metilcitosina/química , ADN/química , Nucleótidos de Desoxiuracil/química , Uracilo/química , Línea Celular , Cromatografía Líquida de Alta Presión , Relación Dosis-Respuesta a Droga , Metilación , Hibridación de Ácido Nucleico , Espectrometría de Masa por Ionización de Electrospray
4.
Nutrients ; 11(8)2019 Jul 31.
Artículo en Inglés | MEDLINE | ID: mdl-31370166

RESUMEN

Iron is an essential nutrient needed for physiological functions, particularly during the developmental period of the early childhood of at-risk populations. The purpose of this study was to investigate, in an experimental colitis, the consequences of daily oral iron ingestion in the early period on the inflammatory response, the spleen T helper (Th) profiles and the associated molecular mechanisms. Juvenile mice orally received microencapsulated ferric iron or water for 6 weeks. On adult mice, we induced a sham or experimental trinitrobenzene sulfonic acid (TNBS) moderate colitis during the last week of the experiment before sacrificing the animals 7 days later. The severity of the gut inflammation was assessed by macroscopic damage scores (MDS) and the myeloperoxidase activity (MPO). Th profiles were evaluated by the examination of the splenic gene expression of key transcription factors of the Th differentiation (Tbet, Gata3, Foxp3 and RORγ) and the methylation of their respective promoter. While TNBS-induced colitis was associated with a change of the Th profile (notably an increase in the Tbet/Gata3 ratio in the spleen), the colitis-inhibition induced by ferric iron was associated with a limitation of the splenic Th profiles perturbation. The inhibition of the splenic Tbet gene overexpression was associated with an inhibition of promoter hypomethylation. In summary, mice treated by long-term oral ferric iron in the early period of life exhibited an inhibition of colitis associated with the inhibition of the splenic Tbet promoter hypomethylation and gene overexpression.


Asunto(s)
Colitis/prevención & control , Compuestos Férricos/farmacología , Regulación de la Expresión Génica/efectos de los fármacos , Regiones Promotoras Genéticas/fisiología , Factores de Transcripción/metabolismo , Administración Oral , Adulto , Envejecimiento , Animales , Islas de CpG , Composición de Medicamentos , Compuestos Férricos/administración & dosificación , Humanos , Masculino , Metilación , Ratones , Ratones Endogámicos BALB C , Factores de Transcripción/genética
5.
Carcinogenesis ; 28(11): 2291-7, 2007 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-17615259

RESUMEN

Fumonisin B1 (FB1) induces apoptosis and decreases the cellular uptake of 5-methyltetrahydrofolate. Two folate transporters (folate receptor, FR, and Reduced Folate Carrier, hRFC1) are involved in the cell uptake of folate. We aimed to study whether FB1 modifies the expression of the FR and the hRFC1 and whether its apoptotic effect is influenced by folate. Incubation of HepG2 cells with FB1 induced apoptosis in concentration and time-dependent manner in complete medium (experimental control medium, ECM), as well as in folate-depleted medium (FDM). FDM increased the toxicity of FB1 as the cells developed apoptosis within 24 h at 1 microM of FB1 instead of 100 microM in ECM. Whereas FR protein expression in cells grown in ECM was significantly inhibited after apoptosis event, protein expression of the hRFC1 was rather increased. The hrfc1 transcription was decreased in the treated cells. Under folate-deficient conditions, dramatic changes were observed on both transcriptional and post-transcriptional expression of the two transporters. FDM alone reduced FR protein expression by 12 +/- 2% and 43 +/- 1% at 48 and 72 h, respectively. The 5-methytetrahydrofolate attenuates apoptosis in a greater extent than the folic acid. However, its effects in preventing decrease of both folate transporters have not been observed. In conclusion, this study shows that the changes in the expression of FR after FB1 addition are probably a consequence of the FB1 toxicity. The response to FB1 by HepG2 cell lines is influenced by folate status and by folate form. 5-methyltetrahydrofolate appears to be more effective in preventing apoptosis than folic acid.


Asunto(s)
Proteínas Portadoras/metabolismo , Deficiencia de Ácido Fólico/metabolismo , Fumonisinas/farmacología , Proteínas de Transporte de Membrana/metabolismo , Micotoxinas/farmacología , Receptores de Superficie Celular/metabolismo , Apoptosis , Proteínas Portadoras/genética , Línea Celular , Medios de Cultivo , Receptores de Folato Anclados a GPI , Humanos , Proteínas de Transporte de Membrana/genética , ARN Mensajero/genética , Receptores de Superficie Celular/genética , Proteína Portadora de Folato Reducido
6.
Chem Biol Interact ; 159(1): 18-46, 2006 Jan 05.
Artículo en Inglés | MEDLINE | ID: mdl-16293235

RESUMEN

Ochratoxin A (OTA) is a mycotoxin produced by fungi of two genera: Penicillium and Aspergillus. OTA has been shown to be nephrotoxic, hepatotoxic, teratogenic and immunotoxic to several species of animals and to cause kidney and liver tumours in mice and rats. Because of differences in the physiology of animal species, wide variations are seen in the toxicokinetic patterns of absorption, distribution and elimination of the toxin. Biotransformation of OTA has not been entirely elucidated. At present, data regarding OTA metabolism are controversial. Several metabolites have been characterized in vitro and/or in vivo, whereas other metabolites remain to be characterized. Several major mechanisms have been shown as involved in the toxicity of OTA: inhibition of protein synthesis, promotion of membrane peroxidation, disruption of calcium homeostasis, inhibition of mitochondrial respiration and DNA damage. The contribution of metabolites in OTA genotoxicity and carcinogenicity is still unclear. The genotoxic status of OTA is still controversial because contradictory results were obtained in various microbial and mammalian tests, notably regarding the formation of DNA adducts. More recent studies are focused on the OTA ability to disturb cellular signalling and regulation, to modulate physiological signals and thereby to influence cells viability and proliferation. The present paper offers an update on these different issues. In addition since humans and animals are likely to be simultaneously exposed to several mycotoxins, especially through their diet, the little information available on the combined effects of OTA and other mycotoxins has also been reviewed.


Asunto(s)
Ocratoxinas/farmacocinética , Ocratoxinas/toxicidad , Animales , Aspergillus/metabolismo , Estructura Molecular , Ocratoxinas/química , Penicillium/metabolismo
7.
J Clin Res Pediatr Endocrinol ; 8(3): 257-63, 2016 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-27087124

RESUMEN

Despite health policies as well as clinical and research efforts, diabetes prevalence is still rising around the world. A multitude of causes have been suggested for this increase, mostly related to familial background, the occidental diet which is rich in fat/carbohydrates, and sedentary life style. Type 2 diabetes involves malfunctions of the primary pancreatic beta-cells, usually attributed to local damage; however, it can be associated with other stressful environmental agents, such as chemical contaminants from food, plastic and air, among others. Indeed, exposure to these chemical agents during perinatal and adolescent life can increase the risk of developing cardiometabolic diseases later in life. This review explores data showing which environmental chemical agents may produce injury in beta-cells and further impair the insulinotropic process of type 2 diabetes. Additionally, it points the need to also consider unusual causes of metabolic diseases, such as environmental contaminants.


Asunto(s)
Diabetes Mellitus Tipo 2/fisiopatología , Contaminantes Ambientales/envenenamiento , Células Secretoras de Insulina/efectos de los fármacos , Efectos Tardíos de la Exposición Prenatal/fisiopatología , Adolescente , Diabetes Mellitus Tipo 2/inducido químicamente , Femenino , Humanos , Recién Nacido , Células Secretoras de Insulina/patología , Enfermedades Metabólicas/inducido químicamente , Enfermedades Metabólicas/fisiopatología , Embarazo , Efectos Tardíos de la Exposición Prenatal/inducido químicamente
8.
Nutrients ; 7(4): 2748-70, 2015 Apr 14.
Artículo en Inglés | MEDLINE | ID: mdl-25875118

RESUMEN

Fetal life is characterized by a tremendous plasticity and ability to respond to various environmental and lifestyle factors, including maternal nutrition. Identification of the role of dietary factors that can modulate and reshape the cellular epigenome during development, including methyl group donors (e.g., folate, choline) and bioactive compounds (e.g., polyphenols) is of great importance; however, there is insufficient knowledge of a particular effect of each type of modulator and/or their combination on fetal life. To enhance the quality and safety of food products for proper fetal health and disease prevention in later life, a better understanding of the underlying mechanisms of dietary epigenetic modulators during the critical prenatal period is necessary. This review focuses on the influence of maternal dietary components on DNA methylation, histone modification, and microRNAs, and summarizes current knowledge of the effect and importance of dietary components on epigenetic mechanisms that control the proper expression of genetic information. Evidence reveals that some components in the maternal diet can directly or indirectly affect epigenetic mechanisms. Understanding the underlying mechanisms of how early-life nutritional environment affects the epigenome during development is of great importance for the successful prevention of adult chronic diseases through optimal maternal nutrition.


Asunto(s)
Dieta , Epigénesis Genética , Fenómenos Fisiologicos de la Nutrición Prenatal/fisiología , Carbohidratos de la Dieta/administración & dosificación , Grasas de la Dieta/administración & dosificación , Proteínas en la Dieta/administración & dosificación , Ingestión de Energía , Epigenómica , Femenino , Feto , Humanos , Micronutrientes/administración & dosificación , Embarazo
9.
Am J Clin Nutr ; 80(6): 1551-7, 2004 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-15585767

RESUMEN

BACKGROUND: The effects of supplementation with B vitamins and of common polymorphisms in genes involved in homocysteine metabolism on plasma total homocysteine (tHcy) concentrations in trisomy 21 are unknown. OBJECTIVES: We aimed to determine the effects of orally administered folic acid and of folic acid combined with vitamin B-12, vitamin B-6, or both on tHcy in adults with trisomy 21. The study was also intended to analyze the possible influence of gene polymorphisms. DESIGN: One hundred sixty adults with trisomy 21 and 160 healthy, unrelated subjects aged 26 +/- 4 y were included. Plasma tHcy, red blood cell folate, serum folate, and vitamin B-12 were measured. Genotyping for the common methylenetetrahydrofolate reductase (MTHFR) 677C-->T, MTHFR 1298A-->C, cystathionine beta-synthase 844Ins68, methionine synthase 2756A-->C, methionine synthase reductase 66A-->G, and reduced folate carrier 80G-->A polymorphisms was carried out. RESULTS: The mean tHcy concentration (9.8 +/- 0.7 micromol/L) of cases who did not use vitamins was not significantly different from that of controls (9.4 +/- 0.3 micromol/L). Plasma tHcy concentrations (7.6 +/- 0.3 mmol/L) in cases who used folic acid were significantly lower than in cases who did not. Folic acid combined with vitamin B-12 did not significantly change tHcy concentrations compared with those in cases who used only folic acid. Folic acid combined with vitamins B-6 and B-12 significantly lowered tHcy (6.5 +/- 0.5 micromol/L). The difference in tHcy according to MTHFR genotype was not significant. However, tHcy concentrations were slightly higher in TT homozygotes among the controls but not among the cases. CONCLUSION: This study provides information on the relation between several polymorphisms in genes involved in homocysteine and folate metabolism in adults with trisomy 21.


Asunto(s)
Síndrome de Down/sangre , Ácido Fólico/administración & dosificación , Homocisteína/sangre , Polimorfismo Genético , Vitamina B 12/administración & dosificación , Vitamina B 6/administración & dosificación , 5-Metiltetrahidrofolato-Homocisteína S-Metiltransferasa/genética , Administración Oral , Adolescente , Adulto , Estudios de Casos y Controles , Cistationina betasintasa/genética , Suplementos Dietéticos , Síndrome de Down/tratamiento farmacológico , Síndrome de Down/genética , Sinergismo Farmacológico , Femenino , Ferredoxina-NADP Reductasa/genética , Ácido Fólico/sangre , Genotipo , Homocisteína/efectos de los fármacos , Homocigoto , Humanos , Masculino , Proteínas de Transporte de Membrana/genética , Metilenotetrahidrofolato Reductasa (NADPH2)/genética , Persona de Mediana Edad , Proteína Portadora de Folato Reducido , Vitamina B 12/sangre
10.
Ann Biol Clin (Paris) ; 72(6): 669-80, 2014.
Artículo en Francés | MEDLINE | ID: mdl-25486663

RESUMEN

Advanced glycation endproducts or advanced glycation end products (AGEs) levels increase in blood or tissue during aging and in diseases such as diabetes and renal failure. The receptor of advanced glycation endproducts (RAGE), is a multi-ligand receptor belonging to the immunoglobulin superfamily. It is weakly expressed in most adult tissues. The link between the RAGE and its ligands triggers a cascade of intracellular events, followed by the transcription of a range of genes involved in different biological systems, as well as other reactions such as the formation of oxidative stress. All of these reactions lead to a series of functional changes that participate in neurological and vascular complications of diabetes, metabolic syndrome. They are also involved in Alzheimer's disease, arthritis, and some cancers. Besides the presence of these ligands, the chronic stimulation of the RAGE, or its isoforms trigger different signaling pathways and reactions. This makes complex the analysis of biological networks associated with multiple clinical traits. Many issues remain to be clarified in the pathogenicity of RAGE. In the era of the development of systems biology, integrative approach is expected allowing a better understanding of the overall effects of RAGE system. In this review, we discuss recent advances in our understanding of the biology of the RAGE, focusing on how systems biology influence our view it processes. The review should contribute to the better understanding of pathogenicity of "RAGE system" and to the transposition this new knowledge into clinical and translational research.


Asunto(s)
Receptores Inmunológicos/fisiología , Adulto , Envejecimiento/metabolismo , Empalme Alternativo/genética , Diabetes Mellitus/metabolismo , Diabetes Mellitus/terapia , Regulación de la Expresión Génica , Productos Finales de Glicación Avanzada/metabolismo , Humanos , Estrés Oxidativo/fisiología , Receptor para Productos Finales de Glicación Avanzada , Receptores Inmunológicos/química , Insuficiencia Renal/metabolismo , Insuficiencia Renal/terapia , Biología de Sistemas
11.
Ann Biol Clin (Paris) ; 72(3): 265-9, 2014.
Artículo en Francés | MEDLINE | ID: mdl-24876137

RESUMEN

The qPCR has been introduced in clinical and biomedical research for over 10 years from now. Its use in trials and diagnostics is continuously increasing. Due to this heavy use, the question of relyability and relevance of qPCR results has to be asked. This review proposes a documented and evidence based answer to this question, thanks to the MIQE (minimum information for publication of quantitative real-time PCR experiments) guideline. The whole analysis process is addressed, from nucleic acids extraction to data management. Simple answers are given, taking into account the technical constraints from clinical research in order to allow a realistic application of this guideline.


Asunto(s)
Guías de Práctica Clínica como Asunto/normas , Reacción en Cadena en Tiempo Real de la Polimerasa/normas , Adhesión a Directriz , Humanos , Reacción en Cadena en Tiempo Real de la Polimerasa/estadística & datos numéricos , Transcripción Reversa/genética , Terminología como Asunto
12.
Mol Nutr Food Res ; 56(6): 976-85, 2012 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-22707272

RESUMEN

SCOPE: Prenatal folate and methyl donor malnutrition lead to epigenetic alterations that could enhance susceptibility to disease. Methyl-deficient diet (MDD) and fumonisin FB1 are risk factors for neural tube defects and cancers. Evidence indicates that FB1 impairs folate metabolism. METHODS AND RESULTS: Folate receptors and four heterochromatin markers were investigated in rat fetuses liver derived from dams exposed to MDD and/or FB1 administered at a dose twice higher than the provisional maximum tolerable daily intake (PMTDI = 2 µg/kg/day). Even though folate receptors transcription seemed up-regulated by methyl depletion regardless of FB1 treatment, combined MDD/FB1 exposure might reverse this up-regulation since folate receptors transcripts were lower in the MDD/FB1 versus MDD group. Methyl depletion decreased H4K20me3. Combined MDD/FB1 decreased H4K20me3 even more and increased H3K9me3. The elevated H3K9me3 can be viewed as a defense mechanism inciting the cell to resist heterochromatin disorganization. H3R2me2 and H4K16Ac varied according to this mechanism even though statistical significance was not consistent. CONCLUSION: Considering that humans are exposed to FB1 levels above the PMTDI, this study is relevant because it suggests that low doses of FB1 interact with MDD thus contributing to disrupt the epigenetic landscape.


Asunto(s)
Anomalías Inducidas por Medicamentos/metabolismo , Ácido Fólico/metabolismo , Fumonisinas/toxicidad , Histonas/metabolismo , Hígado/metabolismo , Fenómenos Fisiologicos Nutricionales Maternos , Teratógenos/toxicidad , Anomalías Inducidas por Medicamentos/embriología , Anomalías Inducidas por Medicamentos/patología , Anomalías Inducidas por Medicamentos/fisiopatología , Animales , Deficiencia de Colina/complicaciones , Deficiencia de Colina/embriología , Deficiencia de Colina/metabolismo , Deficiencia de Colina/patología , Hígado Graso/etiología , Femenino , Deficiencia de Ácido Fólico/complicaciones , Deficiencia de Ácido Fólico/embriología , Deficiencia de Ácido Fólico/metabolismo , Deficiencia de Ácido Fólico/patología , Transportadores de Ácido Fólico/genética , Transportadores de Ácido Fólico/metabolismo , Fumonisinas/administración & dosificación , Regulación del Desarrollo de la Expresión Génica/efectos de los fármacos , Heterocromatina/efectos de los fármacos , Heterocromatina/metabolismo , Hígado/efectos de los fármacos , Hígado/embriología , Hígado/patología , Metilación/efectos de los fármacos , Defectos del Tubo Neural/inducido químicamente , Defectos del Tubo Neural/etiología , Embarazo , ARN Mensajero/metabolismo , Ratas , Ratas Wistar , Deficiencia de Vitamina B 12/complicaciones , Deficiencia de Vitamina B 12/embriología , Deficiencia de Vitamina B 12/metabolismo , Deficiencia de Vitamina B 12/patología
13.
Nutr Rev ; 69(1): 34-40, 2011 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-21198633

RESUMEN

Malaria induced by Plasmodium falciparum is a major cause of mortality. P. falciparum has the ability to use host plasma folate as its primary folate source. Folate is a cofactor needed for both malaria parasite growth and host erythrocyte production. This review examines the possible impairment of the folate-mediated one-carbon metabolism pathway as a result of P. falciparum malaria infection during pregnancy. Folate deficiency during malaria infection is presented, with an emphasis on the controversy regarding the decrease of plasma or erythrocyte folate secondary to malaria. Maternal folate deficiency increases the risk of adverse pregnancy outcomes. Functional folate deficiency and/or increased plasma homocysteine levels during pregnancy of infected women in areas endemic for malaria is a probable scenario accentuating the impairment of placenta function leading to the occurrence of neural tube defects, low birth weights, and intrauterine growth retardations. Potential questions that may be answered in future investigations using an appropriate protocol to study pregnant women with malaria are also addressed.


Asunto(s)
Ácido Fólico/metabolismo , Malaria Falciparum/metabolismo , Plasmodium falciparum/metabolismo , Complicaciones Parasitarias del Embarazo/metabolismo , Animales , Eritrocitos/metabolismo , Eritrocitos/parasitología , Femenino , Deficiencia de Ácido Fólico/complicaciones , Deficiencia de Ácido Fólico/metabolismo , Humanos , Recién Nacido , Plasmodium falciparum/crecimiento & desarrollo , Embarazo , Resultado del Embarazo
14.
Mol Biosyst ; 7(8): 2508-21, 2011 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-21647531

RESUMEN

Dynamical modeling is an accurate tool for describing the dynamic regulation of one-carbon metabolism (1CM) with emphasis on the alteration of DNA methylation and/or dUMP methylation into dTMP. Using logic programming we present a comprehensive and adaptative mathematical model to study the impact of folate deficiency, including folate transport and enzymes activities. 5-Methyltetrahydrofolate (5mTHF) uptake and DNA and dUMP methylation were studied by simulating nutritional 5mTHF deficiency and methylenetetrahydrofolate reductase (MTHFR) gene defects. Both conditions had distinct effects on 1CM metabolite synthesis. Simulating severe 5mTHF deficiency (25% of normal levels) modulated 11 metabolites. However, simulating a severe decrease in MTHFR activity (25% of normal activity) modulated another set of metabolites. Two oscillations of varying amplitude were observed at the steady state for DNA methylation with severe 5mTHF deficiency, and the dUMP/dTMP ratio reached a steady state after 2 h, compared to 2.5 h for 100% 5mTHF. MTHFR activity with 25% of V(max) resulted in an increased methylated DNA pool after half an hour. We observed a deviation earlier in the profile compared to 50% and 100% V(max). For dUMP methylation, the highest level was observed with 25%, suggesting a low rate of dUMP methylation into dTMP with 25% of MTHFR activity. In conclusion, using logic programming we were able to construct the 1CM for analyzing the dynamic system behavior. This model may be used to refine biological interpretations of data or as a tool that can provide new hypotheses for pathogenesis.


Asunto(s)
Inteligencia Artificial , Metilación de ADN , Deficiencia de Ácido Fólico/metabolismo , Biología de Sistemas/métodos , Deficiencia de Ácido Fólico/genética , Humanos , Lógica , Redes y Vías Metabólicas , Metilenotetrahidrofolato Reductasa (NADPH2) , Modelos Biológicos , Transferasas del Grupo 1-Carbono/química , Transferasas del Grupo 1-Carbono/metabolismo , Tetrahidrofolatos
16.
J Nutr Biochem ; 20(4): 312-20, 2009 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-18602821

RESUMEN

The integrated view of the expression of genes involved in folate-dependent one-carbon metabolism (FOCM) under folate deficiency remains unknown. Dynamics of changes in the transcriptional expression of 28 genes involved in the FOCM network were evaluated at different time points (0, 2, 4, 6, 12, 24 and 48 h) in human hepatoma HepG2 cell line. Combined experimental and computational approaches were conducted for emphasizing characteristic patterns in the gene expression changes produced by cellular folate deficiency. Bivariate analysis showed that folate deficiency (0.3 nmol/L of folate vs. 2.27 mumol/L in control medium) displayed rapid and coordinated regulation during the first 2 h with differential expression for hRfc1 (increased by 69%) and Ahcy (decreased by 437%). Density analysis through the time points gave evidence of differential expression for five genes (Ahcy, Cth, Gnmt, Mat1A, Mtrr and hRfc1). Differential expression of Ahcy, Gnmt, Mat1A and Mtrr was confirmed by time-series analysis gene expression. We also found a marked differential expression of Mtrr. Qualitative analysis of genes allowed identifying four clusters of gene that was coexpressed. Two of these clusters were consistent with specific metabolic functions as they associated genes involved in the remethylation (Mthfr and Mtrr) and in the transmethylation (Dnmt1and Dnmt3B) pathways. The study shows a strong influence of folate status on Mtrr transcription in HepG2 cells. It suggests also that folate deficiency produces transcription changes that particularly involve the clusters of genes related with the remethylation and the transmethylation pathways.


Asunto(s)
Deficiencia de Ácido Fólico/genética , Deficiencia de Ácido Fólico/metabolismo , Ácido Fólico/metabolismo , Expresión Génica , Proteínas Portadoras/genética , Proteínas Portadoras/metabolismo , Línea Celular Tumoral , Ferredoxina-NADP Reductasa/genética , Ferredoxina-NADP Reductasa/metabolismo , Receptores de Folato Anclados a GPI , Humanos , Proteínas de Transporte de Membrana/genética , Proteínas de Transporte de Membrana/metabolismo , Receptores de Superficie Celular/genética , Receptores de Superficie Celular/metabolismo , Factores de Tiempo
17.
Biochem Biophys Res Commun ; 349(2): 492-6, 2006 Oct 20.
Artículo en Inglés | MEDLINE | ID: mdl-16949045

RESUMEN

Relative levels of DNA hypermethylation were quantified in DS individuals using a new method based on a combination of methylation-sensitive arbitrarily primed polymerase chain reaction (MS-AP-PCR) and quantification of DNA fragments with the Agilent 2100 bioanalyzer. Four of the DS individuals had low plasma total homocysteine (tHcy) level (4.3 +/- 0.3 micromol/l) and 4 other had high-tHcy level (14.1 +/- 0.9 micromol/l). Eight healthy control individuals were matched to the DS cases for age, sex, and tHcy levels. We have identified and quantified six hypermethylated fragments. Their sizes ranged from 230-bp to 700-bp. In cases and controls, low-tHcy did not affect methylation level of identified fragments, mean methylation values were 68.0 +/- 39.7% and 52.1 +/- 40.3%, respectively. DNA methylation in DS individuals did not change significantly (59.7+/-34.5%) in response to high-tHcy level in contrast to controls (23.4 +/- 17.7%, P = 0.02). Further, the quantitative MS-AP-PCR using this microfludic system is a useful method for determining differential genomic DNA methylation.


Asunto(s)
Metilación de ADN , Síndrome de Down/diagnóstico , Síndrome de Down/genética , Reacción en Cadena de la Polimerasa/métodos , Adolescente , Adulto , Estudios de Casos y Controles , Fragmentación del ADN , Femenino , Técnicas Genéticas , Homocisteína/sangre , Humanos , Masculino , Análisis de Secuencia por Matrices de Oligonucleótidos
18.
Br J Nutr ; 94(2): 166-9, 2005 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-16115349

RESUMEN

The cause of the non-disjunction leading to trisomy 21 remains unclear. Recent evidence has suggested that 5,10-methylenetetrahydrofolate reductase (MTHFR) and/or methionine synthase reductase (MTRR) might contribute to the maternal risk of trisomy 21. The purpose of the present study was to analyse these findings among the French population and to investigate whether common polymorphisms in genes of the folate and homocysteine pathway, including the MTHFR 677C > T, MTHFR 1298A > C, the methionine synthase (MTR) 2756A > G, the cystathionine beta-synthase (CBS) 844Ins68 and the reduced folate carrier (RFC-1) 80G > A polymorphisms, contribute to the risk of trisomy 21. The risk was studied by analysing independent and combined genotypes in 119 case mothers and 119 control mothers. The MTHFR 677T, MTHFR 1298C, MTR2756G, MTRR66G, CBSIns68+ and the RFC-1 80G allele frequencies were not significantly different among French case mothers, compared with control mothers. The risk of having a child with trisomy 21 did not appear to be linked to polymorphisms in genes associated with folate and homocysteine metabolism.


Asunto(s)
Síndrome de Down/genética , Ácido Fólico/genética , Homocisteína/genética , Polimorfismo Genético/genética , 5-Metiltetrahidrofolato-Homocisteína S-Metiltransferasa/genética , Adulto , Síndrome de Down/metabolismo , Femenino , Ferredoxina-NADP Reductasa/genética , Ácido Fólico/metabolismo , Francia , Genotipo , Homocisteína/metabolismo , Humanos , Proteínas de Transporte de Membrana/genética , Metilenotetrahidrofolato Reductasa (NADPH2)/genética , Madres , Proteína Portadora de Folato Reducido , Factores de Riesgo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA