Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 100
Filtrar
1.
Braz J Med Biol Res ; 54(11): e11396, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34586326

RESUMEN

Current understanding of the genetic factors contributing to the etiology of non-syndromic craniosynostosis (NSC) remains scarce. The present work investigated the presence of variants in ALX4, EFNA4, and TWIST1 genes in children with NSC to verify if variants within these genes may contribute to the occurrence of these abnormal phenotypes. A total of 101 children (aged 45.07±40.94 months) with NSC participated in this cross-sectional study. Parents and siblings of the probands were invited to participate. Medical and family history of craniosynostosis were documented. Biological samples were collected to obtain genomic DNA. Coding exons of human TWIST1, ALX4, and EFNA4 genes were amplified by polymerase chain reaction and Sanger sequenced. Five missense variants were identified in ALX4 in children with bilateral coronal, sagittal, and metopic synostosis. A de novo ALX4 variant, c.799G>A: p.Ala267Thr, was identified in a proband with sagittal synostosis. Three missense variants were identified in the EFNA4 gene in children with metopic and sagittal synostosis. A TWIST1 variant occurred in a child with unilateral coronal synostosis. Variants were predicted to be among the 0.1% (TWIST1, c.380C>A: p. Ala127Glu) and 1% (ALX4, c.769C>T: p.Arg257Cys, c.799G>A: p.Ala267Thr, c.929G>A: p.Gly310Asp; EFNA4, c.178C>T: p.His60Tyr, C.283A>G: p.Lys95Glu, c.349C>A: Pro117Thr) most deleterious variants in the human genome. With the exception of ALX4, c.799G>A: p.Ala267Thr, all other variants were present in at least one non-affected family member, suggesting incomplete penetrance. Thus, these variants may contribute to the development of craniosynostosis, and should not be discarded as potential candidate genes in the diagnosis of this condition.


Asunto(s)
Craneosinostosis , Secuencia de Bases , Niño , Craneosinostosis/genética , Estudios Transversales , Proteínas de Unión al ADN/genética , Familia , Humanos , Mutación Missense/genética , Factores de Transcripción/genética
3.
Braz. j. med. biol. res ; 54(11): e11396, 2021. graf
Artículo en Inglés | LILACS | ID: biblio-1339444

RESUMEN

Current understanding of the genetic factors contributing to the etiology of non-syndromic craniosynostosis (NSC) remains scarce. The present work investigated the presence of variants in ALX4, EFNA4, and TWIST1 genes in children with NSC to verify if variants within these genes may contribute to the occurrence of these abnormal phenotypes. A total of 101 children (aged 45.07±40.94 months) with NSC participated in this cross-sectional study. Parents and siblings of the probands were invited to participate. Medical and family history of craniosynostosis were documented. Biological samples were collected to obtain genomic DNA. Coding exons of human TWIST1, ALX4, and EFNA4 genes were amplified by polymerase chain reaction and Sanger sequenced. Five missense variants were identified in ALX4 in children with bilateral coronal, sagittal, and metopic synostosis. A de novo ALX4 variant, c.799G>A: p.Ala267Thr, was identified in a proband with sagittal synostosis. Three missense variants were identified in the EFNA4 gene in children with metopic and sagittal synostosis. A TWIST1 variant occurred in a child with unilateral coronal synostosis. Variants were predicted to be among the 0.1% (TWIST1, c.380C>A: p. Ala127Glu) and 1% (ALX4, c.769C>T: p.Arg257Cys, c.799G>A: p.Ala267Thr, c.929G>A: p.Gly310Asp; EFNA4, c.178C>T: p.His60Tyr, C.283A>G: p.Lys95Glu, c.349C>A: Pro117Thr) most deleterious variants in the human genome. With the exception of ALX4, c.799G>A: p.Ala267Thr, all other variants were present in at least one non-affected family member, suggesting incomplete penetrance. Thus, these variants may contribute to the development of craniosynostosis, and should not be discarded as potential candidate genes in the diagnosis of this condition.


Asunto(s)
Humanos , Niño , Craneosinostosis/genética , Factores de Transcripción/genética , Secuencia de Bases , Familia , Estudios Transversales , Mutación Missense/genética , Proteínas de Unión al ADN/genética
5.
Hum Reprod ; 26(8): 2232-8, 2011 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-21576080

RESUMEN

BACKGROUND: Folic acid intake during pregnancy can reduce the risk of neural tube defects (NTDs) and perhaps also oral facial clefts. Maternal autoantibodies to folate receptors can impair folic acid binding. We explored the relationship of these birth defects to inhibition of folic acid binding to folate receptor α (FRα), as well as possible effects of parental demographics or prenatal exposures. METHODS: We conducted a nested case-control study within the Norwegian Mother and Child Cohort Study. The study included mothers of children with an NTD (n = 11), cleft lip with or without cleft palate (CL/P, n= 72), or cleft palate only (CPO, n= 27), and randomly selected mothers of controls (n = 221). The inhibition of folic acid binding to FRα was measured in maternal plasma collected around 17 weeks of gestation. On the basis of prior literature, the maternal age, gravidity, education, smoking, periconception folic acid supplement use and milk consumption were considered as potential confounding factors. RESULTS: There was an increased risk of NTDs with increased binding inhibition [adjusted odds ratio (aOR) = 1.4, 95% confidence interval (CI) 1.0-1.8]. There was no increased risk of oral facial clefts from inhibited folic acid binding to FRα (CL/P aOR = 0.7, 95% CI 0.6-1.0; CPO aOR = 1.1, 95% CI 0.8-1.4). No association was seen between smoking, folate supplementation or other cofactors and inhibition of folic acid binding to FRα. CONCLUSIONS: Inhibition of folic acid binding to FRα in maternal plasma collected during pregnancy was associated with increased risk of NTDs but not oral facial clefts.


Asunto(s)
Receptor 1 de Folato/sangre , Ácido Fólico/metabolismo , Defectos del Tubo Neural/etiología , Adulto , Autoanticuerpos/análisis , Estudios de Casos y Controles , Labio Leporino/etiología , Fisura del Paladar/etiología , Femenino , Receptor 1 de Folato/inmunología , Deficiencia de Ácido Fólico/complicaciones , Humanos , Noruega , Embarazo
6.
Clin Genet ; 75(5): 409-23, 2009 May.
Artículo en Inglés | MEDLINE | ID: mdl-19459879

RESUMEN

It is generally understood that both genetic and environmental factors contribute to the highly complex etiology of structural birth defects, including neural tube defects, oral clefts and congenital heart defects, by disrupting highly regulated embryonic developmental processes. The intrauterine environment of the developing embryo/fetus is determined by maternal factors such as health/disease status, lifestyle, medication, exposure to environmental teratogens, as well as the maternal genotype. Certain genetic characteristics of the embryo/fetus also predispose it to developmental abnormalities. Epidemiologic and animal studies conducted over the last few decades have suggested that the interplay between genes and environmental factors underlies the etiological heterogeneity of these defects. It is now widely believed that the study of gene-environment interactions will lead to better understanding of the biological mechanisms and pathological processes that contribute to the development of complex birth defects. It is only through such an understanding that more efficient measures will be developed to prevent these severe, costly and often deadly defects. In this review, we attempt to summarize the complex clinical and experimental literature on current hypotheses of interactions between several select environmental factors and those genetic pathways in which they are most likely to have significant modifying effects. These include maternal folate nutritional status, maternal diabetes/obesity-related conditions, and maternal exposure to selected medications and environmental contaminants. Our goal is to highlight the potential gene-environment interactions affecting early embryogenesis that deserve comprehensive study.


Asunto(s)
Anomalías Congénitas/etiología , Exposición a Riesgos Ambientales , Animales , Fisura del Paladar/etiología , Anomalías Congénitas/genética , Susceptibilidad a Enfermedades , Exposición a Riesgos Ambientales/efectos adversos , Femenino , Ácido Fólico/metabolismo , Predisposición Genética a la Enfermedad/etiología , Humanos , Exposición Materna , Defectos del Tubo Neural/etiología
7.
Dev Dyn ; 236(4): 1044-54, 2007 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-17326132

RESUMEN

The amino acid homocysteine increases in the serum when there is insufficient folic acid or vitamin B(12), or with certain mutations in enzymes important in methionine metabolism. Elevated homocysteine is related to increased risk for cardiovascular and other diseases in adults and elevated maternal homocysteine increases the risk for certain congenital defects, especially those that result from abnormal development of the neural crest and neural tube. Experiments with the avian embryo model have shown that elevated homocysteine perturbs neural crest/neural tube migration in vitro and in vivo. Whereas there have been numerous studies of homocysteine-induced changes in gene expression in adult cells, there is no previous report of a homocysteine-responsive transcriptome in the embryonic neural crest. We treated neural crest cells in vitro with exogenous homocysteine in a protocol that induces significant changes in neural crest cell migration. We used microarray analysis and expression profiling to identify 65 transcripts of genes of known function that were altered by homocysteine. The largest set of effected genes (19) included those with a role in cell migration and adhesion. Other major groups were genes involved in metabolism (13); DNA/RNA interaction (11); cell proliferation/apoptosis (10); and transporter/receptor (6). Although the genes identified in this experiment were consistent with prior observations of the effect of homocysteine upon neural crest cell function, none had been identified previously as response to homocysteine in adult cells.


Asunto(s)
Regulación del Desarrollo de la Expresión Génica/efectos de los fármacos , Homocisteína/farmacología , Mioblastos Cardíacos/metabolismo , Cresta Neural/embriología , Análisis de Secuencia por Matrices de Oligonucleótidos , Animales , Adhesión Celular/efectos de los fármacos , Adhesión Celular/genética , Movimiento Celular/efectos de los fármacos , Movimiento Celular/genética , Células Cultivadas , Embrión de Pollo , Perfilación de la Expresión Génica , Cresta Neural/metabolismo
8.
Neurology ; 67(3): 407-12, 2006 Aug 08.
Artículo en Inglés | MEDLINE | ID: mdl-16894099

RESUMEN

BACKGROUND: Pregnancy outcomes following in utero exposure to antiepileptic drugs (AEDs) are uncertain, limiting an evidenced-based approach. OBJECTIVE: To determine if fetal outcomes vary as a function of different in utero AED exposures. METHODS: This ongoing prospective observational study across 25 epilepsy centers in the USA and UK enrolled pregnant women with epilepsy from October 1999 to February 2004 to determine if differential long-term cognitive and behavioral neurodevelopmental effects exist across the four most commonly used AEDs. This initial report focuses on the incidence of serious adverse outcomes including major congenital malformations (which could be attributable to AEDs) or fetal death. A total of 333 mother/child pairs were analyzed for monotherapy exposures: carbamazepine (n = 110), lamotrigine (n = 98), phenytoin (n = 56), and valproate (n = 69). RESULTS: Response frequencies of pregnancies resulting in serious adverse outcomes for each AED were as follows: carbamazepine 8.2%, lamotrigine 1.0%, phenytoin 10.7%, and valproate 20.3%. Distribution of serious adverse outcomes differed significantly across AEDs and was not explained by factors other than in utero AED exposure. Valproate exhibited a dose-dependent effect. CONCLUSIONS: More adverse outcomes were observed in pregnancies with in utero valproate exposure vs the other antiepileptic drugs (AEDs). These results combined with several recent studies provide strong evidence that valproate poses the highest risk to the fetus. For women who fail other AEDs and require valproate, the dose should be limited if possible.


Asunto(s)
Anomalías Inducidas por Medicamentos/etiología , Anticonvulsivantes/efectos adversos , Muerte Fetal/inducido químicamente , Complicaciones del Embarazo/inducido químicamente , Adulto , Anticonvulsivantes/administración & dosificación , Carbamazepina/efectos adversos , Cognición/efectos de los fármacos , Femenino , Humanos , Lamotrigina , Fenitoína/efectos adversos , Embarazo , Resultado del Embarazo , Estudios Prospectivos , Triazinas/efectos adversos , Útero/efectos de los fármacos , Ácido Valproico/efectos adversos
9.
Am J Med Genet A ; 135(3): 268-73, 2005 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-15887293

RESUMEN

Repairing DNA damage is critical during embryogenesis because development involves sensitive periods of cell proliferation, and abnormal cell growth or death can result in malformations. Knockout mouse experiments have demonstrated that disruption of DNA repair genes results in embryolethality and structural defects. Studies using mid-organogenesis rat embryos showed that DNA repair genes were variably expressed. It is hypothesized that polymorphisms that alter the functionality of DNA repair enzymes may modify the risk of malformations. We conducted a case-control analysis to investigate the relationship between DNA repair gene polymorphisms and the risk of spina bifida and oral clefts. Newborn screening blood spot DNA was obtained for 250 cases (125 spina bifida, 125 oral clefts) identified by the California Birth Defects Monitoring Program, and 350 non-malformation controls identified from birth records. Six single nucleotide polymorphisms of five DNA repair genes representing three distinct repair pathways were interrogated including: XRCC1 (Arg399Gln), APE1 (Asp148Glu), XRCC3 (Thr241Met), hOGG1(Ser326Cys), XPD (Asp312Asn, Lys751Gln). Elevated or decreased odds ratios (OR, adjusted for race/ethnicity) for spina bifida were found for genotypes containing at least one copy of the variant allele for XPD [751Gln, OR = 1.62; 95% confidence interval (CI) = 1.05-2.50] and APE 148 (OR = 0.58; CI = 0.37-0.90). A decreased risk of oral clefts was found for XRCC3 (OR = 0.62; CI = 0.39-0.99) and hOGG1 (326 Cys/Cys, OR = 0.22; CI = 0.06-0.78). This study suggested that polymorphisms of DNA repair genes, representing different major repair pathways, may affect risk of two major birth defects. Future, larger studies, examining additional repair genes, birth defects, and interaction with exposures are recommended.


Asunto(s)
Reparación del ADN/genética , Anomalías de la Boca/genética , Polimorfismo Genético , Disrafia Espinal/genética , Alelos , Estudios de Casos y Controles , Labio Leporino/diagnóstico , Labio Leporino/genética , Fisura del Paladar/diagnóstico , Fisura del Paladar/genética , ADN Glicosilasas/genética , ADN Helicasas/genética , ADN-(Sitio Apurínico o Apirimidínico) Liasa/genética , Proteínas de Unión al ADN/genética , Frecuencia de los Genes , Pruebas Genéticas , Genotipo , Humanos , Recién Nacido , Desequilibrio de Ligamiento , Anomalías de la Boca/diagnóstico , Mutación Missense , Tamizaje Neonatal , Oportunidad Relativa , Factores de Riesgo , Disrafia Espinal/diagnóstico , Factores de Transcripción/genética , Proteína 1 de Reparación por Escisión del Grupo de Complementación Cruzada de las Lesiones por Rayos X , Proteína de la Xerodermia Pigmentosa del Grupo D
10.
Neurology ; 64(6): 1088-90, 2005 Mar 22.
Artículo en Inglés | MEDLINE | ID: mdl-15781839

RESUMEN

The authors describe a 6-year-old girl with developmental delay, psychomotor regression, seizures, mental retardation, and autistic features associated with low CSF levels of 5-methyltetrahydrofolate, the biologically active form of folates in CSF and blood. Folate and B12 levels were normal in peripheral tissues, suggesting cerebral folate deficiency. Treatment with folinic acid corrected CSF abnormalities and improved motor skills.


Asunto(s)
Trastorno Autístico/tratamiento farmacológico , Corteza Cerebral/efectos de los fármacos , Corteza Cerebral/crecimiento & desarrollo , Discapacidades del Desarrollo/tratamiento farmacológico , Deficiencia de Ácido Fólico/tratamiento farmacológico , Leucovorina/administración & dosificación , Convulsiones/tratamiento farmacológico , Adaptación Fisiológica/efectos de los fármacos , Adaptación Fisiológica/fisiología , Trastorno Autístico/líquido cefalorraquídeo , Trastorno Autístico/etiología , Corteza Cerebral/metabolismo , Niño , Discapacidades del Desarrollo/líquido cefalorraquídeo , Discapacidades del Desarrollo/etiología , Progresión de la Enfermedad , Femenino , Ácido Fólico/metabolismo , Deficiencia de Ácido Fólico/líquido cefalorraquídeo , Deficiencia de Ácido Fólico/fisiopatología , Predisposición Genética a la Enfermedad , Humanos , Discapacidad Intelectual/tratamiento farmacológico , Discapacidad Intelectual/etiología , Discapacidad Intelectual/metabolismo , Mutación/genética , Recuperación de la Función/efectos de los fármacos , Recuperación de la Función/fisiología , Proteína Portadora de Folato Reducido/genética , Convulsiones/líquido cefalorraquídeo , Convulsiones/etiología , Tetrahidrofolatos/líquido cefalorraquídeo , Factores de Transcripción/genética , Resultado del Tratamiento
11.
Toxicol Appl Pharmacol ; 177(3): 238-46, 2001 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-11749123

RESUMEN

Arsenic is a well-known carcinogen, which has been suspected of being a human teratogen, although there is currently insufficient and inadequate supportive data to make any definitive judgments. In addition, the significance of individual genetic differences on pregnancy outcomes following in utero exposure to arsenic is currently unknown. In order to better understand the role of folate transport mechanisms in arsenic-induced neural tube defects, we examined the effect of in utero exposure to sodium arsenate in a genetically altered murine model in which the folate binding protein 2 (Folbp2) gene has been inactivated by homologous recombination. In utero sodium arsenate exposure induced exencephaly in 40.6% of Folbp2(-/-) embryos compared with 24.0% in control Folbp2(+/+) embryos. The differences in response frequencies were further exacerbated when the dams were fed a folate-deficient diet. Under these conditions, exencephaly was observed in 64.0% of Folbp2(-/-) embryos compared with 25.7% in control Folbp2(+/+) embryos. Analysis of arsenic metabolites excreted in the urine following sodium arsenate injection to Folbp2(-/-) and Folbp2(+/+) mice indicated that there were no significant differences in arsenic metabolism between the two groups. Thus, the increased susceptibility of Folbp2(-/-) mice to arsenate-induced teratogenicity may not be due to differences in biomethylation and exposure. In conclusion, the data suggest that impaired folate transport in the developing mouse embryo increases the risk for developmental defects following in utero exposure to sodium arsenate and that these differences are not due to differences in metabolism of arsenic.


Asunto(s)
Arseniatos/toxicidad , Proteínas Portadoras/metabolismo , Predisposición Genética a la Enfermedad , Defectos del Tubo Neural/inducido químicamente , Efectos Tardíos de la Exposición Prenatal , Receptores de Superficie Celular , Teratógenos/toxicidad , Animales , Arseniatos/metabolismo , Arsenicales/orina , Arsenitos/orina , Ácido Cacodílico/orina , Proteínas Portadoras/genética , Relación Dosis-Respuesta a Droga , Femenino , Reabsorción del Feto/inducido químicamente , Receptores de Folato Anclados a GPI , Deficiencia de Ácido Fólico , Genotipo , Homocisteína/sangre , Ratones , Ratones Noqueados , Defectos del Tubo Neural/patología , Embarazo , Teratógenos/metabolismo
13.
Semin Pediatr Neurol ; 8(3): 160-4, 2001 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-11575845

RESUMEN

Neural tube defects (NTDs) are common congenital malformations that occur when the embryonic neural tube fails to close properly during early development. Although multifactorial in origin, NTDs appear to have a strong genetic component. Mouse NTD mutants provide useful models for the study of candidate genes involved in neural tube development and closure. Because maternal nutrition, specifically folate supplementation, is a significant modulator of NTD risk, genes involved in folate transport and metabolism are a focus of investigation. In addition, transcription factors, as well as genes involved in mitosis, actin regulation, and methylation appear to be implicated in the causes of NTDs. The heterogeneity of function of candidate genes suggests that alterations in multiple developmental pathways may lead to the same clinical malformation.


Asunto(s)
Defectos del Tubo Neural/genética , 5-Metiltetrahidrofolato-Homocisteína S-Metiltransferasa/genética , 5-Metiltetrahidrofolato-Homocisteína S-Metiltransferasa/metabolismo , Animales , Modelos Animales de Enfermedad , Método Doble Ciego , Femenino , Ácido Fólico/administración & dosificación , Ácido Fólico/genética , Ácido Fólico/metabolismo , Ácido Fólico/uso terapéutico , Humanos , Recién Nacido , Masculino , Metilación , Metilenotetrahidrofolato Reductasa (NADPH2) , Ratones , Defectos del Tubo Neural/etiología , Defectos del Tubo Neural/prevención & control , Oxidorreductasas actuantes sobre Donantes de Grupo CH-NH/genética , Oxidorreductasas actuantes sobre Donantes de Grupo CH-NH/metabolismo , Placebos , Embarazo , Ensayos Clínicos Controlados Aleatorios como Asunto , Factores de Riesgo , Factores de Transcripción/genética , Vitaminas/administración & dosificación , Vitaminas/uso terapéutico
14.
Proc Nutr Soc ; 60(1): 53-61, 2001 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-11310424

RESUMEN

Population-based studies of human pregnancies show that periconceptional folate supplementation has a significant protective effect for embryos during early development, resulting in a significant reduction in developmental defects of the face, the neural tube, and the cono-truncal region of the heart. These results have been supported by experiments with animal models. An obvious quality held in common by these three anatomical regions is that the normal development of each region depends on a set of multi-potent cells that originate in the mid-dorsal region of the neural epithelium. However, the reason for the sensitive dependence of these particular cells on folic acid for normal development has not been obvious, and there is no consensus about the biological basis of the dramatic rescue with periconceptional folate supplementation. There are two principal hypotheses for the impact of folate insufficiency on development; each of these hypotheses has a micronutrient component and a genetic component. In the first hypothesis the effect of low folate is direct, limiting the availability of folic acid to cells within the embryo itself; thus compromising normal function and limiting proliferation. The second hypothetical effect is indirect; low folate disrupts methionine metabolism; homocysteine increases in the maternal serum; homocysteine induces abnormal development by inhibiting the function of N-methyl-D-aspartate (NMDA) receptors in the neural epithelium. There are three general families of genes whose level of expression may need to be considered in the context of these two related hypotheses: folate-receptor genes; genes that regulate methionine-homocysteine metabolism; NMDA-receptor genes.


Asunto(s)
Desarrollo Embrionario y Fetal/fisiología , Ácido Fólico/administración & dosificación , Homocisteína/metabolismo , Defectos del Tubo Neural/prevención & control , Animales , Suplementos Dietéticos , Desarrollo Embrionario y Fetal/genética , Femenino , Ácido Fólico/metabolismo , Homocisteína/genética , Humanos , Embarazo
15.
Teratology ; 63(5): 167-75, 2001 May.
Artículo en Inglés | MEDLINE | ID: mdl-11320527

RESUMEN

BACKGROUND: Very little is known about the identity of genetic factors involved in the complex etiology of nonsyndromic neural tube defects (NTD). Potential susceptibility genes have emerged from the vast number of mutant mouse strains displaying NTD. Reasonable candidates are the human homologues of mice exencephaly genes Tfap2alpha and Msx2, which are expressed in the developing neural tube. METHODS: A single-strand conformation analysis (SSCA) mutation screen of the coding sequences of TFAP2alpha and MSX2 was performed for 204 nonsyndromic NTD patients including cases of anencephaly (n = 10), encephalocele (n = 8), and spina bifida aperta, SBA (n = 183). A selected number of SBA patients was additionally tested for specific mutations in MTHFD, FRalpha, and PAX1 already shown to be related to NTD. RESULTS: Two TFAP2alpha point mutations in individual SBA patients were silent on the amino acid level (C308C, T396T). On nucleic acid level, these mutations change evolutionary conserved codons and thus may influence mRNA processing and translation efficiency. One SBA patient displayed an exonic 9-bp deletion in MSX2 leading to a shortened and possibly less functional protein. None of these mutations was found in 222 controls. Seven polymorphisms detected in TFAP2alpha and MSX2 were equally distributed in patients and controls. Patients with combined heterozygosity of an exonic MSX2 and an intronic TFAP2alpha polymorphism were at a slightly increased risk of NTD (OR 1.71; 95% CI 0.57-5.39). CONCLUSIONS: Although several new genetic variants were found in TFAP2 and MSX2, no statistically significant association was found between NTD cases and the new alleles or their combinations. Further studies are necessary to finally decide if these gene variants may have acted as susceptibility factors in our individual cases.


Asunto(s)
Proteínas de Unión al ADN/genética , Mutación , Defectos del Tubo Neural/genética , Receptores de Superficie Celular , Factores de Transcripción/genética , Alelos , Anencefalia/genética , Animales , Secuencia de Bases , Proteínas Portadoras/genética , Codón , ADN Complementario/metabolismo , Encefalocele/genética , Exones , Receptores de Folato Anclados a GPI , Ácido Fólico/metabolismo , Eliminación de Gen , Genotipo , Proteínas de Homeodominio , Humanos , Ratones , Datos de Secuencia Molecular , Linaje , Mutación Puntual , Polimorfismo Genético , Polimorfismo Conformacional Retorcido-Simple , Disrafia Espinal/genética , Factor de Transcripción AP-2
16.
Eur J Pediatr Surg ; 11 Suppl 1: S14-7, 2001 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-11813127

RESUMEN

Moderate hyperhomocysteinemia in pregnant women has been associated with an increased risk of neural tube defects (NTDs). Periconceptional supplementation with multi-vitamins containing folic acid may normalize homocysteine metabolism and decrease the NTD risk. The C677 T variant of the MTHFR gene coding for a thermolabile enzyme has been described as the first genetic risk factor that accounts for a group of NTDs characterized by low maternal folate status and high homocysteine concentrations. Another common mutation of the same MTHFR gene, A1298 C, has also been described as an NTD risk factor. In addition to abnormal folate metabolism, anything that compromises the internalization of folate into the cell may be involved in the pathogenesis of NTDs. For this reason, a common polymorphism in the RFC-1 gene encoding the reduced folate carrier protein (A80 G) could also be an additional NTD risk factor. In the present study we examined the genotypic distributions and the allele frequencies of MTHFR A1298 C and RFC-1 A80 G polymorphisms in DNA samples from healthy Italian individuals and compared them to the frequencies observed in NTD cases and their parents. By means of restriction enzymatic analysis, we determined that the frequency of the mutated C allele of the A1298 C mutation was 0.25 among control individuals, which is in the range of that recently reported in other ethnic groups. However, we report that the mutant C allele frequencies are significantly higher among NTD cases and case mothers than among controls (0.39, 0.44, 0.25). Furthermore, for the RFC-1 A80 G mutation, we found that the frequency of the G allele of the RFC-1 mutation was 0.46 in the control population, suggesting that this is a common polymorphism in the Italian population. In spite of the high prevalence of the 80 G/G genotype among healthy subjects, we observed an increased frequency of the G allele in NTD-affected children, and their mothers and fathers. These preliminary results indicate that both the MTHFR and RFC-1 polymorphisms may play a role in NTD risk, at least in the Italian population. Further studies should be directed toward the evaluation of the level of risk conferred by the mutant MTHFR and RFC-1 genotypes, as well as the interaction between these genetic determinants and other nutritional and environmental factors.


Asunto(s)
Homocisteína/genética , Proteínas de Transporte de Membrana , Defectos del Tubo Neural/genética , Oxidorreductasas actuantes sobre Donantes de Grupo CH-NH/genética , Polimorfismo Genético/genética , Adulto , Proteínas Portadoras/genética , Estudios de Casos y Controles , Femenino , Ácido Fólico/metabolismo , Genotipo , Homocisteína/metabolismo , Humanos , Italia/etnología , Masculino , Proteínas de la Membrana/genética , Metilenotetrahidrofolato Reductasa (NADPH2) , Mutación/genética , Oxidorreductasas actuantes sobre Donantes de Grupo CH-NH/deficiencia , Embarazo
17.
Am J Med Genet ; 95(3): 216-23, 2000 Nov 27.
Artículo en Inglés | MEDLINE | ID: mdl-11102926

RESUMEN

Periconceptional folate supplementation reduces the recurrence and occurrence risk of neural tube defects (NTD) by as much as 70%, yet the protective mechanism remains unknown. Inborn errors of folate and homocysteine metabolism may be involved in the aetiology of NTDs. Previous studies have demonstrated that both homozygosity for the C677T mutation in the methylenetetrahydrofolate reductase (MTHFR) gene, and combined heterozygosity for the C677T and for another mutation in the same gene, the A1298C polymorphism, represent genetic risk factors for NTDs. In an attempt to identify additional folate related genes that contribute to NTD pathogenesis, we performed molecular genetic analysis of folate receptors (FRs). We identified 4 unrelated patients out of 50 with de novo insertions of pseudogene (PS)-specific mutations in exon 7 and 3'UTR of the FRalpha gene, arising by microconversion events. All of the substitutions affect the carboxy-terminal amino acid membrane tail, or the GPI anchor region of the nascent protein. Furthermore, among 150 control individuals, we also identified one infant with a gene conversion event within the FRalpha coding region. This study, though preliminary, provides the first genetic association between molecular variations of the FRalpha gene and NTDs and suggests that this gene can act as a risk factor for human NTD.


Asunto(s)
Ácido Fólico/genética , Defectos del Tubo Neural/etiología , Receptores de Superficie Celular , Regiones no Traducidas 3' , Secuencia de Bases , Southern Blotting , Proteínas Portadoras/genética , Niño , Preescolar , Análisis Mutacional de ADN , Exones , Femenino , Receptores de Folato Anclados a GPI , Ácido Fólico/fisiología , Glicosilfosfatidilinositoles/genética , Humanos , Lactante , Recién Nacido , Masculino , Datos de Secuencia Molecular , Mutación , Defectos del Tubo Neural/genética , Sistemas de Lectura Abierta , Linaje , Polimorfismo Conformacional Retorcido-Simple , Factores de Riesgo , Alineación de Secuencia
18.
Gene ; 258(1-2): 117-25, 2000 Nov 27.
Artículo en Inglés | MEDLINE | ID: mdl-11111049

RESUMEN

Utilizing a 'database mining' strategy to detect novel folate receptors (FR), we identified two potential novel members in the mouse and human. The mouse gene (Folbp3) was sequenced and found to predict a 28.2 kDa protein that consists of 244 amino acids that is highly expressed in both the thymus and spleen, suggesting a potential role in the immune system. The human gene (FR-delta) is mapped to chromosome 11q14, and predicts a 27.7 kDa protein that is comprised of 241 amino acids. However, expression of the human gene was not detected in 59 samples from both adult and embryonic tissue sources, suggesting a highly restricted spatial/temporal expression pattern, an alternatively spliced variant or an additional FR pseudogene. Using T31 mouse radiation hybrid mapping, Folbp3 was mapped to a region on mouse chromosome 9 that is syntenic to human chromosome 19p13. As the chromosomal locations of Folbp1 murine and Folbp2 genes were previously unknown, we utilized the same approach and mapped both genes to a region of mouse chromosome 7 that is syntenic to the human FR loci on chromosome 11q13.


Asunto(s)
Proteínas Portadoras/genética , Receptores de Superficie Celular , Adulto , Secuencia de Aminoácidos , Animales , Northern Blotting , Mapeo Cromosómico , Cromosomas Humanos Par 11/genética , ADN Complementario/química , ADN Complementario/genética , Embrión de Mamíferos/metabolismo , Femenino , Receptores de Folato Anclados a GPI , Expresión Génica , Regulación del Desarrollo de la Expresión Génica , Humanos , Masculino , Ratones , Datos de Secuencia Molecular , ARN Mensajero/genética , ARN Mensajero/metabolismo , Mapeo de Híbrido por Radiación , Alineación de Secuencia , Análisis de Secuencia de ADN , Homología de Secuencia de Aminoácido , Distribución Tisular
20.
Toxicol Lett ; 117(3): 129-37, 2000 Nov 20.
Artículo en Inglés | MEDLINE | ID: mdl-11087978

RESUMEN

It has been proposed that arsenic exerts its toxic effects, in part, by perturbing cellular methyl metabolism. Based on the hypothesis that folic acid treatment will attenuate the cytotoxic and growth inhibitory effects of arsenic, SWV/Fnn embryo fibroblasts were cultured in media supplemented with various concentrations of folic acid during treatment with sodium arsenite or dimethylarsinic acid (DMA). It was found that folic acid protects SWV/Fnn embryo fibroblasts from sodium arsenite and DMA cytotoxicity in a dose-dependent manner. In contrast, folic acid supplementation has no effect on toxicity resulting from treatment with ethanol or staurosporine, suggesting that folic acid is not generally protective against necrosis and apoptosis. Although folic acid protects against acute arsenic toxicity, this agent shows a modest and delayed ability to attenuate the growth inhibitory effect of arsenic on these cells. These results support a model in which perturbations of methyl metabolism contribute to the acute cytotoxicity of arsenic.


Asunto(s)
Arsénico/antagonistas & inhibidores , Arsénico/toxicidad , Ácido Fólico/uso terapéutico , Animales , Arsenitos/antagonistas & inhibidores , Arsenitos/toxicidad , Ácido Cacodílico/antagonistas & inhibidores , Ácido Cacodílico/toxicidad , Carcinógenos/toxicidad , División Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Depresores del Sistema Nervioso Central/toxicidad , Etanol/toxicidad , Femenino , Fibroblastos/efectos de los fármacos , Ratones , Ratones Endogámicos , Compuestos de Sodio/antagonistas & inhibidores , Compuestos de Sodio/toxicidad , Estaurosporina/toxicidad
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...