Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
1.
Neurochem Int ; 138: 104778, 2020 09.
Artigo em Inglês | MEDLINE | ID: mdl-32474175

RESUMO

Neurotrophins play a critical role in the development, maintenance, and proper function of the brain. We investigated the effects of maternal diet high in omega (n)-3 polyunsaturated fatty acids (PUFA) on fatty acids composition and the gene expression of neurotrophins in fetal brain at different gestation stages. Female C57BL/6 mice (7-weeks old, n = 8/group) were fed a diet containing high, low or very low n-3 PUFA (9, 3 or 1% w/w, respectively), with an n-6:n-3 PUFA of 5:1, 20:1 and 40:1, respectively, for two weeks before mating and throughout pregnancy. Animals were sacrificed during pregnancy at gestation day 12.5 and 18.5 to determine placental and fetal-brain fatty acids composition. The gene expressions of endothelial lipase (EL) and plasma membrane fatty acid-binding protein (FABPpm) were measured in the placenta, while major facilitator superfamily domain-containing 2a (Mfsd2a), brain-derived neurotrophic factor (BDNF), tropomyosin-receptor kinase (TrK)-B, and cAMP response element-binding protein (CREB) were measured in fetal-brain, using qPCR. The protein expression of phosphorylated CREB (pCREB) was determined using ELISA. The high n-3 PUFA diet increased the mRNA expression of EL, FABPpm, and Mfsd2a at both gestation days, compared to other groups. Docosahexaenoic acid (DHA) and total n-3 PUFA were significantly higher in the high n-3 PUFA group, compared to the other groups at both gestation days. The high n-3 PUFA diet also increased the mRNA expressions of BDNF, TrKB and CREB, as well as the protein concentration of pCREB as gestation progressed, compared to the other groups. Our findings show for the first time that maternal diet high in n-3 PUFA increased the mRNA expression of Mfsd2a, which correlated with an increase in DHA accretion in the fetal-brain. A diet high in n-3 PUFA increased neurotrophin signalling in fetal-brain as gestation progressed, demonstrating the importance of n-3 PUFA during brain development.


Assuntos
Gorduras na Dieta/administração & dosagem , Ácidos Graxos Ômega-3/administração & dosagem , Desenvolvimento Fetal/fisiologia , Fatores de Crescimento Neural/genética , Fatores de Crescimento Neural/metabolismo , Regulação para Cima/fisiologia , Animais , Encéfalo/efeitos dos fármacos , Encéfalo/embriologia , Encéfalo/metabolismo , Feminino , Desenvolvimento Fetal/efeitos dos fármacos , Saúde Materna , Camundongos , Camundongos Endogâmicos C57BL , Placenta/efeitos dos fármacos , Placenta/embriologia , Placenta/metabolismo , Gravidez , Efeitos Tardios da Exposição Pré-Natal/genética , Efeitos Tardios da Exposição Pré-Natal/metabolismo , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/fisiologia , Regulação para Cima/efeitos dos fármacos
2.
Endocrinology ; 161(6)2020 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-32270179

RESUMO

Vitamin D insufficiency during pregnancy is widespread. The effects of active vitamin D on the human placenta in vivo are unknown. We test the hypotheses that 25(OH)D sufficiency (arbitrarily defined as 25(OH)D ≥32 ng/mL) modulates placental structure and function in vivo in a population of women whose offspring are at risk for childhood asthma, and that placental pathology is more common in offspring that evolve asthma at age 3. Pregnant volunteers in the St. Louis, MO, cohort of the Vitamin D Antenatal Asthma Reduction Trial (VDAART, NIH grant #HL091528) participated in a nested case-control study and consented for the study of placentas after delivery. Maternal concentrations of 25(OH)D were measured at trial entry and in the third trimester. The histopathology of the placentas from women with sufficient 25(OH)D, versus insufficient, showed no clinically significant differences, but morphometry revealed villi of women with sufficient third-trimester 25(OH)D had a higher villous surface density. Notably, analyses of transcripts, extracted from formalin-fixed paraffin-embedded specimens, revealed higher expression of INTS9, vWF, MACC1, and ARMS2, and diminished expression of the CNTN5 genes in the insufficient group. A larger proportion of placentas showed chronic chorioamnionitis in offspring with versus without asthma at age 3. These findings suggest that maternal 25(OH)D insufficiency has a limited effect on human placental villous histopathology and morphometry, but attenuates a small number of placental gene expression profiles in this selected population. The association of placental chronic chorioamnionitis and offspring asthma is worthy of further study.


Assuntos
Corioamnionite/tratamento farmacológico , Placenta/anatomia & histologia , Deficiência de Vitamina D/tratamento farmacológico , Vitamina D/administração & dosagem , Adulto , Asma/epidemiologia , Estudos de Casos e Controles , Pré-Escolar , Corioamnionite/genética , Corioamnionite/metabolismo , Corioamnionite/patologia , Suplementos Nutricionais/análise , Feminino , Humanos , Masculino , Placenta/efeitos dos fármacos , Placenta/embriologia , Placenta/patologia , Gravidez , Proteínas/genética , Proteínas/metabolismo , Transativadores/genética , Transativadores/metabolismo , Estados Unidos/epidemiologia , Vitamina D/sangue , Deficiência de Vitamina D/sangue , Deficiência de Vitamina D/metabolismo , Deficiência de Vitamina D/patologia , Adulto Jovem
3.
Eur J Neurosci ; 42(10): 2742-60, 2015 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-26215319

RESUMO

The prevalence of autism spectrum disorders (ASDs) has been on the rise over recent years. The presence of diverse subsets of candidate genes in each individual with an ASD and the vast variability of phenotypical differences suggest that the interference of an exogenous environmental component may greatly contribute to the development of ASDs. The lipid mediator prostaglandin E2 (PGE2 ) is released from phospholipids of cell membranes, and is important in brain development and function; PGE2 is involved in differentiation, synaptic plasticity and calcium regulation. The previous review already described extrinsic factors, including deficient dietary supplementation, and exposure to oxidative stress, infections and inflammation that can disrupt signaling of the PGE2 pathway and contribute to ASDs. In this review, the structure and establishment of two key protective barriers for the brain during early development are described: the blood-brain barrier; and the placental barrier. Then, the first comprehensive summary of other environmental factors, such as exposure to chemicals in air pollution, pesticides and consumer products, which can also disturb PGE2 signaling and increase the risk for developing ASDs is provided. Also, how these exogenous agents are capable of crossing the protective barriers of the brain during critical developmental periods when barrier components are still being formed is described. This review underlines the importance of avoiding or limiting exposure to these factors during vulnerable periods in development.


Assuntos
Transtorno do Espectro Autista/metabolismo , Barreira Hematoencefálica/metabolismo , Encéfalo/metabolismo , Dinoprostona/metabolismo , Exposição Ambiental/efeitos adversos , Placenta/metabolismo , Efeitos Tardios da Exposição Pré-Natal/metabolismo , Animais , Barreira Hematoencefálica/embriologia , Encéfalo/embriologia , Proteínas de Transporte/metabolismo , Membrana Celular/química , Membrana Celular/metabolismo , Feminino , Humanos , Placenta/embriologia , Gravidez , Fatores de Risco , Transdução de Sinais
4.
Proc Natl Acad Sci U S A ; 112(22): 6834-40, 2015 Jun 02.
Artigo em Inglês | MEDLINE | ID: mdl-25404322

RESUMO

Mammalian viviparity (intrauterine development of the fetus) introduced a new dimension to brain development, with the fetal hypothalamus and fetal placenta developing at a time when the fetal placenta engages hypothalamic structures of the maternal generation. Such transgenerational interactions provide a basis for ensuring optimal maternalism in the next generation. This success has depended on genomic imprinting and a biased role of the matriline. Maternal methylation imprints determine parent of origin expression of genes fundamental to both placental and hypothalamic development. The matriline takes a further leading role for transgenerational reprogramming of these imprints. Developmental errors are minimized by the tight control that imprinted genes have on regulation of downstream evolutionary expanded gene families important for placental and hypothalamic development. Imprinted genes themselves have undergone purifying selection, providing a framework of stability for in utero development with most growth variance occurring postnatally. Mothers, not fathers, take the lead in the endocrinological and behavior adaptations that nurture, feed, and protect the infant. In utero coadaptive development of the placenta and hypothalamus has thus required a concomitant development to ensure male masculinization. Only placental male mammals evolved the sex determining SRY, which activates Sox9 for testes formation. SRY is a hybrid gene of Dgcr8 expressed in the developing placenta and Sox3 expressed in hypothalamic development. This hybridization of genes that take their origin from the placenta and hypothalamus has enabled critical in utero timing for the development of fetal Leydig cells, and hence testosterone production for hypothalamic masculinization.


Assuntos
Adaptação Biológica/genética , Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Impressão Genômica/fisiologia , Instabilidade Genômica/genética , Hipotálamo/embriologia , Mamíferos/embriologia , Placenta/embriologia , Caracteres Sexuais , Animais , Feminino , Humanos , Masculino , Gravidez
5.
FASEB J ; 28(4): 1854-69, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24421398

RESUMO

Patients with congenital disorder of glycosylation (CDG), type Ib (MPI-CDG or CDG-Ib) have mutations in phosphomannose isomerase (MPI) that impair glycosylation and lead to stunted growth, liver dysfunction, coagulopathy, hypoglycemia, and intestinal abnormalities. Mannose supplements correct hypoglycosylation and most symptoms by providing mannose-6-P (Man-6-P) via hexokinase. We generated viable Mpi hypomorphic mice with residual enzymatic activity comparable to that of patients, but surprisingly, these mice appeared completely normal except for modest (~15%) embryonic lethality. To overcome this lethality, pregnant dams were provided 1-2% mannose in their drinking water. However, mannose further reduced litter size and survival to weaning by 40 and 66%, respectively. Moreover, ~50% of survivors developed eye defects beginning around midgestation. Mannose started at birth also led to eye defects but had no effect when started after eye development was complete. Man-6-P and related metabolites accumulated in the affected adult eye and in developing embryos and placentas. Our results demonstrate that disturbing mannose metabolic flux in mice, especially during embryonic development, induces a highly specific, unanticipated pathological state. It is unknown whether mannose is harmful to human fetuses during gestation; however, mothers who are at risk for having MPI-CDG children and who consume mannose during pregnancy hoping to benefit an affected fetus in utero should be cautious.


Assuntos
Cegueira/etiologia , Suplementos Nutricionais/toxicidade , Manose-6-Fosfato Isomerase/metabolismo , Manose/toxicidade , Animais , Cegueira/genética , Cegueira/metabolismo , Western Blotting , Células Cultivadas , Defeitos Congênitos da Glicosilação/genética , Defeitos Congênitos da Glicosilação/metabolismo , Embrião de Mamíferos/citologia , Embrião de Mamíferos/efeitos dos fármacos , Embrião de Mamíferos/metabolismo , Olho/embriologia , Olho/crescimento & desenvolvimento , Olho/metabolismo , Feminino , Humanos , Imuno-Histoquímica , Masculino , Manose/sangue , Manose/metabolismo , Manose-6-Fosfato Isomerase/genética , Manosefosfatos/metabolismo , Camundongos , Camundongos da Linhagem 129 , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Placenta/efeitos dos fármacos , Placenta/embriologia , Placenta/metabolismo , Gravidez
6.
Biol Trace Elem Res ; 154(3): 338-44, 2013 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-23857380

RESUMO

Placental type 3 iodothyronine deiodinase (D3) potentially protects the fetus from the elevated maternal thyroid hormones. Na(+)/I(-) symporter (NIS) is a plasma membrane glycoprotein, which mediates active iodide uptake. Our objectives were to establish the distribution of NIS and D3 gene expressions in the placenta and the amniotic membrane and to investigate the relationship between placental D3 and NIS gene expressions and maternal iodine, selenium, and thyroid hormone status. Thyroid hormones, urinary iodine concentration (UIC), and selenium levels were measured in 49 healthy term pregnant women. NIS and D3 gene expressions were studied with the total mRNA RT-PCR method in tissues from maternal placenta (n = 49), fetal placenta (n = 9), and amniotic membrane (n = 9). NIS and D3 gene expressions were shown in the fetal and maternal sides of the placenta and amniotic membrane. Mean blood selenium level was 66 ± 26.5 µg/l, and median UIC was 143 µg/l. We could not demonstrate any statistically significant relationship of spot UIC and blood selenium with NIS and D3 expression (p > 0.05). Positive correlations were found between NIS and thyroxine-binding globulin (TBG) (r = 0.3, p = 0.042) and between D3 and preoperative glucose levels (r = 0.4, p = 0.006). D3 and NIS genes are expressed in term placenta and amniotic membrane; thus, in addition to placenta, amniotic membrane contributes to regulation of maternofetal iodine and thyroid hormone transmission. Further studies are needed to clarify the relationship between maternal glucose levels and placental D3 expression and between TBG and placental NIS expression.


Assuntos
Âmnio/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Iodeto Peroxidase/genética , Placenta/metabolismo , Simportadores/genética , Hormônios Tireóideos/sangue , Adulto , Âmnio/embriologia , Feminino , Idade Gestacional , Humanos , Iodo/metabolismo , Iodo/urina , Placenta/embriologia , Gravidez , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Selênio/sangue , Globulina de Ligação a Tiroxina/análise , Globulina de Ligação a Tiroxina/genética
7.
FASEB J ; 27(8): 3249-56, 2013 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-23651543

RESUMO

Selenoprotein P (Sepp1) is taken up by receptor-mediated endocytosis for its selenium. The other extracellular selenoprotein, glutathione peroxidase-3 (Gpx3), has not been shown to transport selenium. Mice with genetic alterations of Sepp1, the Sepp1 receptors apolipoprotein E receptor-2 (apoER2) and megalin, and Gpx3 were used to investigate maternal-fetal selenium transfer. Immunocytochemistry (ICC) showed receptor-independent uptake of Sepp1 and Gpx3 in the same vesicles of d-13 visceral yolk sac cells, suggesting uptake by pinocytosis. ICC also showed apoER2-mediated uptake of maternal Sepp1 in the d-18 placenta. Thus, two selenoprotein-dependent maternal-fetal selenium transfer mechanisms were identified. Selenium was quantified in d-18 fetuses with the mechanisms disrupted. Maternal Sepp1 deletion, which lowers maternal whole-body selenium, decreased fetal selenium under selenium-adequate conditions but deletion of fetal apoER2 did not. Fetal apoER2 deletion did decrease fetal selenium, by 51%, under selenium-deficient conditions, verifying function of the placental Sepp1-apoER2 mechanism. Maternal Gpx3 deletion decreased fetal selenium, by 13%, but only under selenium-deficient conditions. These findings indicate that the selenoprotein uptake mechanisms ensure selenium transfer to the fetus under selenium-deficient conditions. The failure of their disruptions (apoER2 deletion, Gpx3 deletion) to affect fetal selenium under selenium-adequate conditions indicates the existence of an additional maternal-fetal selenium transfer mechanism.


Assuntos
Glutationa Peroxidase/metabolismo , Proteínas Relacionadas a Receptor de LDL/metabolismo , Proteína-2 Relacionada a Receptor de Lipoproteína de Baixa Densidade/metabolismo , Troca Materno-Fetal/fisiologia , Selênio/metabolismo , Selenoproteína P/metabolismo , Animais , Transporte Biológico , Feminino , Glutationa Peroxidase/genética , Imuno-Histoquímica , Proteínas Relacionadas a Receptor de LDL/genética , Proteína-2 Relacionada a Receptor de Lipoproteína de Baixa Densidade/genética , Masculino , Troca Materno-Fetal/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Microscopia de Fluorescência , Placenta/embriologia , Placenta/metabolismo , Gravidez , Selenoproteína P/genética , Fatores de Tempo , Saco Vitelino/embriologia , Saco Vitelino/metabolismo
8.
J Pregnancy ; 2013: 320413, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23691322

RESUMO

Extrapolating from animal studies to human pregnancy, our studies showed that folate (FA) deficiency as well as one-time exposure to environmental factors in the first two to three weeks of human gestation can result in severe congenital heart defects (CHDs). Considering that approximately 49% of pregnancies are unplanned, this period of pregnancy can be considered high-risk for cardiac, as well as for neural, birth defects, as the woman usually is not aware of her pregnancy and may not yet be taking precautionary actions to protect the developing embryo. Using avian and mouse vertebrate models, we demonstrated that FA supplementation prevents CHD induced by alcohol, lithium, or elevation of the metabolite homocysteine, a marker for FA deficiency. All three factors affected the important Wnt signaling pathway by suppressing Wnt-mediated gene expression in the heart fields, resulting in a delay of cardiomyocyte migration, cardiomyogenesis, and CHD. Optimal protection of cardiogenesis was observed to occur with FA supplementation provided upon morning after conception and at higher doses than the presently available in prenatal vitamin supplementation. Our studies demonstrate pathways and cell processes that are involved with protection of one-carbon metabolism during heart development.


Assuntos
Ácido Fólico/farmacologia , Cardiopatias Congênitas/prevenção & controle , Coração/embriologia , Placenta/embriologia , Animais , Aves , Diferenciação Celular/fisiologia , Suplementos Nutricionais , Modelos Animais de Doenças , Etanol/toxicidade , Feminino , Ácido Fólico/administração & dosagem , Deficiência de Ácido Fólico/dietoterapia , Deficiência de Ácido Fólico/embriologia , Deficiência de Ácido Fólico/prevenção & controle , Idade Gestacional , Cardiopatias Congênitas/induzido quimicamente , Cardiopatias Congênitas/embriologia , Homocisteína/toxicidade , Humanos , Compostos de Lítio/toxicidade , Exposição Materna , Camundongos , Miócitos Cardíacos/citologia , Gravidez , Efeitos Tardios da Exposição Pré-Natal/prevenção & controle , Transdução de Sinais , Proteínas Wnt/fisiologia
9.
Proc Natl Acad Sci U S A ; 108(37): 15237-41, 2011 Sep 13.
Artigo em Inglês | MEDLINE | ID: mdl-21810990

RESUMO

The fetal genome regulates maternal physiology and behavior via its placenta, which produces hormones that act on the maternal hypothalamus. At the same time, the fetus itself develops a hypothalamus. In this study we show that many of the genes that regulate placental development also regulate the developing hypothalamus, and in mouse the coexpression of these genes is particularly high on embryonic days 12 and 13 (days E12-13). Such synchronized expression is regulated, in part, by the maternally imprinted gene, paternally expressed gene 3 (Peg3), which also is developmentally coexpressed in the hypothalamus and placenta at days E12-13. We further show that challenging this genomic linkage of hypothalamus and placenta with 24-h food deprivation results in disruption to coexpressed genes, primarily by affecting placental gene expression. Food deprivation also produces a significant decrease in Peg3 gene expression in the placenta, with consequences similar to many of the placental gene changes induced by Peg3 mutation. Such genomic dysregulation does not occur in the hypothalamus, where Peg3 expression increases with food deprivation. Thus, changes in gene expression brought about by food deprivation are consistent with the fetal genome's maintaining hypothalamic development at a cost to its placenta. This biased change to gene dysregulation in the placenta is linked to autophagy and ribosomal turnover, which sustain, in the short term, nutrient supply for the developing hypothalamus. Thus, the fetus controls its own destiny in times of acute starvation by short-term sacrifice of the placenta to preserve brain development.


Assuntos
Encéfalo/embriologia , Feto/embriologia , Privação de Alimentos/fisiologia , Placenta/embriologia , Animais , Autofagia/genética , Encéfalo/metabolismo , Sincronização Cortical/genética , Feminino , Feto/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Redes Reguladoras de Genes/genética , Hipotálamo/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Placenta/citologia , Placenta/metabolismo , Gravidez , Fatores de Tempo
10.
Endocrinology ; 131(5): 2120-6, 1992 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-1425412

RESUMO

Glucocorticoids promote the development of many organ systems vital for extrauterine survival, and fetal cortisol provides the trigger for birth in sheep. The activity of glucocorticoids may be influenced at a cellular level by 11 beta-hydroxysteroid dehydrogenase (11 beta-HSD), which is responsible for the interconversion of cortisol and cortisone. To examine 11 beta-HSD gene expression during fetal development, two overlapping clones which yield a 1.4 kilobase (kb) complementary DNA encoding sheep 11 beta-HSD from a liver library were isolated by using a rat 11 beta-HSD cDNA as the probe. This cDNA contains a 879 base pair open reading frame for a protein of 292 amino acids that has more than 70% sequence identity to rat and human 11 beta-HSDs. To define the tissue distribution of 11 beta-HSD messenger RNA in sheep, selected tissues were collected from one fetus at day 130 and term (approximately 145 days), and from a nonpregnant ewe. Cellular RNA was extracted and subjected to Northern blot analysis, and a single 1.8 kb transcript was detected in the fetal and adult liver, lung, hypothalamus, anterior pituitary, and placenta. This was undetectable in adrenals and kidneys, but a smaller (1.5 kb) transcript was present in fetal and adult kidney RNA. The relative abundance of 11 beta-HSD mRNA was greatest in fetal and adult livers, and it was much higher in adult liver, lung, and kidney than in the corresponding fetal tissues. To examine whether 11 beta-HSD gene expression is developmentally regulated in the fetal sheep, liver, lung, and kidney tissues were taken from fetuses at day 60-70, day 100-110, day 125-130, at term, and from newborn lambs (24-48 h old). In the lung and kidney, the relative abundance of 11 beta-HSD mRNA did not change from day 60 to term but increased in the lungs of newborn lambs. In contrast, 11 beta-HSD mRNA levels in the liver increased between day 125 and term and rose further in the newborn. Collectively, these results demonstrate that 11 beta-HSD gene expression in sheep is regulated in a tissue-specific and developmentally programmed manner.


Assuntos
Animais Recém-Nascidos/genética , DNA/genética , Desenvolvimento Embrionário e Fetal/genética , Hidroxiesteroide Desidrogenases/genética , Fígado/química , RNA Mensageiro/análise , 11-beta-Hidroxiesteroide Desidrogenases , Glândulas Suprarrenais/química , Glândulas Suprarrenais/embriologia , Glândulas Suprarrenais/enzimologia , Animais , Sequência de Bases , Northern Blotting , Clonagem Molecular , DNA/análise , Feminino , Feto/química , Expressão Gênica , Hipotálamo/química , Hipotálamo/embriologia , Hipotálamo/enzimologia , Fígado/embriologia , Fígado/enzimologia , Pulmão/química , Pulmão/embriologia , Pulmão/enzimologia , Dados de Sequência Molecular , Adeno-Hipófise/química , Adeno-Hipófise/embriologia , Adeno-Hipófise/enzimologia , Placenta/química , Placenta/embriologia , Placenta/enzimologia , Gravidez , RNA Mensageiro/genética , Ovinos , Distribuição Tecidual
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA