Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 44
Filtrar
Más filtros

Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
Int J Mol Sci ; 20(5)2019 Mar 04.
Artículo en Inglés | MEDLINE | ID: mdl-30836646

RESUMEN

Supplementation of micronutrients like folate is a double-edged sword in terms of their ambivalent role in cell metabolism. Although several epidemiological studies support a protective role of folate in carcinogenesis, there are also data arguing for an opposite effect. To address this issue in the context of human papillomavirus (HPV)-induced transformation, the molecular events of different folate availability on human keratinocytes immortalized by HPV16 E6 and E7 oncoproteins were examined. Several sublines were established: Control (4.5 µM folate), folate deficient (0.002 µM folate), and repleted cells (4.5 µM folate). Cells were analyzed in terms of oncogene expression, DNA damage and repair, karyotype changes, whole-genome sequencing, and transcriptomics. Here we show that folate depletion irreversibly induces DNA damage, impairment of DNA repair fidelity, and unique chromosomal alterations. Repleted cells additionally underwent growth advantage and enhanced clonogenicity, while the above mentioned impaired molecular properties became even more pronounced. Overall, it appears that a period of folate deficiency followed by repletion can shape immortalized cells toward an anomalous phenotype, thereby potentially contributing to carcinogenesis. These observations should elicit questions and inquiries for broader additional studies regarding folate fortification programs, especially in developing countries with micronutrient deficiencies and high HPV prevalence.


Asunto(s)
Deficiencia de Ácido Fólico/genética , Papillomavirus Humano 16/genética , Infecciones por Papillomavirus/genética , Transcripción Genética , Carcinogénesis/genética , Daño del ADN/ética , Reparación del ADN/genética , Ácido Fólico/genética , Deficiencia de Ácido Fólico/patología , Deficiencia de Ácido Fólico/virología , Genómica , Papillomavirus Humano 16/patogenicidad , Humanos , Queratinocitos/virología , Proteínas Oncogénicas Virales/genética , Proteínas E7 de Papillomavirus/genética , Infecciones por Papillomavirus/virología , Proteínas Represoras/genética
2.
J Clin Neurosci ; 59: 341-344, 2019 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-30420205

RESUMEN

INTRODUCTION: Folate is essential for production of DNA, neurotransmitters and myelin and regulation of genetic activity. A specific transporter protein is required to transport folate from blood to CSF. Various inherited brain-specific folate transport defects have been recognized due to mutation in Folate Receptor alpha (FOLR1). FOLR1 mutation is one of the vitamin responsive encephalopathies and is inherited as an autosomal recessive condition. It has a wide spectrum of phenotype, commonly presenting as epileptic encephalopathy. Less frequently the condition may manifest with subtle hypotonia, movement disorder as tremors, ataxia or intellectual disability and autistic spectrum disorder. We present a case of folate transporter deficiency with non-epileptic manifestations, presenting with tremors, speech delay and stable white matter changes in MRI brain. OBJECTIVE: We present a case of Folate transporter defect with Non-epileptic presentation. CONCLUSION: Folate transporter deficiency has a wide range of presenting symptoms. Presentation with slowly progressive atypical symptoms, stable white matter changes in brain MRI that does not fit a specific diagnosis, should raise a high suspicion of FOLR1 mutation, even in absence of seizures. Since folate transporter deficiency is a treatable neurodegenerative disorder, early diagnosis and supplementation with folinic acid is vital.


Asunto(s)
Deficiencia de Ácido Fólico/patología , Síndromes de Malabsorción/patología , Fenotipo , Encéfalo/metabolismo , Niño , Femenino , Receptor 1 de Folato/genética , Deficiencia de Ácido Fólico/diagnóstico por imagen , Deficiencia de Ácido Fólico/genética , Humanos , Síndromes de Malabsorción/diagnóstico por imagen , Síndromes de Malabsorción/genética , Mutación
3.
Environ Mol Mutagen ; 59(5): 366-374, 2018 06.
Artículo en Inglés | MEDLINE | ID: mdl-29668043

RESUMEN

Folate deficiency causes megaloblastic anemia and neural tube defects, and is also associated with some cancers. In vitro, folate deficiency increases mutation frequency and genome instability, as well as exacerbates the mutagenic potential of known environmental mutagens. Conversely, it remains unclear whether or not elevated folic acid (FA) intakes are beneficial or detrimental to the induction of DNA mutations and by proxy human health. We used the MutaMouse transgenic model to examine the in vivo effects of FA deficient, control, and supplemented diets on somatic DNA mutant frequency (MF) and genome instability in hematopoietic cells. We also examined the interaction between FA intake and exposure to the known mutagen N-ethyl-N-nitrosourea (ENU) on MF. Male mice were fed the experimental diets for 20 weeks from weaning. Half of the mice from each diet group were gavaged with 50 mg/kg body weight ENU after 10 weeks on diet and remained on their respective diet for an additional 10 weeks. Mice fed a FA-deficient diet had a 1.3-fold increase in normochromatic erythrocyte micronucleus (MN) frequency (P = 0.034), and a doubling of bone marrow lacZ MF (P = 0.035), compared to control-fed mice. Mice exposed to ENU showed significantly higher bone marrow lacZ and Pig-a MF, but there was no effect of FA intake on ENU-induced MF. These data indicate that FA deficiency increases mutations and MN formation in highly proliferative somatic cells, but that FA intake does not mitigate ENU-induced mutations. Also, FA intake above adequacy had no beneficial or detrimental effect on mutations or MN formation. Environ. Mol. Mutagen. 59:366-374, 2018. © 2018 Her Majesty the Queen in Right of Canada 2018.


Asunto(s)
Anemia Megaloblástica/genética , Deficiencia de Ácido Fólico/genética , Ácido Fólico/genética , Células Madre Hematopoyéticas/efectos de los fármacos , Anemia Megaloblástica/inducido químicamente , Anemia Megaloblástica/metabolismo , Anemia Megaloblástica/patología , Animales , Daño del ADN/efectos de los fármacos , Suplementos Dietéticos , Etilnitrosourea/toxicidad , Femenino , Ácido Fólico/metabolismo , Deficiencia de Ácido Fólico/metabolismo , Deficiencia de Ácido Fólico/patología , Inestabilidad Genómica/efectos de los fármacos , Células Madre Hematopoyéticas/patología , Humanos , Operón Lac/efectos de los fármacos , Masculino , Ratones , Ratones Transgénicos , Mutagénesis/efectos de los fármacos , Mutágenos/toxicidad , Mutación/efectos de los fármacos , Defectos del Tubo Neural/genética , Defectos del Tubo Neural/metabolismo , Defectos del Tubo Neural/patología
4.
Congenit Anom (Kyoto) ; 57(5): 138-141, 2017 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-28603928

RESUMEN

Folate functions as a coenzyme to transfer one-carbon units that are necessary for deoxythymidylate synthesis, purine synthesis, and various methylation reactions. Ingested folate becomes a functional molecule through intestinal absorption, circulation, transport to cells, and various modifications to its structure. Associations between nutritional folate status and chronic diseases such as cardiovascular disease, cancer, and cognitive dysfunction have been reported. It has also been reported that maternal folate nutritional status is related to the risk of neural tube defects (NTDs) in the offspring. It has also been recommended that folate be consumed in the diet to promote the maintenance of good health. To reduce the risk of NTDs, supplementation with folic acid (a synthetic form of folate) during the periconceptional period has also been recommended. This paper describes the basic features and nutritional role of folate.


Asunto(s)
Suplementos Dietéticos , Deficiencia de Ácido Fólico/prevención & control , Ácido Fólico/metabolismo , Defectos del Tubo Neural/prevención & control , Tubo Neural/metabolismo , Embrión de Mamíferos , Femenino , Feto , Ácido Fólico/administración & dosificación , Deficiencia de Ácido Fólico/diagnóstico , Deficiencia de Ácido Fólico/metabolismo , Deficiencia de Ácido Fólico/patología , Humanos , Redes y Vías Metabólicas/fisiología , Tubo Neural/anomalías , Tubo Neural/efectos de los fármacos , Tubo Neural/embriología , Defectos del Tubo Neural/diagnóstico , Defectos del Tubo Neural/metabolismo , Defectos del Tubo Neural/patología , Ingesta Diaria Recomendada
5.
Birth Defects Res ; 109(2): 81-91, 2017 01 30.
Artículo en Inglés | MEDLINE | ID: mdl-28008752

RESUMEN

BACKGROUND: Periconception maternal nutrition and folate in particular are important factors influencing the incidence of neural tube defects (NTDs). Many but not all NTDs are prevented by folic acid supplementation and there is a pressing need for additional strategies to prevent these birth defects. Other micronutrients such as iron are potential candidates, yet a clear role for iron deficiency in contributing to NTDs is lacking. Our previous studies with the flatiron (ffe) mouse model of Ferroportin1 (Fpn1) deficiency suggest that iron is required for neural tube closure and forebrain development raising the possibility that iron supplementation could prevent NTDs. METHODS: We determined the effect of periconception iron and/or folic acid supplementation on the penetrance of NTDs in the Fpn1ffe mouse model. Concurrently, measurements of folate and iron were made to ensure supplementation had the intended effects. RESULTS: High levels of iron supplementation significantly reduced the incidence of NTDs in Fpn1ffe mutants. Fpn1 deficiency resulted in reduced folate levels in both pregnant dams and embryos. Yet folic acid supplementation did not prevent NTDs in the Fpn1ffe model. Similarly, forebrain truncations were rescued with iron. Surprisingly, the high levels of iron supplementation used in this study caused folate deficiency in wild-type dams and embryos. CONCLUSION: Our results demonstrate that iron supplementation can prevent NTDs and forebrain truncations in the Fpn1ffe model. Surprisingly, high levels of iron supplementation and iron overload can cause folate deficiency. If iron is essential for neural tube closure, it is possible that iron deficiency might contribute to NTDs. Birth Defects Research 109:81-91, 2017. © 2016 The Authors Birth Defects Research Published by Wiley Periodicals, Inc.


Asunto(s)
Proteínas de Transporte de Catión/genética , Suplementos Dietéticos , Deficiencia de Ácido Fólico/dietoterapia , Ácido Fólico/administración & dosificación , Hierro/administración & dosificación , Defectos del Tubo Neural/prevención & control , Animales , Proteínas de Transporte de Catión/deficiencia , Cruzamientos Genéticos , Modelos Animales de Enfermedad , Embrión de Mamíferos , Femenino , Deficiencia de Ácido Fólico/genética , Deficiencia de Ácido Fólico/metabolismo , Deficiencia de Ácido Fólico/patología , Eliminación de Gen , Humanos , Fenómenos Fisiologicos Nutricionales Maternos , Ratones , Ratones Transgénicos , Tubo Neural/anomalías , Tubo Neural/efectos de los fármacos , Tubo Neural/metabolismo , Defectos del Tubo Neural/genética , Defectos del Tubo Neural/metabolismo , Defectos del Tubo Neural/patología , Penetrancia , Embarazo , Prosencéfalo/anomalías , Prosencéfalo/efectos de los fármacos , Prosencéfalo/metabolismo
6.
Birth Defects Res ; 109(2): 68-80, 2017 01 30.
Artículo en Inglés | MEDLINE | ID: mdl-27324558

RESUMEN

Susceptibility to neural tube defects (NTDs), such as anencephaly and spina bifida is influenced by genetic and environmental factors including maternal nutrition. Maternal periconceptional supplementation with folic acid significantly reduces the risk of an NTD-affected pregnancy, but does not prevent all NTDs, and "folic acid non-responsive" NTDs continue to occur. Similarly, among mouse models of NTDs, some are responsive to folic acid but others are not. Among nutritional factors, inositol deficiency causes cranial NTDs in mice while supplemental inositol prevents spinal and cranial NTDs in the curly tail (Grhl3 hypomorph) mouse, rodent models of hyperglycemia or induced diabetes, and in a folate-deficiency induced NTD model. NTDs also occur in mice lacking expression of certain inositol kinases. Inositol-containing phospholipids (phosphoinositides) and soluble inositol phosphates mediate a range of functions, including intracellular signaling, interaction with cytoskeletal proteins, and regulation of membrane identity in trafficking and cell division. Myo-inositol has been trialed in humans for a range of conditions and appears safe for use in human pregnancy. In pilot studies in Italy and the United Kingdom, women took inositol together with folic acid preconceptionally, after one or more previous NTD-affected pregnancies. In nonrandomized cohorts and a randomized double-blind study in the United Kingdom, no recurrent NTDs were observed among 52 pregnancies reported to date. Larger-scale fully powered trials are needed to determine whether supplementation with inositol and folic acid would more effectively prevent NTDs than folic acid alone. Birth Defects Research 109:68-80, 2017. © 2016 The Authors Birth Defects Research Published by Wiley Periodicals, Inc.


Asunto(s)
Suplementos Dietéticos , Deficiencia de Ácido Fólico/prevención & control , Ácido Fólico/administración & dosificación , Inositol/administración & dosificación , Defectos del Tubo Neural/prevención & control , Tubo Neural/efectos de los fármacos , Animales , Ensayos Clínicos como Asunto , Modelos Animales de Enfermedad , Femenino , Deficiencia de Ácido Fólico/genética , Deficiencia de Ácido Fólico/metabolismo , Deficiencia de Ácido Fólico/patología , Humanos , Fosfatos de Inositol/metabolismo , Fenómenos Fisiologicos Nutricionales Maternos , Ratones , Tubo Neural/anomalías , Tubo Neural/metabolismo , Defectos del Tubo Neural/genética , Defectos del Tubo Neural/metabolismo , Defectos del Tubo Neural/patología , Fosfatidilinositoles/metabolismo , Embarazo
7.
Int J Biochem Cell Biol ; 79: 222-230, 2016 10.
Artículo en Inglés | MEDLINE | ID: mdl-27592453

RESUMEN

Folate deficiency contributes to impaired adult hippocampal neurogenesis, yet the mechanisms remain unclear. Here we use HT-22 hippocampal neuron cells as model to investigate the effect of folate deprivation (FD) on cell proliferation and apoptosis, and to elucidate the underlying mechanism. FD caused cell cycle arrest at G0/G1 phase and increased the rate of apoptosis, which was associated with disrupted expression of folate transport and methyl transfer genes. FOLR1 and SLC46A1 were (P<0.01) down-regulated, while SLC19A1 was up-regulated (P<0.01) in FD group. FD cells exhibited significantly (P<0.05) higher protein content of BHMT, MAT2b and DNMT3a, as well as increased SAM/SAH concentrations and global DNA hypermethylation. The expression of the total and all the 3 classes of IGF-1 mRNA variants was significantly (P<0.01) down-regulated and IGF-1 concentration was decreased (P<0.05) in the culture media. IGF-1 signaling pathway was also compromised with diminished activation (P<0.05) of STAT3, AKT and mTOR. CpG hypermethylation was detected in the promoter regions of IGF-1 and FOLR1 genes, while higher SLC19A1 mRNA corresponded to hypomethylation of its promoter. IGF-1 supplementation in FD media significantly abolished FD-induced decrease in cell viability. However, IGF-1 had limited effect in rescuing the cell phenotype when added 24h after FD. Taken together, down-regulation of IGF-1 expression and signaling is involved in FD-induced cell cycle arrest and apoptosis in HT-22 hippocampal neuron cells, which is associated with an abnormal activation of methyl transfer pathway and hypermethylation of IGF-1 gene promoter.


Asunto(s)
Apoptosis , Deficiencia de Ácido Fólico/patología , Puntos de Control de la Fase G1 del Ciclo Celular , Hipocampo/patología , Factor I del Crecimiento Similar a la Insulina/metabolismo , Neuronas/patología , Fase de Descanso del Ciclo Celular , Animales , Transporte Biológico , Línea Celular , Proliferación Celular , Supervivencia Celular , Islas de CpG/genética , Metilación de ADN , Regulación hacia Abajo , Epigénesis Genética , Receptor 1 de Folato/genética , Ácido Fólico/metabolismo , Deficiencia de Ácido Fólico/genética , Deficiencia de Ácido Fólico/metabolismo , Transportadores de Ácido Fólico/genética , Ratones , Regiones Promotoras Genéticas/genética , Proteína Portadora de Folato Reducido/genética , Transducción de Señal
8.
Lab Med ; 47(3): 259-62, 2016 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-27406144

RESUMEN

The clinical presentation of celiac disease has evolved from chronic diarrhea and malnutrition to mild nutrient insufficiencies. Recently diagnosed adults with celiac disease should be assessed for micronutrient deficiencies because early institution of a gluten-free diet (GFD) prevents morbidity and reduces the incidence of gastrointestinal malignant neoplasms and osteoporosis. In this report, we present the case of a 49-year-old woman of Southeast Asian-Indian descent living in the United States who had folate insufficiency, as manifested by low serum and red blood cell (RBC) folate levels. Further investigation, including serologic testing and intestinal biopsy, confirmed a diagnosis of celiac disease and other nutrient deficiencies. Managing the condition of this patient with folate supplements and implementation of a recommended GFD reversed the folate insufficiency. In conclusion, when serum and/or RBC levels are low in a person of Southeast Asian-Indian descent living in a country with folate fortification of the grain supply, such as the United States, the medical team needs to look for an organic cause, as in our patient, to diagnose and manage celiac disease early and, hopefully, forestall complications.


Asunto(s)
Enfermedad Celíaca/complicaciones , Deficiencia de Ácido Fólico/diagnóstico , Deficiencia de Ácido Fólico/patología , Pueblo Asiatico , Enfermedad Celíaca/terapia , Dieta/métodos , Femenino , Deficiencia de Ácido Fólico/terapia , Humanos , Persona de Mediana Edad , Estados Unidos
9.
Oncotarget ; 7(22): 33246-56, 2016 May 31.
Artículo en Inglés | MEDLINE | ID: mdl-27119349

RESUMEN

Clinically, serum level of folate has been negatively correlated to the stage and progression of liver cancer. Nevertheless, the functional consequence of folate deficiency (FD) in malignancy has not been fully investigated. Human hepatocellular carcinoma (HCC) cells (as study model) and other cancer types such as lung and glioma were cultured under folate deficient (FD) and folate complete (FD) conditions. Molecular characterization including intracellular ROS/RNS (reactive oxygen/nitrogen species), viability, colony formation, cancer stem-like cell (CSC) phenotype analyses were performed. In vivo tumorigenesis under FD and FC conditions were also examined. FD induced a significant increase in ROS and RNS, suppressing proliferative ability but inducing metastatic potential. Mesenchymal markers such as Snail, ZEB2, and Vimentin were significantly up-regulated while E-cadherin down-regulated. Importantly, CSC markers such as Oct4, ß-catenin, CD133 were induced while PRRX1 decreased under FD condition. Furthermore, FD-conditioned HCC cells showed a decreased miR-22 level, leading to the increased expression of its target genes including HDAC4, ZEB2 and Oct4. Finally, xenograft mouse model demonstrated that FD diet promoted tumorigenesis and metastasis as compared to their FC counterparts. Our data provides rationales for the consideration of folate supplement as a metastasis preventive measure.


Asunto(s)
Transición Epitelial-Mesenquimal , Deficiencia de Ácido Fólico/metabolismo , Ácido Fólico/metabolismo , Neoplasias/metabolismo , Células Madre Neoplásicas/metabolismo , Microambiente Tumoral , Animales , Línea Celular Tumoral , Proliferación Celular , Supervivencia Celular , Deficiencia de Ácido Fólico/genética , Deficiencia de Ácido Fólico/patología , Regulación Neoplásica de la Expresión Génica , Humanos , Ratones Endogámicos NOD , Ratones SCID , MicroARNs/genética , MicroARNs/metabolismo , Neoplasias/genética , Neoplasias/patología , Células Madre Neoplásicas/patología , Estrés Nitrosativo , Estrés Oxidativo , Fenotipo , Especies de Nitrógeno Reactivo/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Transducción de Señal , Factores de Tiempo
10.
Biochimie ; 126: 31-42, 2016 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-26924398

RESUMEN

Folates are essential in the intermediary metabolism of amino acids, synthesis of nucleotides and for maintaining methylation reactions. They are also linked to the production of neurotransmitters through GTP needed for the synthesis of tetrahydrobiopterin. During pregnancy, folate is needed for fetal development. Folate deficiency during this period has been linked to increased risk of neural tube defects. Disturbances of folate metabolism due to genetic abnormalities or the presence of autoantibodies to folate receptor alpha (FRα) can impair physiologic processes dependent on folate, resulting in a variety of developmental disorders including cerebral folate deficiency syndrome and autism spectrum disorders. Overall, adequate folate status has proven to be important during pregnancy as well as neurological development and functioning in neonates and children. Treatment with pharmacologic doses of folinic acid has led to reversal of some symptoms in many children diagnosed with cerebral folate deficiency syndrome and autism, especially in those positive for autoantibodies to FRα. Thus, as the brain continues to develop throughout fetal and infant life, it can be affected and become dysfunctional due to a defective folate transport contributing to folate deficiency. Treatment and prevention of these disorders can be achieved by identification of those at risk and supplementation with folinic acid.


Asunto(s)
Trastorno Autístico , Deficiencia de Ácido Fólico , Ácido Fólico , Defectos del Tubo Neural , Animales , Trastorno Autístico/genética , Trastorno Autístico/metabolismo , Trastorno Autístico/patología , Autoanticuerpos/metabolismo , Transporte Biológico Activo/genética , Femenino , Receptor 1 de Folato/antagonistas & inhibidores , Receptor 1 de Folato/genética , Receptor 1 de Folato/metabolismo , Deficiencia de Ácido Fólico/genética , Deficiencia de Ácido Fólico/metabolismo , Deficiencia de Ácido Fólico/patología , Humanos , Defectos del Tubo Neural/genética , Defectos del Tubo Neural/metabolismo , Defectos del Tubo Neural/patología , Embarazo
11.
Birth Defects Res A Clin Mol Teratol ; 106(3): 201-7, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26900104

RESUMEN

BACKGROUND: Periconceptional supplementation with folic acid results in a significant reduction in the incidence of neural tube defects (NTDs). Nonetheless, NTDs remain a leading cause of perinatal morbidity and mortality worldwide, and the mechanism(s) by which folate exerts its protective effects are unknown. Homocysteine is an amino acid that accumulates under conditions of folate-deficiency, and is suggested as a risk factor for NTDs. One proposed mechanism of homocysteine toxicity is its accumulation into proteins in a process termed homocysteinylation. METHODS & RESULTS: Herein, we used a folate-deficient diet in pregnant mice to demonstrate that there is: (i) a significant inverse correlation between maternal serum folate levels and serum homocysteine; (ii) a significant positive correlation between serum homocysteine levels and titers of autoantibodies against homocysteinylated protein; and (iii) a significant increase in congenital malformations and NTDs in mice deficient in serum folate. Furthermore, in mice administered the folate-deplete diet before conception, supplementation with folic acid during the gestational period completely rescued the embryos from congenital defects, and resulted in homocysteinylated protein titers at term that are comparable to that of mice administered a folate-replete diet throughout both the pre- and postconception period. These results demonstrate that a low-folate diet that induces NTDs also increases protein homocysteinylation and the subsequent generation of autoantibodies against homocysteinylated proteins. CONCLUSION: These data support the hypotheses that homocysteinylation results in neo-self antigen formation under conditions of maternal folate deficiency, and that this process is reversible with folic acid supplementation.


Asunto(s)
Autoanticuerpos/sangre , Proteínas Sanguíneas/metabolismo , Deficiencia de Ácido Fólico/complicaciones , Ácido Fólico/sangre , Homocisteína/química , Defectos del Tubo Neural/etiología , Animales , Proteínas Sanguíneas/inmunología , Dieta , Suplementos Dietéticos , Modelos Animales de Enfermedad , Femenino , Ácido Fólico/administración & dosificación , Ácido Fólico/inmunología , Deficiencia de Ácido Fólico/sangre , Deficiencia de Ácido Fólico/inmunología , Deficiencia de Ácido Fólico/patología , Edad Gestacional , Homocisteína/biosíntesis , Humanos , Ratones , Ratones Endogámicos C57BL , Defectos del Tubo Neural/sangre , Defectos del Tubo Neural/inmunología , Defectos del Tubo Neural/patología , Embarazo , Procesamiento Proteico-Postraduccional
12.
J Inherit Metab Dis ; 38(5): 863-72, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-25633902

RESUMEN

In the folate cycle MTHFD1, encoded by MTHFD1, is a trifunctional enzyme containing 5,10-methylenetetrahydrofolate dehydrogenase, 5,10-methenyltetrahydrofolate cyclohydrolase and 10-formyltetrahydrofolate synthetase activity. To date, only one patient with MTHFD1 deficiency, presenting with hyperhomocysteinemia, megaloblastic anaemia, hemolytic uremic syndrome (HUS) and severe combined immunodeficiency, has been identified (Watkins et al J Med Genet 48:590-2, 2011). We now describe four additional patients from two different families. The second patient presented with hyperhomocysteinemia, megaloblastic anaemia, HUS, microangiopathy and retinopathy; all except the retinopathy resolved after treatment with hydroxocobalamin, betaine and folinic acid. The third patient developed megaloblastic anaemia, infection, autoimmune disease and moderate liver fibrosis but not hyperhomocysteinemia, and was successfully treated with a regime that included and was eventually reduced to folic acid. The other two, elder siblings of the third patient, died at 9 weeks of age with megaloblastic anaemia, infection and severe acidosis and had MTFHD1 deficiency diagnosed retrospectively. We identified a missense mutation (c.806C > T, p.Thr296Ile) and a splice site mutation (c.1674G > A) leading to exon skipping in the second patient, while the other three harboured a missense mutation (c.146C > T, p.Ser49Phe) and a premature stop mutation (c.673G > T, p.Glu225*), all of which were novel. Patient fibroblast studies revealed severely reduced methionine formation from [(14)C]-formate, which did not increase in cobalamin supplemented culture medium but was responsive to folic and folinic acid. These additional cases increase the clinical spectrum of this intriguing defect, provide in vitro evidence of disturbed methionine synthesis and substantiate the effectiveness of folic or folinic acid treatment.


Asunto(s)
Ácido Fólico/uso terapéutico , Leucovorina/uso terapéutico , Metilenotetrahidrofolato Deshidrogenasa (NADP)/deficiencia , Metilenotetrahidrofolato Deshidrogenasa (NADP)/genética , Anemia Megaloblástica/tratamiento farmacológico , Anemia Megaloblástica/genética , Anemia Megaloblástica/patología , Células Cultivadas , Resultado Fatal , Femenino , Deficiencia de Ácido Fólico/tratamiento farmacológico , Deficiencia de Ácido Fólico/genética , Deficiencia de Ácido Fólico/patología , Humanos , Hiperhomocisteinemia/tratamiento farmacológico , Hiperhomocisteinemia/genética , Hiperhomocisteinemia/patología , Lactante , Recién Nacido , Masculino , Antígenos de Histocompatibilidad Menor , Inmunodeficiencia Combinada Grave/tratamiento farmacológico , Inmunodeficiencia Combinada Grave/genética , Inmunodeficiencia Combinada Grave/patología , Adulto Joven
13.
Brain Dev ; 37(1): 163-7, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24534056

RESUMEN

BACKGROUND: Hereditary folate malabsorption is a rare, autosomal recessive disorder of proton-coupled folate transporter deficiency resulting in folate deficiency. Left untreated, the condition can cause severe brain damage and megaloblastic anemia, leading to progressive psychomotor retardation, seizures and other neurological problems. Early diagnosis and treatment are crucial. No case has been documented yet in Mainland China until now. METHODS: A Chinese girl affected by hereditary folate malabsorption was studied. The girl presented with recurrent megaloblastic anemia from the age of 7 months. Paroxysmal limbs trembling and seizures were presented from the age of three years. Intracranial calcification was noted by CT. At her age of 5 years, mental regression, lower-extremity weakness and sleeping problems were observed. Her plasma folate decreased to 4.49 nmol/L (normal control>6.8nmol/L). Plasma total homocysteine elevated to 28.11 µmol/L (normal control<15 µmol/L). Folate and 5-methylterahydrofolate in cerebrospinal fluid were significantly decreased to undetectable level. RESULTS: On SLC46A1 gene, a novel mutation, c.1A>T (M1L), and a reported mutation c.194-195 insG (p.Cys66LeufsX99) were identified, supported the diagnosis of hereditary folate malabsorption. Each parent carries one of two mutations. Folinic calcium supplement resulted in rapid clinical improvement. She is currently 6 years old with normal development and routine blood features. CONCLUSION: Hereditary folate malabsorption is one of the few easily-treatable inherited metabolic diseases. Measurements of folate and 5-methyltetrahydrofolate in cerebrospinal fluid are keys for the diagnosis of the patients.


Asunto(s)
Deficiencia de Ácido Fólico/genética , Síndromes de Malabsorción/genética , Transportador de Folato Acoplado a Protón/genética , Edad de Inicio , Pueblo Asiatico , Preescolar , Femenino , Ácido Fólico/uso terapéutico , Deficiencia de Ácido Fólico/complicaciones , Deficiencia de Ácido Fólico/patología , Humanos , Síndromes de Malabsorción/complicaciones , Síndromes de Malabsorción/patología , Mutación Puntual , Convulsiones/etiología
14.
Neurobiol Dis ; 71: 234-44, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-25131448

RESUMEN

Folate is a nutrient essential for the development, function and regeneration of nervous systems. Folate deficiency has been linked to many neurological disorders including neural tube defects in fetus and Alzheimer's diseases in the elderly. However, the etiology underlying these folate deficiency-associated diseases is not completely understood. In this study, zebrafish transgenic lines with timing and duration-controllable folate deficiency were developed by ectopically overexpressing a recombinant EGFP-γ-glutamyl hydrolase (γGH). Impeded neural crest cell migration was observed in the transgenic embryos when folate deficiency was induced in early stages, leading to defective neural tube closure and hematopoiesis. Adding reduced folate or N-acetylcysteine reversed the phenotypic anomalies, supporting the causal link between the increased oxidative stress and the folate deficiency-induced abnormalities. When folate deficiency was induced in aged fish accumulation of beta-amyloid and phosphorylated Tau protein were found in the fish brain cryo-sections. Increased autophagy and accumulation of acidic autolysosome were apparent in folate deficient neuroblastoma cells, which were reversed by reduced folate or N-acetylcysteine supplementation. Decreased expression of cathepsin B, a lysosomal protease, was also observed in cells and tissue with folate deficiency. We concluded that folate deficiency-induced oxidative stress contributed to the folate deficiency-associated neuropathogenesis in both early and late stages of life.


Asunto(s)
Envejecimiento/genética , Enfermedad de Alzheimer/etiología , Deficiencia de Ácido Fólico , Defectos del Tubo Neural/etiología , Estrés Oxidativo/genética , Acetilcisteína/metabolismo , Acetilcisteína/farmacología , Enfermedad de Alzheimer/genética , Animales , Animales Modificados Genéticamente , Catepsina B/genética , Catepsina B/metabolismo , Movimiento Celular/genética , Embrión no Mamífero , Ácido Fólico/metabolismo , Deficiencia de Ácido Fólico/complicaciones , Deficiencia de Ácido Fólico/genética , Deficiencia de Ácido Fólico/patología , Proteínas Fluorescentes Verdes/genética , Calor/efectos adversos , Proteínas Asociadas a Microtúbulos/metabolismo , Cresta Neural/fisiología , Defectos del Tubo Neural/genética , Estrés Oxidativo/efectos de los fármacos , Factores de Tiempo , Pez Cebra , Proteínas de Pez Cebra/genética , Proteínas de Pez Cebra/metabolismo , gamma-Glutamil Hidrolasa/metabolismo
15.
Mol Nutr Food Res ; 58(6): 1190-8, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24585430

RESUMEN

SCOPE: Fumonisin B1 (FB1 ) is found in corn-based foods and is a possible risk factor for neural tube defects (NTDs). The mechanism(s) underlying NTD induction by FB1 in LM/Bc mice is not understood; however, evidence suggests disrupted folate transport is involved. METHODS AND RESULTS: Female LM/Bc mice were fed folate-sufficient (control) or folate-deficient diet beginning 5 wk before mating, treated with 0 (vehicle), 2.5 or 10 mg/kg FB1 by intraperitoneal injection on embryonic days 7 (E7) and E8, and their fetuses examined on E16. Dose-dependent NTD induction was found in groups fed the control diet: 3 of 13 low-dose and 10 of 11 high-dose litters were affected. Among groups fed folate-deficient diet, NTDs were found only in 4 of 11 high-dose litters. In another trial, consumption of folate-deficient diet also resulted in fewer NTDs at a dose of 10 mg/kg FB1 and reduced maternal red blood cell folate levels by 80%. In utero death did not fully account for the differences in NTD rates. CONCLUSION: Folate deficiency does not exacerbate NTD induction by FB1 in LM/Bc mice. Interactions between folate, other nutritional factors, and FB1 in this mouse model for NTDs are complex and require further investigation.


Asunto(s)
Ácido Fólico/sangre , Fumonisinas/toxicidad , Fenómenos Fisiologicos Nutricionales Maternos , Defectos del Tubo Neural/patología , Animales , Dieta , Modelos Animales de Enfermedad , Eritrocitos/química , Femenino , Ácido Fólico/administración & dosificación , Deficiencia de Ácido Fólico/patología , Ratones , Ratones Endogámicos , Defectos del Tubo Neural/inducido químicamente
16.
Nutr Cancer ; 65(6): 857-65, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23909730

RESUMEN

Hereditary nevoid basal cell carcinoma syndrome (NBCCS) is caused by PTCH1 gene mutations that result in diverse neoplasms including medulloblastoma (MB). Epidemiological studies report reduced pediatric brain tumor risks associated with maternal intake of prenatal vitamins containing folic acid (FA) and FA supplements specifically. We hypothesized that low maternal FA intake during the perigestational period would increase MB incidence in a transgenic NBCCS mouse model, which carries an autosomal dominant mutation in the Ptch1 gene. Female wild-type C57BL/6 mice (n = 126) were randomized to 1 of 3 diets with differing FA amounts: 0.3 mg/kg (low), 2.0 mg/kg (control), and 8.0 mg/kg (high) 1 mo prior to mating with Ptch1 (+/-) C57BL/6 males. Females were maintained on the diet until pup weaning; the pups were then aged for tumor development. Compared to the control group, offspring MB incidence was significantly lower in the low FA group (Hazard Ratio = 0.47; 95% confidence interval 0.27-0.80) at 1 yr. No significant difference in incidence was observed between the control and high FA groups. Low maternal perigestational FA levels may decrease MB incidence in mice genetically predisposed to tumor development. Our results could have implications for prenatal FA intake recommendations in the presence of cancer syndromes.


Asunto(s)
Síndrome del Nevo Basocelular/tratamiento farmacológico , Suplementos Dietéticos , Deficiencia de Ácido Fólico/patología , Ácido Fólico/administración & dosificación , Fenómenos Fisiologicos Nutricionales Maternos , Meduloblastoma/tratamiento farmacológico , Receptores de Superficie Celular/genética , Animales , Síndrome del Nevo Basocelular/complicaciones , Síndrome del Nevo Basocelular/genética , Modelos Animales de Enfermedad , Femenino , Deficiencia de Ácido Fólico/complicaciones , Deficiencia de Ácido Fólico/tratamiento farmacológico , Predisposición Genética a la Enfermedad , Masculino , Meduloblastoma/complicaciones , Meduloblastoma/genética , Ratones , Ratones Endogámicos C57BL , Mutación , Receptores Patched , Receptor Patched-1 , Embarazo , Receptores de Superficie Celular/metabolismo
17.
J Nutr ; 143(7): 1028-35, 2013 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-23700346

RESUMEN

Impaired folate-mediated one-carbon metabolism (OCM) has emerged as a risk factor for several diseases associated with age-related cognitive decline, but the underlying mechanisms remain unknown and thus hinder the identification of subpopulations most vulnerable to OCM disruption. Here we investigated the role of serine hydroxymethyltransferase 1 (SHMT1), a folate-dependent enzyme regulating de novo thymidylate biosynthesis, in influencing neuronal and cognitive function in the adult mouse. We observed Shmt1 expression in the hippocampus, including the granule cell layer of the dentate gyrus (DG), and examined hippocampal neurogenesis and hippocampal-dependent fear conditioning in mice deficient for Shmt1. We used a 3 × 3 design in which adult male Shmt1(+/+), Shmt1(+/-), and Shmt1(-/-) mice were fed folic acid control (2 mg/kg), folic acid-deficient (0 mg/kg), or folic acid-supplemented (8 mg/kg) diets from weaning through the duration of the study. Proliferation within the DG was elevated by 70% in Shmt1(+/-) mice, yet the number of newborn mature neurons was reduced by 98% compared with that in Shmt1(+/+) mice. Concomitant with these alterations, Shmt1(+/-) mice showed a 45% reduction in mnemonic recall during trace fear conditioning. Dietary folate manipulations alone did not influence neural outcomes. Together, these data identify SHMT1 as one of the first enzymes within the OCM pathway to regulate neuronal and cognitive profiles and implicate impaired thymidylate biosynthesis in the etiology of folate-related neuropathogenesis.


Asunto(s)
Glicina Hidroximetiltransferasa/genética , Hipocampo/patología , Memoria/fisiología , Neurogénesis/genética , Animales , Proliferación Celular , Giro Dentado/metabolismo , Miedo/fisiología , Ácido Fólico/administración & dosificación , Ácido Fólico/sangre , Deficiencia de Ácido Fólico/patología , Regulación de la Expresión Génica , Glicina Hidroximetiltransferasa/metabolismo , Hipocampo/metabolismo , Homocisteína/metabolismo , Hibridación in Situ , Masculino , Ratones , Ratones Noqueados , Neuronas/citología , Neuronas/metabolismo , Factores de Riesgo , Timidina Monofosfato/biosíntesis
18.
Clin Chem Lab Med ; 51(3): 555-69, 2013 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-23449524

RESUMEN

Folate deficiency has been linked to diverse clinical manifestations and despite the importance of accurate assessment of folate status, the best test for routine use is uncertain. Both serum and red cell folate assays are widely available in clinical laboratories; however, red cell folate is the more time-consuming and costly test. This review sought to evaluate whether the red cell assay demonstrated superior performance characteristics to justify these disadvantages. Red cell folate, but not serum folate, measurements demonstrated analytical variation due to sample pre-treatment parameters, oxygen saturation of haemoglobin and haematocrit. Neither marker was clearly superior in characterising deficiency but serum folate more frequently showed the higher correlation with homocysteine, a sensitive marker of deficiency. Similarly, both serum and red cell folate were shown to increase in response to folic acid supplementation. However, serum folate generally gave the greater response and was able to distinguish different supplementation doses. The C677T polymorphism of methylenetetrahydrofolate reductase alters the distribution of folate forms in red cells and may thereby cause further analytical variability in routine red cell folate assays. Overall, serum folate is cheaper and faster to perform than red cell folate, is influenced by fewer analytical variables and provides an assessment of folate status that may be superior to red cell folate.


Asunto(s)
Eritrocitos/metabolismo , Deficiencia de Ácido Fólico/sangre , Ácido Fólico/análisis , Cromatografía Líquida de Alta Presión , Suplementos Dietéticos , Ácido Fólico/sangre , Ácido Fólico/uso terapéutico , Deficiencia de Ácido Fólico/tratamiento farmacológico , Deficiencia de Ácido Fólico/patología , Homocisteína/sangre , Humanos , Metilenotetrahidrofolato Reductasa (NADPH2)/genética , Metilenotetrahidrofolato Reductasa (NADPH2)/metabolismo , Polimorfismo de Nucleótido Simple , Espectrometría de Masas en Tándem
19.
J Nutr ; 143(1): 27-33, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-23190761

RESUMEN

Low folate status is a risk factor for colon carcinogenesis; mechanisms proposed to account for this relationship include uracil misincorporation into DNA and global DNA hypomethylation. We investigated whether such biomarkers are related to folate status in isolated colonocytes from colonoscopy patients. In cases with adenomatous polyps (n = 40) or hyperplastic polyps (n = 16), colonocytes were isolated from biopsies from the polyp, from a site adjacent to the polyp, and from normal mucosa 10-15 cm distal to the polyp. In polyp-free controls (n = 53), biopsies were taken from ascending, transverse, and descending areas of colon. Within adenoma cases, there was a trend (P-trend < 0.001) of decreasing colonocyte folate (pg/105 cells, mean ± CI) from the site distal to the polyp (16.9 ± 2.4), to the site adjacent to the polyp (14.7 ± 2.3), to the polyp (12.8 ± 2.0). Correspondingly, there were increases in uracil misincorporation (P-trend < 0.001) and global DNA hypomethylation (P-trend = 0.012) across the 3 sites. Colonocyte folate concentrations were significantly correlated with RBC folate concentrations, but only in individuals with generally lower (≤484 µg/L) RBC folate status (r = 0.54; P = 0.006; n = 24), and were also significantly lower in normal mucosa of cases with adenomatous polyps than in controls matched for colonic segment. In conclusion, localized folate deficiency in specific areas of colon might create carcinogenic fields and affect the development of colorectal polyps through uracil misincorporation and DNA hypomethylation; alternatively, the polyp itself might deplete folate in the surrounding tissue. Folate supplementation trials aimed at colon cancer prevention should target individuals with suboptimal folate status.


Asunto(s)
Disparidad de Par Base , Colon/metabolismo , Pólipos del Colon/metabolismo , Metilación de ADN , Deficiencia de Ácido Fólico/metabolismo , Ácido Fólico/metabolismo , Mucosa Intestinal/metabolismo , Pólipos Adenomatosos/etiología , Pólipos Adenomatosos/metabolismo , Pólipos Adenomatosos/patología , Adulto , Anciano , Biomarcadores/metabolismo , Estudios de Casos y Controles , Colon/patología , Pólipos del Colon/etiología , Pólipos del Colon/patología , ADN/biosíntesis , Daño del ADN , Femenino , Deficiencia de Ácido Fólico/patología , Deficiencia de Ácido Fólico/fisiopatología , Humanos , Hiperplasia , Mucosa Intestinal/patología , Masculino , Persona de Mediana Edad , Especificidad de Órganos , Recto/metabolismo , Recto/patología , Uracilo/metabolismo
20.
Reprod Fertil Dev ; 25(7): 1085-94, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23137932

RESUMEN

Folic acid and vitamin B12 deficiencies are associated with high reproductive risks ranging from infertility to fetal structural defects. The aim of the present study was to examine the effects of preconceptional omega-3 fatty acid supplementation (eicosapentaenoic acid and docosahexaenoic acid) to a micronutrient-deficient diet on the reproductive cycle in Wistar rats. Female rats were divided into five groups from birth and throughout pregnancy: a control group, a folic acid-deficient (FD) group, a vitamin B12-deficient (BD) group, a folic acid-deficient + omega-3 fatty acid-supplemented (FDO) group and a vitamin B12 deficient + omega-3 fatty acid-supplemented (BDO) group. Dams were killed on gestation Day 20 and their ovaries and mammary glands were dissected out and subjected to histological examination. Maternal micronutrient deficiency (FD and BD groups) resulted in an abnormal oestrous cycle (P<0.001), whereas omega-3 fatty acid supplementation (FDO and BDO groups) restored the oestrous cycle to normal. There were fewer corpora lutea in the ovaries of FD rats compared with controls. In addition, rats in both the FD and BD groups exhibited an absence of lactating ducts in their mammary glands compared with controls. The findings of the present study indicate, for the first time, that maternal micronutrient deficiency affects the oestrous cycle and morphology of the ovary and mammary glands. Omega-3 fatty acid supplementation ameliorated these effects. This may have implications for infertility and pregnancy outcomes.


Asunto(s)
Dieta , Ácidos Grasos Omega-3/administración & dosificación , Micronutrientes/deficiencia , Atención Preconceptiva/métodos , Reproducción/efectos de los fármacos , Animales , Cuerpo Lúteo/patología , Suplementos Dietéticos , Ácidos Docosahexaenoicos/administración & dosificación , Ácido Eicosapentaenoico/administración & dosificación , Ciclo Estral/efectos de los fármacos , Femenino , Ácido Fólico/administración & dosificación , Deficiencia de Ácido Fólico/patología , Lactancia , Glándulas Mamarias Animales/patología , Micronutrientes/administración & dosificación , Tamaño de los Órganos , Embarazo , Resultado del Embarazo , Ratas , Ratas Wistar , Vitamina B 12/administración & dosificación , Deficiencia de Vitamina B 12/patología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA