Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Front Cell Dev Biol ; 11: 1175917, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37377737

RESUMO

In 2018, data from a surveillance study in Botswana evaluating adverse birth outcomes raised concerns that women on antiretroviral therapy (ART) containing dolutegravir (DTG) may be at increased risk for neural tube defects (NTDs). The mechanism of action for DTG involves chelation of Mg2+ ions in the active site of the viral integrase. Plasma Mg2+ homeostasis is maintained primarily through dietary intake and reabsorption in the kidneys. Inadequate dietary Mg2+ intake over several months results in slow depletion of plasma Mg2+ and chronic latent hypomagnesemia, a condition prevalent in women of reproductive age worldwide. Mg2+ is critical for normal embryonic development and neural tube closure. We hypothesized that DTG therapy might slowly deplete plasma Mg2+ and reduce the amount available to the embryo, and that mice with pre-existing hypomagnesemia due to genetic variation and/or dietary Mg2+ insufficiency at the time of conception and initiation of DTG treatment would be at increased risk for NTDs. We used two different approaches to test our hypothesis: 1) we selected mouse strains that had inherently different basal plasma Mg2+ levels and 2) placed mice on diets with different concentrations of Mg2+. Plasma and urine Mg2+ were determined prior to timed mating. Pregnant mice were treated daily with vehicle or DTG beginning on the day of conception and embryos examined for NTDs on gestational day 9.5. Plasma DTG was measured for pharmacokinetic analysis. Our results demonstrate that hypomagnesemia prior to conception, due to genetic variation and/or insufficient dietary Mg2+ intake, increases the risk for NTDs in mice exposed to DTG. We also analyzed whole-exome sequencing data from inbred mouse strains and identified 9 predicted deleterious missense variants in Fam111a that were unique to the LM/Bc strain. Human FAM111A variants are associated with hypomagnesemia and renal Mg2+ wasting. The LM/Bc strain exhibits this same phenotype and was the strain most susceptible to DTG-NTDs. Our results suggest that monitoring plasma Mg2+ levels in patients on ART regimens that include DTG, identifying other risk factors that impact Mg2+ homeostasis, and correcting deficiencies in this micronutrient might provide an effective strategy for mitigating NTD risk.

2.
Adv Food Nutr Res ; 56: 145-81, 2009.
Artigo em Inglês | MEDLINE | ID: mdl-19389609

RESUMO

Fumonisins are mycotoxins produced by the fungus F. verticillioides, a common contaminant of maize (corn) worldwide. Maternal consumption of fumonisin B(1)-contaminated maize during early pregnancy has recently been associated with increased risk for neural tube defects (NTDs) in human populations that rely heavily on maize as a dietary staple. Experimental administration of purified fumonisin to mice early in gestation also results in an increased incidence of NTDs in exposed offspring. Fumonisin inhibits the enzyme ceramide synthase in de novo sphingolipid biosynthesis, resulting in an elevation of free sphingoid bases and depletion of downstream glycosphingolipids. Increased sphingoid base metabolites (i.e., sphinganine-1-phosphate) may perturb signaling cascades involved in embryonic morphogenesis by functioning as ligands for sphingosine-1-P (S1P) receptors, a family of G-protein-coupled receptors that regulate key biological processes such as cell survival/proliferation, differentiation and migration. Fumonisin-induced depletion of glycosphingolipids impairs expression and function of the GPI-anchored folate receptor (Folr1), which may also contribute to adverse pregnancy outcomes. NTDs appear to be multifactorial in origin, involving complex gene-nutrient-environment interactions. Vitamin supplements containing folic acid have been shown to reduce the occurrence of NTDs, and may help protect the developing fetus from environmental teratogens. Fumonisins appear to be an environmental risk factor for birth defects, although other aspects of maternal nutrition and genetics play interactive roles in determining pregnancy outcome. Minimizing exposures to mycotoxins through enhanced agricultural practices, identifying biomarkers of exposure, characterizing mechanisms of toxicity, and improving maternal nutrition are all important strategies for reducing the NTD burden in susceptible human populations.


Assuntos
Fumonisinas/toxicidade , Exposição Materna/efeitos adversos , Defeitos do Tubo Neural/etiologia , Teratogênicos/toxicidade , Animais , Feminino , Deficiência de Ácido Fólico , Contaminação de Alimentos/legislação & jurisprudência , Fumonisinas/farmacologia , Fumonisinas/normas , Predisposição Genética para Doença , Humanos , Recém-Nascido , Masculino , Fenômenos Fisiológicos da Nutrição Materna , Defeitos do Tubo Neural/induzido quimicamente , Defeitos do Tubo Neural/epidemiologia , Defeitos do Tubo Neural/prevenção & controle , Gravidez , Fatores de Risco , Esfingolipídeos/metabolismo , Teratogênicos/farmacologia , Teratogênicos/normas , Zea mays
3.
Exp Eye Res ; 86(2): 419-33, 2008 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-18191835

RESUMO

Microphthalmia-associated transcription factor (Mitf) is expressed in neural crest cell-derived melanocytes, and in the retinal pigment epithelium (RPE) during ocular development. Mutations in Mitf are associated with auditory/visual/pigmentary syndromes in humans. Mitf(mi/mi) mouse mutants lack pigmentation, and are microphthalmic, while Mitf(vit/vit) mouse mutants display abnormal RPE pigmentation, and progressive retinal degeneration. Microarray analysis was used to identify novel downstream gene targets/pathways in the RPE that are altered by mutations in the transcription factor Mitf. Using the Affymetrix platform, gene expression profiles were generated using the eyes of E13.5 mouse fetuses that were wildtype, heterozygous, or homozygous for the Mitf(mi) mutation. In a separate experiment, eyes from E13.5 mouse fetuses homozygous for the Mitf(vit) mutation were compared to eyes from the C57BL/6 control background strain. Statistical analyses were performed using robust multiarray average, mixed-effects ANOVA and random-variance t-tests. Altered expression of genes involved in pigment formation, melanosome biogenesis/transport, and redox homeostasis were observed. Twelve genes were commonly mis-regulated in the eyes of both Mitf mutants: 10 of these genes were downregulated in both mutants relative to controls, while 2 of the genes (Nramp1 (Slc11a1) and epoxide hydrolase) were downregulated in Mitf(mi/mi) mutants, and conversely, upregulated in Mitf(vit/vit) mutants. Quantitative RT-PCR and immunohistochemistry were used to confirm altered gene/protein expression. RPE expression of the Fe(+2) iron transporter Nramp1 (Slc11a1) has not previously been reported. Fe(+2) is an important co-factor utilized by the iron-dependent isomerohydrolase RPE65 in the retinoid visual cycle. However, excess accumulation of Fe(+2) in the RPE has recently been associated with oxidative damage and age-related macular degeneration. Abnormal pigmentation and increased activity of Slc11a1 in the RPE of Mitf(vit) mice may contribute to the pathology and progressive retinal degeneration observed in these mutants.


Assuntos
Proteínas de Transporte de Cátions/metabolismo , Desenvolvimento Fetal/genética , Fator de Transcrição Associado à Microftalmia/genética , Epitélio Pigmentado Ocular/embriologia , Degeneração Retiniana/genética , Animais , Antígenos de Neoplasias , Perfilação da Expressão Gênica/métodos , Regulação da Expressão Gênica no Desenvolvimento , Homeostase/genética , Ferro/metabolismo , Antígenos Específicos de Melanoma , Melanossomas/ultraestrutura , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Mutantes , Fator de Transcrição Associado à Microftalmia/metabolismo , Microftalmia/genética , Microftalmia/metabolismo , Microscopia Eletrônica , Proteínas de Neoplasias/metabolismo , Análise de Sequência com Séries de Oligonucleotídeos , Oxirredução , Epitélio Pigmentado Ocular/metabolismo , Degeneração Retiniana/metabolismo , Pigmentos da Retina/biossíntese , Reação em Cadeia da Polimerase Via Transcriptase Reversa/métodos , Especificidade da Espécie
4.
Mycotoxin Res ; 22(1): 61-9, 2006 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-23605503

RESUMO

Fumonisin B1 (FB1) is a mycotoxin produced byFusarium verticillioides that is found in maize and maize-based foods. Reproductive studies in CD1 mice, rats and rabbits initially found no evidence that fumonisins are teratogenic. However, more recent findings suggest that they might increase the risk of neural tube defects (NTDs) in populations consuming large amounts of fumonisin-contaminated corn. When ≥15 mg/kg body weight fumonisin B1 (FB1) was given to pregnant LM/Bc mice by intraperitoneal (ip) injection, all litters were positive for NTDs. To determine if NTD induction is unique to the inbred LM/Bc mouse strain, NTD induction in LM/Bc and CD1 mice was compared: (a) in a study in whichF. verticillioides culture material providing ≤150 ppm FB1 was fed to female mice before and during gestation, and (b) in a study in which FB1 was given by ip injection to CD1 dams on gestation days 7 and 8, the critical time for NTD development. In the feeding study, one of five LM/Bc litters from dams fed the 150 ppm FB1 diet was positive for NTDs whereas no NTDs were found in the CD1 litters. In the ip injection study, 40% of the litters at the highest dose tested, 45 mg/kg body weight, were positive for NTDs and one of nine low-dose (15 mg/kg body weight) litters was also positive. Thus, FB1 induced NTDs in both LM/Bc and CD1 mice although the latter strain appears less sensitive. Comparative investigations using these strains will be useful for elucidating the mechanisms underlying fumonisin-induced NTDs in mice and determining the suitability of mouse models for studying the relationships between fumonisins and NTDs in humans.

5.
Toxicol Appl Pharmacol ; 177(3): 238-46, 2001 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-11749123

RESUMO

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.


Assuntos
Arseniatos/toxicidade , Proteínas de Transporte/metabolismo , Predisposição Genética para Doença , Defeitos do Tubo Neural/induzido quimicamente , Efeitos Tardios da Exposição Pré-Natal , Receptores de Superfície Celular , Teratogênicos/toxicidade , Animais , Arseniatos/metabolismo , Arsenicais/urina , Arsenitos/urina , Ácido Cacodílico/urina , Proteínas de Transporte/genética , Relação Dose-Resposta a Droga , Feminino , Reabsorção do Feto/induzido quimicamente , Receptores de Folato com Âncoras de GPI , Deficiência de Ácido Fólico , Genótipo , Homocisteína/sangue , Camundongos , Camundongos Knockout , Defeitos do Tubo Neural/patologia , Gravidez , Teratogênicos/metabolismo
6.
Semin Pediatr Neurol ; 8(3): 160-4, 2001 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-11575845

RESUMO

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.


Assuntos
Defeitos do Tubo Neural/genética , 5-Metiltetra-Hidrofolato-Homocisteína S-Metiltransferase/genética , 5-Metiltetra-Hidrofolato-Homocisteína S-Metiltransferase/metabolismo , Animais , Modelos Animais de Doenças , Método Duplo-Cego , Feminino , Ácido Fólico/administração & dosagem , Ácido Fólico/genética , Ácido Fólico/metabolismo , Ácido Fólico/uso terapêutico , Humanos , Recém-Nascido , Masculino , Metilação , Metilenotetra-Hidrofolato Redutase (NADPH2) , Camundongos , Defeitos do Tubo Neural/etiologia , Defeitos do Tubo Neural/prevenção & controle , Oxirredutases atuantes sobre Doadores de Grupo CH-NH/genética , Oxirredutases atuantes sobre Doadores de Grupo CH-NH/metabolismo , Placebos , Gravidez , Ensaios Clínicos Controlados Aleatórios como Assunto , Fatores de Risco , Fatores de Transcrição/genética , Vitaminas/administração & dosagem , Vitaminas/uso terapêutico
7.
Ann N Y Acad Sci ; 919: 261-77, 2000.
Artigo em Inglês | MEDLINE | ID: mdl-11083116

RESUMO

Neural tube defects (NTDs) are among the most common of all human congenital defects, with multifactorial etiologies comprising both environmental and genetic components. Several murine model systems have been developed in an effort to elucidate genetic factors regulating expression of NTDs. Strain-dependent differences in susceptibility to teratogenic insults and altered patterns of gene expression observed within the neuroepithelium of affected embryos support the hypothesis that subtle genetic changes can result in NTDs. Since several affected genes are folate-regulated, transgenic knockout mice lacking a functional folate receptor were developed. Nullizygous embryos died in utero with significant morphological defects, supporting the critical role of folic acid in early embryogenesis. While epidemiological studies have not established an association between polymorphisms in the human folate receptor gene and NTDs, it is known that folate supplementation reduces infant NTD risk. Continued efforts are therefore necessary to reveal the mechanism by which folate works and the nature of the gene(s) responsible for human NTDs.


Assuntos
Poluentes Ambientais/toxicidade , Predisposição Genética para Doença/genética , Defeitos do Tubo Neural/induzido quimicamente , Defeitos do Tubo Neural/genética , Receptores de Superfície Celular , Animais , Proteínas de Transporte/genética , Ciclo Celular/genética , Impressões Digitais de DNA , Modelos Animais de Doenças , Desenvolvimento Embrionário e Fetal/genética , Receptores de Folato com Âncoras de GPI , Ácido Fólico/metabolismo , Ácido Fólico/farmacologia , Idade Gestacional , Substâncias de Crescimento/genética , Substâncias de Crescimento/metabolismo , Humanos , Hipertermia Induzida/efeitos adversos , Camundongos , Camundongos Knockout , Defeitos do Tubo Neural/epidemiologia , Defeitos do Tubo Neural/patologia , Polimorfismo Conformacional de Fita Simples , Ácido Valproico/farmacologia
8.
Teratology ; 59(1): 23-34, 1999 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-9988880

RESUMO

In utero exposure to the anticonvulsant drug phenytoin has been shown to alter normal embryonic development, leading to a pattern of dysmorphogenesis known as the Fetal Hydantoin Syndrome. This embryopathy is characterized by growth retardation, microcephaly, mental deficiency, and craniofacial malformations, although the precise mechanism(s) by which phenytoin alters normal developmental pathways remains unknown. To better understand the molecular events involved in the pathogenesis of phenytoin-induced congenital defects, alterations in gene expression were examined during critical periods of craniofacial development. Pregnant SWV mice were administered phenytoin (60 mg/kg/day) from gestational day 6.5 until they were sacrificed at selected developmental time points. Tissue from the craniofacial region of control and exposed embryos was isolated, and samples were subjected to in situ transcription, antisense RNA amplification, and hybridization on reverse Northern blots to quantitatively assess expression of 36 candidate genes. Chronic phenytoin exposure significantly altered expression of several genes at distinct times during morphogenesis. Results of these studies show that expression of the retinoic acid receptors (RAR) alpha, beta, and gamma were significantly increased by phenytoin exposure. Elevations in gene expression of laminin beta 1, and the growth factors IGF-2, TGF alpha, and TGF beta 1, were also demonstrated in the craniofacial region of phenytoin-exposed embryos. As several of these genes are transcriptionally regulated by retinoic-acid-responsive elements in their promoter regions, phenytoin-induced alterations in expression of the RAR isoforms may have severe downstream consequences in the regulation of events necessary for normal craniofacial development. Such alterations occurring coordinately at critical times during craniofacial development may account for the dysmorphogenesis often associated with phenytoin exposure.


Assuntos
Anticonvulsivantes/toxicidade , Expressão Gênica/efeitos dos fármacos , Fenitoína/toxicidade , Anormalidades Induzidas por Medicamentos/genética , Animais , DNA Complementar , Desenvolvimento Embrionário e Fetal/efeitos dos fármacos , Face/embriologia , Feminino , Camundongos , Camundongos Endogâmicos , Morfogênese , Gravidez , Receptores do Ácido Retinoico/genética , Crânio/embriologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA