Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
1.
AIDS ; 38(4): 439-446, 2024 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-37382903

RESUMEN

OBJECTIVES: In 2018, the Botswana Tsepamo Study reported a nine-fold increased risk of neural tube defects in infants whose mothers were treated with dolutegravir (DTG) from the time of conception. As maternal folate supplementation and status is a well known modifier of neural tube defect (NTD) risk, we sought to evaluate birth outcomes in mice fed normal and low folic acid diets treated with DTG during pregnancy. DESIGN: DTG was evaluated for developmental toxicity using pregnant mice fed normal or low folic acid diet. METHODS: CD-1 mice were provided diet with normal (3 mg/kg) or low (0.3 mg/kg) folic acid. They were treated with water, a human therapeutic-equivalent dose, or supratherapeutic dose of DTG from mouse embryonic day E6.5 to E12.5. Pregnant dams were sacrificed at term (E18.5) and fetuses were inspected for gross, internal, and skeletal defects. RESULTS: Fetuses with exencephaly, an NTD, were present in both therapeutic human equivalent and supratherapeutic exposures in dams fed low folic acid diet. Cleft palates were also found under both folate conditions. CONCLUSIONS: Recommended dietary folic acid levels during mouse pregnancy ameliorate developmental defects that arise from DTG exposure. Since low folate status in mice exposed to DTG increases the risk for NTDs, it is possible that DTG exposures in people living with HIV with low folate status during pregnancy may explain, at least in part, the elevated NTD risk signal observed in Botswana. Based on these results, future studies should consider folate status as a modifier for DTG-associated NTD risk.


Asunto(s)
Infecciones por VIH , Defectos del Tubo Neural , Oxazinas , Piperazinas , Piridonas , Humanos , Embarazo , Femenino , Animales , Ratones , Ácido Fólico/uso terapéutico , Infecciones por VIH/tratamiento farmacológico , Infecciones por VIH/complicaciones , Defectos del Tubo Neural/inducido químicamente , Defectos del Tubo Neural/tratamiento farmacológico , Compuestos Heterocíclicos con 3 Anillos/efectos adversos
2.
FASEB J ; 38(1): e23346, 2024 01.
Artículo en Inglés | MEDLINE | ID: mdl-38095297

RESUMEN

Folate deficiency contribute to neural tube defects (NTDs) which could be rescued by folate supplementation. However, the underlying mechanisms are still not fully understood. Besides, there is considerable controversy concerning the forms of folate used for supplementation. To address this controversy, we prepared culture medium with different forms of folate, folic acid (FA), and 5-methyltetrahydrofolate (5mTHF), at concentrations of 5 µM, 500 nM, 50 nM, and folate free, respectively. Mouse embryonic fibroblasts (MEFs) were treated with different folates continuously for three passages, and cell proliferation and F-actin were monitored. We determined that compared to 5mTHF, FA showed stronger effects on promoting cell proliferation and F-actin formation. We also found that FOLR1 protein level was positively regulated by folate concentration and the non-canonical Wnt/planar cell polarity (PCP) pathway signaling was significantly enriched among different folate conditions in RNA-sequencing analyses. We demonstrated for the first time that FOLR1 could promote the transcription of Vangl2, one of PCP core genes. The transcription of Vangl2 was down-regulated under folate-deficient condition, which resulted in a decrease in PCP activity and F-actin formation. In summary, we identified a distinct advantage of FA in cell proliferation and F-actin formation over 5mTHF, as well as demonstrating that FOLR1 could promote transcription of Vangl2 and provide a new mechanism by which folate deficiency can contribute to the etiology of NTDs.


Asunto(s)
Deficiencia de Ácido Fólico , Defectos del Tubo Neural , Animales , Ratones , Ácido Fólico/metabolismo , Actinas/metabolismo , Receptor 1 de Folato/genética , Receptor 1 de Folato/metabolismo , Polaridad Celular/genética , Fibroblastos/metabolismo , Vía de Señalización Wnt , Defectos del Tubo Neural/genética , Defectos del Tubo Neural/metabolismo , Deficiencia de Ácido Fólico/metabolismo
3.
Front Genet ; 12: 659612, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34040637

RESUMEN

Human structural congenital malformations are the leading cause of infant mortality in the United States. Estimates from the United States Center for Disease Control and Prevention (CDC) determine that close to 3% of all United States newborns present with birth defects; the worldwide estimate approaches 6% of infants presenting with congenital anomalies. The scientific community has recognized for decades that the majority of birth defects have undetermined etiologies, although we propose that environmental agents interacting with inherited susceptibility genes are the major contributing factors. Neural tube defects (NTDs) are among the most prevalent human birth defects and as such, these malformations will be the primary focus of this review. NTDs result from failures in embryonic central nervous system development and are classified by their anatomical locations. Defects in the posterior portion of the neural tube are referred to as meningomyeloceles (spina bifida), while the more anterior defects are differentiated as anencephaly, encephalocele, or iniencephaly. Craniorachischisis involves a failure of the neural folds to elevate and thus disrupt the entire length of the neural tube. Worldwide NTDs have a prevalence of approximately 18.6 per 10,000 live births. It is widely believed that genetic factors are responsible for some 70% of NTDs, while the intrauterine environment tips the balance toward neurulation failure in at risk individuals. Despite aggressive educational campaigns to inform the public about folic acid supplementation and the benefits of providing mandatory folic acid food fortification in the United States, NTDs still affect up to 2,300 United States births annually and some 166,000 spina bifida patients currently live in the United States, more than half of whom are now adults. Within the context of this review, we will consider the role of maternal nutritional status (deficiency states involving B vitamins and one carbon analytes) and the potential modifiers of NTD risk beyond folic acid. There are several well-established human teratogens that contribute to the population burden of NTDs, including: industrial waste and pollutants [e.g., arsenic, pesticides, and polycyclic aromatic hydrocarbons (PAHs)], pharmaceuticals (e.g., anti-epileptic medications), and maternal hyperthermia during the first trimester. Animal models for these teratogens are described with attention focused on valproic acid (VPA; Depakote). Genetic interrogation of model systems involving VPA will be used as a model approach to discerning susceptibility factors that define the gene-environment interactions contributing to the etiology of NTDs.

4.
AIDS ; 33(13): 1967-1976, 2019 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-31259764

RESUMEN

OBJECTIVE: Maternal folate (vitamin B9) status is the largest known modifier of neural tube defect risk, so we evaluated folate-related mechanisms of action for dolutegravir (DTG) developmental toxicity. DESIGN: Folate receptor 1 (FOLR1) was examined as a target for DTG developmental toxicity using protein and cellular interaction studies and an animal model. METHODS: FOLR1 competitive binding studies were used to test DTG for FOLR1 antagonism. Human placenta cell line studies were used to test interactions with DTG, folate, and cations. Zebrafish were selected as an animal model to examine DTG-induced developmental toxicity and rescue strategies. RESULTS: FOLR1 binding studies indicate DTG is a noncompetitive FOLR1 antagonist at therapeutic concentrations. In-vitro testing indicates calcium (2 mmol/l) increases FOLR1-folate interactions and alters DTG-FOLR1-folate interactions and cytotoxicity. DTG does not inhibit downstream folate metabolism by dihydrofolate reductase. Early embryonic exposure to DTG is developmentally toxic in zebrafish, and supplemental folic acid can mitigate DTG developmental toxicity. CONCLUSION: Folates and FOLR1 are established modifiers of risk for neural tube defects, and binding data indicates DTG is a partial antagonist of FOLR1. Supplemental folate can ameliorate increased developmental toxicity due to DTG in zebrafish. The results from these studies are expected to inform and guide future animal models and clinical studies of DTG-based antiretroviral therapy in women of childbearing age.


Asunto(s)
Receptor 1 de Folato/antagonistas & inhibidores , Ácido Fólico/administración & dosificación , Compuestos Heterocíclicos con 3 Anillos/toxicidad , Proteínas de Pez Cebra/antagonistas & inhibidores , Pez Cebra/embriología , Animales , Línea Celular , Suplementos Dietéticos , Desarrollo Embrionario/efectos de los fármacos , Femenino , Receptor 1 de Folato/genética , Infecciones por VIH/tratamiento farmacológico , Inhibidores de Integrasa VIH/toxicidad , Humanos , Modelos Animales , Oxazinas , Piperazinas , Embarazo , Piridonas , Pruebas de Toxicidad , Pez Cebra/genética , Proteínas de Pez Cebra/genética
5.
Birth Defects Res ; 111(14): 1013-1023, 2019 08 15.
Artículo en Inglés | MEDLINE | ID: mdl-30325584

RESUMEN

OBJECTIVES: The anticonvulsant valproic acid (VPA) has a known teratogenic effect capable of inducing major congenital malformations and developmental disorders. A comparative teratogenicity study of VPA and its analog valnoctamide (VCD), which is a new generation candidate antiepileptic drug, was carried out using Swiss Vancouver (SWV) mice. METHODS: Pregnant SWV dams were treated with either a single intraperitoneal injection of VPA (1.8 and 2.7 mmol/kg), VCD (1.8 and 2.7 mmol/kg), or vehicle on E8:12 (gestational day:hour). The numbers of implantation and resorption, viable and dead fetuses, and the presence of gross fetal visceral and skeletal abnormalities were determined (E18). Real-time Polymerase chain reaction (RT-PCR) arrays were used to analyze the expression of 84 genes related to the processes of neurogenesis and neural stem cell differentiation. RESULTS: Significant decreases in pregnancy weight gain and the number of live fetuses were observed when VPA was administered at the high dose, whereas the percentage of exencephalic fetuses was significantly increased in VPA treated compared with an equivalent VCD dosage group. There was a dose-related increase in visceral defects in the VPA-exposed fetuses. Missing skull bones and fused vertebrae in fetuses occurred at the high dose of VPA. Three genes (Mtap2, Bmp8b, and Stat3) were significantly upregulated and one (Heyl) was downregulated in samples from VPA-treated dams. CONCLUSIONS: The study demonstrates that the teratogenicity of VPA was significantly greater than that of an equimolar dose of VCD. Four genes (Mtap2, Bmp8b, Stat3, and Heyl) represent candidate target genes for the underlying teratogenic mechanism responsible for VPA-induced malformations.


Asunto(s)
Amidas/efectos adversos , Teratogénesis/efectos de los fármacos , Ácido Valproico/efectos adversos , Anomalías Inducidas por Medicamentos/etiología , Anomalías Inducidas por Medicamentos/fisiopatología , Amidas/farmacología , Animales , Anticonvulsivantes/efectos adversos , Femenino , Muerte Fetal , Feto/efectos de los fármacos , Ratones , Defectos del Tubo Neural/inducido químicamente , Embarazo , Teratógenos/metabolismo , Teratoma/etiología , Ácido Valproico/análogos & derivados , Ácido Valproico/farmacología
6.
Proc Natl Acad Sci U S A ; 115(18): 4690-4695, 2018 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-29666258

RESUMEN

Periconceptional folic acid (FA) supplementation significantly reduces the prevalence of neural tube defects (NTDs). Unfortunately, some NTDs are FA resistant, and as such, NTDs remain a global public health concern. Previous studies have identified SLC25A32 as a mitochondrial folate transporter (MFT), which is capable of transferring tetrahydrofolate (THF) from cellular cytoplasm to the mitochondria in vitro. Herein, we show that gene trap inactivation of Slc25a32 (Mft) in mice induces NTDs that are folate (5-methyltetrahydrofolate, 5-mTHF) resistant yet are preventable by formate supplementation. Slc25a32gt/gt embryos die in utero with 100% penetrant cranial NTDs. 5-mTHF supplementation failed to promote normal neural tube closure (NTC) in mutant embryos, while formate supplementation enabled the majority (78%) of knockout embryos to complete NTC. A parallel genetic study in human subjects with NTDs identified biallelic loss of function SLC25A32 variants in a cranial NTD case. These data demonstrate that the loss of functional Slc25a32 results in cranial NTDs in mice and has also been observed in a human NTD patient.


Asunto(s)
Formiatos/farmacología , Proteínas de Transporte de Membrana/genética , Proteínas de Transporte de Membrana/metabolismo , Mutación , Defectos del Tubo Neural , Tubo Neural , Animales , Transporte Biológico Activo/genética , Humanos , Ratones , Ratones Transgénicos , Tubo Neural/embriología , Tubo Neural/patología , Defectos del Tubo Neural/embriología , Defectos del Tubo Neural/genética , Defectos del Tubo Neural/patología , Defectos del Tubo Neural/prevención & control
7.
Birth Defects Res A Clin Mol Teratol ; 106(8): 685-95, 2016 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-27166990

RESUMEN

BACKGROUND: After years of periconceptional folic acid supplementation, the prevalence of neural tube defects (NTDs) remains stable following the remarkable reduction observed immediately after the fortification practice. There is accumulating evidence that folate receptor (FR) autoimmunity may play a role in the etiology of folate-sensitive NTDs. METHODS: From 2011 to 2013, 118 NTD cases and 242 healthy controls were recruited from a population-based birth defects surveillance system in Northern China. Enzyme-linked immunosorbent assay was used to measure FR autoantibodies in maternal and cord blood. Logistic regression models were used to estimate the odds ratios (OR) and 95% confidence intervals (95% CI). RESULTS: Plasma FR autoantibodies levels were significantly elevated in mothers of infants with NTDs compared with mothers of healthy controls. Using the lowest tertile as the referent group, 2.20-fold (95% CI, 0.71-6.80) and 5.53-fold increased odds (95% CI, 1.90-16.08) of NTDs were observed for the second and third tertile of immunoglobulin G (IgG), respectively, and the odds of NTDs for each successive tertile of IgM was 0.98 (95% CI, 0.35-2.75) and 3.49 (95% CI, 1.45-8.39), respectively. A dose-response relationship was found between FR autoantibodies levels and risk of NTDs (P < 0.001 for IgG, P = 0.002 for IgM). The same pattern was observed in both subtypes of spina bifida and anencephaly. No significant difference in levels of cord blood FR autoantibodies was observed. CONCLUSION: Higher levels of FR autoimmunity in maternal plasma are associated with elevated risk of NTDs in a dose-response manner. Birth Defects Research (Part A) 106:685-695, 2016. © 2016 Wiley Periodicals, Inc.


Asunto(s)
Anencefalia/inmunología , Autoanticuerpos/sangre , Autoinmunidad/efectos de los fármacos , Receptor 1 de Folato/antagonistas & inhibidores , Ácido Fólico/inmunología , Inmunoglobulina G/sangre , Adulto , Anencefalia/diagnóstico , Anencefalia/genética , Anencefalia/patología , Estudios de Casos y Controles , China , Relación Dosis-Respuesta Inmunológica , Tolerancia a Medicamentos/inmunología , Ensayo de Inmunoadsorción Enzimática , Femenino , Receptor 1 de Folato/genética , Receptor 1 de Folato/inmunología , Ácido Fólico/administración & dosificación , Expresión Génica , Humanos , Lactante , Modelos Logísticos , Masculino , Oportunidad Relativa , Riesgo
8.
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
9.
FASEB J ; 27(3): 1167-75, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-23212123

RESUMEN

Folate deficiency has been associated with many adverse clinical manifestations. The blood-brain barrier (BBB), formed by brain capillary endothelial cells, protects the brain from exposure to neurotoxicants. The function of BBB is modulated by multiple ABC transporters, particularly P-glycoprotein. A proton-coupled folate transporter (PCFT)-deficient mouse has been previously described as a model for systemic folate deficiency. Herein, we demonstrate that exposing mouse brain capillaries to the antiepileptic drug, valproic acid (VPA; 5 µM), significantly increased P-glycoprotein transport function in the wild-type animals. A ligand to the aryl hydrocarbon receptor, 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD), produced a similar induction of P-glycoprotein, which tightened the BBB, thereby increasing the neuroprotection. However, VPA- or TCDD-induced P-glycoprotein transport was blocked in the PCFT-nullizygous mice, indicating that multiple neuroprotective mechanisms are compromised under folate-deficient conditions. Brain capillaries from S-folinic acid (SFA; 40 mg/kg)-treated PCFT-nullizygous mice exhibited increased P-glycoprotein transport following VPA exposure. This suggests that SFA supplementation restored the normal BBB function. In addition, we show that tight-junction proteins are disintegrated in the PCFT mutant mice. Taken together, these findings strongly suggest that folate deficiency disrupts the BBB function by targeting the transporter and tight junctions, which may contribute to the development of neurological disorders.


Asunto(s)
Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/metabolismo , Barrera Hematoencefálica/metabolismo , Transportador de Folato Acoplado a Protón/metabolismo , Uniones Estrechas/metabolismo , Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/genética , Animales , Anticonvulsivantes/farmacología , Transporte Biológico Activo/efectos de los fármacos , Transporte Biológico Activo/genética , Barrera Hematoencefálica/patología , Leucovorina/farmacología , Ratones , Ratones Mutantes , Enfermedades del Sistema Nervioso/genética , Enfermedades del Sistema Nervioso/metabolismo , Enfermedades del Sistema Nervioso/patología , Dibenzodioxinas Policloradas/farmacología , Transportador de Folato Acoplado a Protón/genética , Teratógenos/farmacología , Uniones Estrechas/genética , Uniones Estrechas/patología , Ácido Valproico/farmacología , Complejo Vitamínico B/farmacología
10.
Neurochem Res ; 29(6): 1105-12, 2004 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-15176467

RESUMEN

Inactivation of the murine folate binding protein-1 (Folbp1) has been shown to play a vital role in embryonic development. Nullizygous embryos (Folbp1-/-) have significant malformations of the neural tube, craniofacies, and conotruncus, and invariably die in utero by gestational day (E) 10. Administration of 25 mg x kg(-1) x day(-1) folinic acid to dams prior to and throughout gestation rescues the majority of embryos from premature death; however, a portion of surviving embryos develops neural tube defects. Using antisense RNA amplification and cDNA microarrays, we examined the expression of approximately 5700 genes in the anterior neural tube of gestational day 9 Folbp1-/- embryos that were supplemented with folinic acid. Genes that appear to be folate regulated include transcription factors, G-proteins, growth factors, methyltransferases, and those that are related to cell proliferation. The potential impact of such changes during neural tube closure is considered in light of the phenotype of Folbp1-/- embryos.


Asunto(s)
Proteínas Portadoras/genética , Ácido Fólico/farmacología , Regulación del Desarrollo de la Expresión Génica/genética , Sistema Nervioso/embriología , Receptores de Superficie Celular/genética , Animales , Desarrollo Embrionario y Fetal/genética , Receptores de Folato Anclados a GPI , Regulación del Desarrollo de la Expresión Génica/efectos de los fármacos , Heterocigoto , Ratones , Ratones Noqueados , Familia de Multigenes , Hibridación de Ácido Nucleico
11.
Mamm Genome ; 15(5): 361-9, 2004 May.
Artículo en Inglés | MEDLINE | ID: mdl-15170225

RESUMEN

Human neural tube defects (NTDs) are among the most common congenital defects. They have a highly heterogeneous etiology, and, in addition to those seen in association with genetic syndromes, there are also NTDs induced by pharmaceutical compounds in utero, such as the widely used anti-epileptic drug valproic acid (VPA). Although familial studies have suggested a genetic contribution to VPA-induced NTDs, this trait has not been adequately studied, nor have the responsible genetic factors been identified. We generated a series of mouse crosses and backcrosses using the highly inbred SWV/Fnn and C57BL/6J strains, in order to identify possible chromosomal loci contributing to VPA sensitivity. When exposed to a high dose of sodium VPA (600 mg/kg) via maternal intraperitoneal injection on gestational day E8.5, the fetuses manifested exencephaly in a strain-dependent manner. Our data show an autosomal recessive trait, plus a gender-related effect or an overall X-Chromosome (Chr) effect, as being primarily responsible for determining sensitivity to VPA-induced exencephaly. Genome scanning and further linkage analysis of 131 exencephalic backcross fetuses identified a major locus linked to D7Mit285 (p < 2 x 10(-6)), exceeding the threshold for significant linkage. These results suggest a major chromosomal locus associated with the sensitivity to VPA-induced exencephaly in mice.


Asunto(s)
Anticonvulsivantes/toxicidad , Predisposición Genética a la Enfermedad , Genoma , Exposición Materna/efectos adversos , Defectos del Tubo Neural/inducido químicamente , Ácido Valproico/toxicidad , Animales , Mapeo Cromosómico , Cruzamientos Genéticos , Femenino , Inyecciones Intraperitoneales , Masculino , Ratones , Ratones Endogámicos C57BL , Embarazo
12.
Birth Defects Res C Embryo Today ; 72(4): 330-44, 2004 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-15662706

RESUMEN

Neural tube defects (NTDs) are serious malformations affecting approximately 1 per 1000 births, yet the mechanisms by which they arise are unknown. There have been consistent efforts in many fields of research to elucidate the etiology of this multifactorial condition. While no single gene has been identified as a major independent risk factor for NTDs, candidate genes have been proposed that may modify the effects of maternal and/or embryonic exposures. Folate supplementation effectively reduces the occurrence of NTDs and, consequently, has focused much research on metabolism of folate-related pathways during pregnancy and development. Further understanding of normal development and how teratogens can perturb these orchestrated processes also remains at the fore of modern scientific endeavors. The composite of these factors remains fragmented; the aim of this review is to provide the reader with a summary of sentinel and current works in the body of literature addressing NTD disease etiology.


Asunto(s)
Sistema Nervioso Central/embriología , Ácido Fólico/metabolismo , Fumonisinas/toxicidad , Defectos del Tubo Neural/etiología , Teratógenos/toxicidad , Vitamina B 12/efectos adversos , Complicaciones de la Diabetes , Efectos Colaterales y Reacciones Adversas Relacionados con Medicamentos , Femenino , Fiebre/complicaciones , Ácido Fólico/uso terapéutico , Fumonisinas/metabolismo , Humanos , Defectos del Tubo Neural/inducido químicamente , Defectos del Tubo Neural/prevención & control , Obesidad/complicaciones , Embarazo , Factores de Riesgo , Teratógenos/metabolismo , Vitamina B 12/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA