Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 26
Filtrar
Más filtros

País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Genes (Basel) ; 12(10)2021 10 17.
Artículo en Inglés | MEDLINE | ID: mdl-34681028

RESUMEN

The maternal environment during the periconceptional period influences foetal growth and development, in part, via epigenetic mechanisms moderated by one-carbon metabolic pathways. During embryonic development, one-carbon metabolism is involved in brain development and neural programming. Derangements in one-carbon metabolism increase (i) the short-term risk of embryonic neural tube-related defects and (ii) long-term childhood behaviour, cognition, and autism spectrum disorders. Here we investigate the association between maternal one-carbon metabolism and foetal and neonatal brain growth and development. Database searching resulted in 26 articles eligible for inclusion. Maternal vitamin B6, vitamin B12, homocysteine, and choline were not associated with foetal and/or neonatal head growth. First-trimester maternal plasma folate within the normal range (>17 nmol/L) associated with increased foetal head size and head growth, and high erythrocyte folate (1538-1813 nmol/L) with increased cerebellar growth, whereas folate deficiency (<7 nmol/L) associated with a reduced foetal brain volume. Preconceptional folic acid supplement use and specific dietary patterns (associated with increased B vitamins and low homocysteine) increased foetal head size. Although early pregnancy maternal folate appears to be the most independent predictor of foetal brain growth, there is insufficient data to confirm the link between maternal folate and offspring risks for neurodevelopmental diseases.


Asunto(s)
Encéfalo/crecimiento & desarrollo , Carbono/metabolismo , Desarrollo Embrionario/genética , Desarrollo Fetal/genética , Encéfalo/metabolismo , Femenino , Desarrollo Fetal/fisiología , Feto/metabolismo , Feto/fisiología , Ácido Fólico/metabolismo , Humanos , Embarazo , Vitamina B 12/metabolismo
2.
J Trace Elem Med Biol ; 65: 126731, 2021 May.
Artículo en Inglés | MEDLINE | ID: mdl-33610057

RESUMEN

BACKGROUND: Zinc deficiency is associated with adverse effects on maternal health and pregnancy outcomes. These consequences have been reported over the years from zinc supplementation trials and observational studies whereby outcomes of maternal, foetal and infant health were measured. Owing to the importance of zinc in the functions of epigenetic enzymes, pre-clinical studies have shown that its deficiency could disrupt biological activities that involve epigenetic mechanisms in offspring. Thus, this review assessed the link between epigenetics and the effects of maternal zinc deficiency on the offspring's health in animal studies. METHODS: Research articles were retrieved without date restriction from PubMed, Web of Science, ScienceDirect, and Google Scholar databases, as well as reference lists of relevant articles. The search terms used were "zinc deficiency", "maternal zinc deficiency", "epigenetics", and "offspring." Six studies met the eligibility criteria and were reviewed. RESULTS: All the eligible studies reported maternal zinc deficiency and observed changes in epigenetic markers on the progeny during prenatal and postnatal stages of development. The main epigenetic markers reported were global and gene specific methylation and/ or acetylation. The epigenetic changes led to mortality, disruption in development, and risk of later life diseases. CONCLUSION: Maternal zinc deficiency is associated with epigenetic modifications in offspring, which induce pathologies and increase the risk of later life diseases. More research and insight into the epigenetic mechanisms could spring up new approaches to combat the associated disease conditions.


Asunto(s)
Epigénesis Genética/genética , Desarrollo Fetal/genética , Zinc/metabolismo , Animales , Humanos , Zinc/deficiencia
3.
J. obstet. gynaecol. Can ; 43(1): 124-139, Jan. 1, 2021.
Artículo en Inglés | BIGG | ID: biblio-1146592

RESUMEN

This revised guideline is intended to provide an update on the genetic aspects, prevention, screening, diagnosis, and management of fetal neural tube defects. Target population: Women who are pregnant or may become pregnant. Neural tube defect screening should be offered to all pregnant women. For prevention: a folate-rich diet, and folic acid and vitamin B12 supplementation, with dosage depending on risk level. For screening: second-trimester anatomical sonography; first-trimester sonographic screening; maternal serum alpha fetoprotein; prenatal magnetic resonance imaging. For genetic testing: diagnostic amniocentesis with chromosomal microarray and amniotic fluid alpha fetoprotein and acetylcholinesterase; fetal exome sequencing. For pregnancy management: prenatal surgical repair; postnatal surgical repair; pregnancy termination with autopsy. For subsequent pregnancies: prevention and screening options and counselling. The research on and implementation of fetal surgery for prenatally diagnosed myelomeningocele has added a significant treatment option to the previous options (postnatal repair or pregnancy termination), but this new option carries an increased risk of maternal morbidity. Significant improvements in health and quality of life, both for the mother and the infant, have been shown to result from the prevention, screening, diagnosis, and treatment of fetal neural tube defects. The benefits for patient autonomy and decision-making are provided in the guideline. Harms include an unexpected fetal diagnosis and the subsequent management decisions. Harm can also result if the patient declines routine sonographic scans or if counselling and access to care for neural tube defects are delayed. Cost analysis (personal, family, health care) is not within the scope of this clinical practice guideline. A directed and focused literature review was conducted using the search terms spina bifida, neural tube defect, myelomeningocele, prenatal diagnosis, fetal surgery, neural tube defect prevention, neural tube defect screening, neural tube defect diagnosis, and neural tube defect management in order to update and revise this guideline. A peer review process was used for content validation and clarity, with appropriate ethical considerations. The authors rated the quality of evidence and strength of recommendations using the Grading of Recommendations Assessment, Development and Evaluation (GRADE) approach. See online Appendix A (Tables A1 for definitions and A2 for interpretations of strong and weak recommendations). Maternity care professionals who provide any part of pre-conception, antenatal, delivery, and neonatal care. This guideline is also appropriate for patient education.


Asunto(s)
Humanos , Femenino , Embarazo , Complicaciones del Embarazo/prevención & control , Manejo de Atención al Paciente/organización & administración , Desarrollo Fetal/genética , Defectos del Tubo Neural/prevención & control , Adenina/uso terapéutico , Ultrasonografía Prenatal/métodos , Ácido Fólico/uso terapéutico
4.
J Anim Sci ; 98(8)2020 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-32687162

RESUMEN

To determine the effects of maternal supplementation on the mRNA abundance of genes associated with metabolic function in fetal muscle and liver, pregnant sows (Landrace × Yorkshire; initial body weight (BW) 221.58 ± 33.26 kg; n = 21) fed a complete gestation diet (corn-soybean meal based diet, CSM) were randomly assigned to 1 of 4 isocaloric supplementation treatments: control (CON, 378 g/d CSM, n = 5), sucrose (SUGAR, 255 g/d crystalized sugar, n = 5), cooked ground beef (BEEF, 330 g/d n = 6), or BEEF + SUGAR (B+S, 165 g/d cooked ground beef and 129 g/d crystalized sugar, n = 5), from days 40 to 110 of gestation. Sows were euthanized on day 111 of gestation. Two male and 2 female fetuses of median BW were selected from each litter, and samples of the longissimus dorsi muscle and liver were collected. Relative transcript level was quantified via qPCR with HPRT1 as the reference gene for both muscle and liver samples. The following genes were selected and analyzed in the muscle: IGF1R, IGF2, IGF2R, GYS-1, IRS-1, INSR, SREBP-1C, and LEPR; while the following were analyzed in the liver: IGF2, IGF2R, FBFase, G6PC, PC, PCK1, FGF21, and LIPC. No effect of fetal sex by maternal treatment interaction was observed in mRNA abundance of any of the genes evaluated (P > 0.11). In muscle, the maternal nutritional treatment influenced (P = 0.02) IGF2 mRNA abundance, with B+S and SUGAR fetuses having lower abundance than CON, which was not different from BEEF. Additionally, SREBP-1 mRNA abundance was greater (P < 0.01) for B+S compared with CON, BEEF, or SUGAR fetuses; and females tended (P = 0.06) to have an increased abundance of SREBP-1 than males. In fetal liver, IGF2R mRNA abundance was greater (P = 0.01) for CON and BEEF than SUGAR and B+S; while FBPase mRNA abundance was greater (P = 0.03) for B+S compared with the other groups. In addition, maternal nutritional tended (P = 0.06) to influence LIPC mRNA abundance, with increased abundance in CON compared with SUGAR and B+S. These data indicate limited changes in transcript abundance due to substitution of supplemental sugar by ground beef during mid to late gestation. However, the differential expression of FBPase and SREBP-1c in response to the simultaneous supplementation of sucrose and ground beef warrants further investigations, since these genes may play important roles in determining the offspring susceptibility to metabolic diseases.


Asunto(s)
Suplementos Dietéticos/análisis , Desarrollo Fetal/efectos de los fármacos , Factor II del Crecimiento Similar a la Insulina/genética , Carne Roja/análisis , Sacarosa/administración & dosificación , Porcinos/fisiología , Alimentación Animal/análisis , Animales , Peso Corporal/efectos de los fármacos , Dieta/veterinaria , Femenino , Desarrollo Fetal/genética , Hígado/efectos de los fármacos , Masculino , Músculos/efectos de los fármacos , Embarazo , ARN Mensajero/genética , Porcinos/genética , Porcinos/crecimiento & desarrollo
5.
Nutrients ; 12(6)2020 Jun 04.
Artículo en Inglés | MEDLINE | ID: mdl-32512764

RESUMEN

Small-for-gestational-age (SGA) is associated with significant perinatal morbidity and mortality. Our aim was to investigate gene-nutrient interactions between maternal one-carbon single nucleotide polymorphisms (SNPs) and folic acid supplement (FAS) use, and their association with SGA. Nulliparous New Zealand women with singleton pregnancy were recruited as part of the Screening for Pregnancy Endpoints prospective cohort study. Data on FAS use was collected via face-to-face interview at 15 weeks' gestation; participants were followed prospectively and birth outcome data collected within 72 h of delivery. Participants were genotyped for MTHFR 677, MTHFR 1298, MTHFD1 1958, MTR 2756, MTRR 66 and TCN2 776 SNPs. Genotype data for at least one SNP was available for 1873 (93%) of eligible participants. Analysis showed a significant SNP-FAS interaction for MTHFR 1298 (p = 0.020), MTHFR 677 (p = 0.019) and TCN2 776 (p = 0.017) in relation to SGA: MTHFR 1298 CC variant non-FAS users had an increased likelihood [Odds Ratio (OR) = 2.91 (95% Confidence Interval (CI) = 1.52, 5.60] compared with wild-type (MTHFR 1298 AA) FAS users. MTHFR 677 variant allele carrier (MTHFR 677 CT + MTHFR 677 TT) non-FAS users had an increased likelihood [OR = 1.87 (95% CI = 1.21, 2.88)] compared to wild-type (MTHFR 677 CC) FAS users. TCN2 776 variant (TCN2 776 GG) non-FAS users had an increased likelihood [OR = 2.16 (95% CI = 1.26, 3.71)] compared with wild type homozygote + heterozygote (TCN2 776 CC + TCN2 776 CG) FAS users. No significant interactions were observed for MTHFD1 1958, MTR 2756 or MTRR 66 (p > 0.05). We observed an overall pattern of FAS attenuating differences in the likelihood of SGA seen between genotype groups in FAS non-users. Future research should focus on how intake of other one-carbon nutrients might mediate these gene-nutrient interactions.


Asunto(s)
Suplementos Dietéticos , Desarrollo Fetal/genética , Desarrollo Fetal/fisiología , Ácido Fólico/administración & dosificación , Genotipo , Recién Nacido Pequeño para la Edad Gestacional , Fenómenos Fisiologicos Nutricionales Maternos/genética , Fenómenos Fisiologicos Nutricionales Maternos/fisiología , Nutrigenómica , Polimorfismo de Nucleótido Simple , 5-Metiltetrahidrofolato-Homocisteína S-Metiltransferasa/genética , Adulto , Femenino , Ferredoxina-NADP Reductasa/genética , Humanos , Metilenotetrahidrofolato Deshidrogenasa (NADP)/genética , Metilenotetrahidrofolato Reductasa (NADPH2)/genética , Antígenos de Histocompatibilidad Menor/genética , Nueva Zelanda , Embarazo , Estudios Prospectivos , Adulto Joven
6.
Nutrients ; 12(5)2020 May 19.
Artículo en Inglés | MEDLINE | ID: mdl-32438566

RESUMEN

Fetal brain development is closely dependent on maternal nutrition and metabolic status. Maternal protein restriction (PR) is known to be associated with alterations in the structure and function of the hypothalamus, leading to impaired control of energy homeostasis and food intake. The objective of this study was to identify the cellular and molecular systems underlying these effects during fetal development. We combined a global transcriptomic analysis on the fetal hypothalamus from a rat model of maternal PR with in vitro neurosphere culture and cellular analyses. Several genes encoding proteins from the mitochondrial respiratory chain complexes were overexpressed in the PR group and mitochondrial metabolic activity in the fetal hypothalamus was altered. The level of the N6-methyladenosine epitranscriptomic mark was reduced in the PR fetuses, and the expression of several genes involved in the writing/erasing/reading of this mark was indeed altered, as well as genes encoding several RNA-binding proteins. Additionally, we observed a higher number of neuronal-committed progenitors at embryonic day 17 (E17) in the PR fetuses. Together, these data strongly suggest a metabolic adaptation to the amino acid shortage, combined with the post-transcriptional control of protein expression, which might reflect alterations in the control of the timing of neuronal progenitor differentiation.


Asunto(s)
Dieta con Restricción de Proteínas/efectos adversos , Feto/metabolismo , Hipotálamo/embriología , Fenómenos Fisiologicos Nutricionales Maternos/genética , Mitocondrias/genética , Animales , Femenino , Desarrollo Fetal/genética , Hipotálamo/metabolismo , Embarazo , Ratas
7.
Sci Adv ; 6(1): eaay6350, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31911949

RESUMEN

Temperature homeostasis is critical for fetal development. The heat sensor protein TRPM2 (transient receptor potential channel M2) plays crucial roles in the heat response, but its function and specific mechanism in brain development remain largely unclear. Here, we observe that TRPM2 is expressed in neural stem cells. In hyperthermia, TRPM2 knockdown and knockout reduce the proliferation of neural progenitor cells (NPCs) and, accordingly, increase premature cortical neuron differentiation. In terms of the mechanism, TRPM2 regulates neural progenitor self-renewal by targeting SP5 (specificity protein 5) via inhibiting the phosphorylation of ß-catenin and increasing ß-catenin expression. Furthermore, the constitutive expression of TRPM2 or SP5 partly rescues defective NPC proliferation in the TRPM2-deficient embryonic brain. Together, the data suggest that TRPM2 has a critical function in maintaining the NPC pool during heat stress, and the findings provide a framework for understanding how the disruption of the TRPM2 gene may contribute to neurological disorders.


Asunto(s)
Desarrollo Fetal/genética , Neurogénesis/genética , Canales Catiónicos TRPM/genética , Factores de Transcripción/genética , beta Catenina/genética , Animales , Encéfalo/crecimiento & desarrollo , Encéfalo/patología , Diferenciación Celular/genética , Proliferación Celular/genética , Autorrenovación de las Células/genética , Regulación del Desarrollo de la Expresión Génica/genética , Homeostasis , Humanos , Hipertermia Inducida , Ratones , Células-Madre Neurales/metabolismo , Células-Madre Neurales/patología , Neuronas/metabolismo , Neuronas/patología
8.
Brain Dev ; 41(7): 567-576, 2019 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-30954358

RESUMEN

BACKGROUNDS: Metabotropic glutamate receptors, besides ionotropic receptors, mediate the complicated effect of glutamate on neurogenesis. Previous studies showed that metabotropic glutamate receptor 4 (mGluR4) regulated the proliferation and differentiation of neural stem/progenitor cells in vitro. However, little is known about the expression pattern of mGluR4 on prenatal central nervous system in vivo, especially the human being. METHODS: The normal brain tissues of human fetus were collected and divided into 4 groups according to the gestational age: 9-11 W, 14-16 W, 22-24 W and 32-36 W. Then the expression of mGluR4 was evaluated at mRNA and protein levels by means of PCR or immunohistochemistry method, respectively. The type of cell expressing mGluR4 was further investigated using double-labeling immunofluorescence. RESULTS: RT-PCR showed that the mRNA of mGluR4 could be detected in frontal lobe from 9 W to 32 W and real-time PCR quantificationally demonstrated the mRNA increased with development. Similarly, immnoreactivity was found in all layers of frontal lobe, VZ/SVZ. The intensity scores analysis showed that the staining became stronger and the range extended gradually with development. The double-labeling immunofluorescence showed that mGluR4 was present in neural stem/progenitor cells (nestin-positive cells after 9 W), young neurons (DCX-positive cells after 9 W), mature neurons (NeuN-positive cells in cortex after 32 W), as well as typical astrocytes (GFAP-positive cells in medulla after 32 W). CONCLUSION: These results supply an important evidence that mGluR4 is expressed in prenatal human cerebrum, and main kinds of cells related to neurogenesis are involved in its expression.


Asunto(s)
Encéfalo/embriología , Lóbulo Frontal/embriología , Receptores de Glutamato Metabotrópico/metabolismo , Encéfalo/metabolismo , Diferenciación Celular/fisiología , Sistema Nervioso Central/citología , Sistema Nervioso Central/embriología , Sistema Nervioso Central/metabolismo , Corteza Cerebral/citología , Corteza Cerebral/embriología , Corteza Cerebral/metabolismo , Femenino , Desarrollo Fetal/genética , Lóbulo Frontal/citología , Lóbulo Frontal/metabolismo , Ácido Glutámico/metabolismo , Humanos , Inmunohistoquímica , Masculino , Células-Madre Neurales/citología , Células-Madre Neurales/metabolismo , Neurogénesis/fisiología , Neuronas/metabolismo , Embarazo , Receptores de Glutamato Metabotrópico/genética
9.
Reproduction ; 157(1): 13-25, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-30394704

RESUMEN

A number of studies have demonstrated effects of gestational undernutrition on fetal ovarian development and postnatal female fertility. However, the mechanism underlying these effects remains elusive. Using a cohort of animals in which altered gestational nutrition affected indicators of postnatal fertility, this study applies RNAseq to fetal ovaries to identify affected genes and pathways that may underlie the relationship between gestational plane of nutrition and postnatal fertility. Pregnant ewes were exposed to either a maintenance diet or 0.6 of maintenance for the first 55 days of gestation followed by an ad libitum diet. Complementary DNA libraries were constructed from 5 to 6 fetal ovaries from each nutritional group at both days 55 and 75 of gestation and sequenced using Ion Proton. Of approximately 16,000 transcripts, 69 genes were differentially expressed at day 55 and 145 genes differentially expressed at day 75. At both gestational ages, genes expressed preferentially in germ cells were common among the differentially expressed genes. Enriched gene ontology terms included ion transport, nucleic acid binding, protease inhibitor activity and carrier proteins of the albumin family. Affected pathways identified by IPA analysis included LXR/RXR activation, FXR/RXR activation, pathways associated with nitric oxide production and citrullination (by NOS1), vitamin C transport and metabolism and REDOX reactions. The data offer some insights into potential mechanisms underlying the relationship between gestational plane of nutrition and postnatal fertility observed in these animals. In particular, the roles of nitric oxide and protease inhibitors in germ cell development are highlighted and warrant further study.


Asunto(s)
Feto/metabolismo , Desnutrición/genética , Ovario/embriología , Ovario/metabolismo , Fenómenos Fisiologicos de la Nutrición Prenatal , Ovinos , Animales , Femenino , Desarrollo Fetal/genética , Feto/embriología , Regulación del Desarrollo de la Expresión Génica , Edad Gestacional , Desnutrición/metabolismo , Embarazo , Complicaciones del Embarazo/genética , Complicaciones del Embarazo/metabolismo , Fenómenos Fisiologicos de la Nutrición Prenatal/genética , Ovinos/embriología , Ovinos/genética , Ovinos/metabolismo
10.
Basic Clin Pharmacol Toxicol ; 125 Suppl 3: 70-80, 2019 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-30387920

RESUMEN

Much progress has happened in understanding developmental vulnerability to preventable environmental hazards. Along with the improved insight, the perspective has widened, and developmental toxicity now involves latent effects that can result in delayed adverse effects in adults or at old age and additional effects that can be transgenerationally transferred to future generations. Although epidemiology and toxicology to an increasing degree are exploring the adverse effects from developmental exposures in human beings, the improved documentation has resulted in little progress in protection, and few environmental chemicals are currently regulated to protect against developmental toxicity, whether it be neurotoxicity, endocrine disruption or other adverse outcome. The desire to obtain a high degree of certainty and verification of the evidence used for decision-making must be weighed against the costs and necessary duration of research, as well as the long-term costs to human health because of delayed protection of vulnerable early-life stages of human development and, possibly, future generations. Although two-generation toxicology tests may be useful for initial test purposes, other rapidly emerging tools need to be seriously considered from computational chemistry and metabolomics to CLARITY-BPA-type designs, big data and population record linkage approaches that will allow efficient generation of new insight; epigenetic mechanisms may necessitate a set of additional regulatory tests to reveal such effects. As reflected by the Prenatal Programming and Toxicity (PPTOX) VI conference, the current scientific understanding and the timescales involved require an intensified approach to protect against preventable adverse health effects that can harm the next generation and generations to come. While further research is needed, the main emphasis should be on research translation and timely public health intervention to avoid serious, irreversible and perhaps transgenerational harm.


Asunto(s)
Ecotoxicología/métodos , Disruptores Endocrinos/efectos adversos , Exposición a Riesgos Ambientales/efectos adversos , Medicina Ambiental/métodos , Efectos Tardíos de la Exposición Prenatal/diagnóstico , Animales , Macrodatos , Química Computacional/métodos , Congresos como Asunto , Modelos Animales de Enfermedad , Epigénesis Genética/efectos de los fármacos , Epigenómica/métodos , Femenino , Desarrollo Fetal/efectos de los fármacos , Desarrollo Fetal/genética , Humanos , Metabolómica/métodos , Embarazo , Efectos Tardíos de la Exposición Prenatal/etiología , Efectos Tardíos de la Exposición Prenatal/prevención & control , Proyectos de Investigación , Medición de Riesgo/métodos , Factores de Tiempo
11.
Artículo en Inglés | MEDLINE | ID: mdl-30293593

RESUMEN

Poor fetal growth is associated with long-term behavioral, metabolic and psychiatric alterations, including impulsivity, insulin resistance, and mood disorders. However, the consumption of omega-3 polyunsaturated fatty acid (n-3 PUFA) seems to be protective for this population, improving inhibitory control and behavioral reactivity. We investigated whether the presence of the A allele of rs8887 SNP (PLIN4 gene), known to be associated with increased sensitivity to the consumption of n-3 PUFAs, interacts with fetal growth influencing inhibitory control. 152 five-year-old children were genotyped and performed the Stop Signal Task (SSRT). There was a significant interaction between birth weight and the presence of the A allele on SSRT performance, in which lower birth weight associated with poorer inhibitory control only in non-carriers. These results suggest that a higher responsiveness to n-3 PUFAS protects small for gestational age children from developing poor response inhibition, highlighting that optimizing n-3 PUFA intake may benefit this population.


Asunto(s)
Peso al Nacer/efectos de los fármacos , Ácidos Grasos Omega-3/administración & dosificación , Desarrollo Fetal/genética , Perilipina-4/genética , Alelos , Preescolar , Femenino , Desarrollo Fetal/efectos de los fármacos , Estudios de Asociación Genética , Predisposición Genética a la Enfermedad , Humanos , Masculino , Polimorfismo de Nucleótido Simple/genética
12.
Artículo en Inglés | MEDLINE | ID: mdl-29991034

RESUMEN

Growth characteristics during periods of early developmental plasticity are linked with later health outcomes and with disease risks. Infant growth is modulated by genetic and exogenous factors including nutrition. We try to explore their underlying mechanisms using targeted metabolomic profiling of small molecules in biological samples using high-performance liquid chromatography (LC) coupled to tandem mass spectrometry (MS/MS) to quantify hundreds of molecules in small biosamples, e.g., 50 µL plasma. In the large German LISA birth cohort study, cord blood lysophosphatidylcholines and fatty acids were closely associated with infant birth weight, with a nonsignificant trend towards an association with infant weight gain and later BMI. Studies in infants randomized to different protein intakes in the European CHOP Study show conventional high protein intakes to markedly increase plasma-indispensable amino acids (AA), particularly branched-chain AA (BCAA), while exceeding the infant's capacity of BCAA breakdown, and an increase in the dispensable AA tyrosine previously associated with insulin resistance. In a path model analysis of the relationship of infant plasma AA, growth factors, and infant growth, AA were generally found to induce a stronger response of insulin than IGF-I although effects of individual AA were very different. We conclude that targeted improvement in nutrient supply in pregnancy and infancy may offer large opportunities for promoting desirable child growth patterns and long-term health.


Asunto(s)
Desarrollo Infantil/fisiología , Desarrollo Fetal/fisiología , Adolescente , Aminoácidos/sangre , Peso al Nacer , Índice de Masa Corporal , Preescolar , Cromatografía Líquida de Alta Presión/métodos , Proteínas en la Dieta/administración & dosificación , Ácidos Grasos/sangre , Femenino , Sangre Fetal/química , Desarrollo Fetal/genética , Humanos , Lactante , Fenómenos Fisiológicos Nutricionales del Lactante , Recién Nacido , Lisofosfatidilcolinas/sangre , Metabolómica/métodos , Embarazo , Espectrometría de Masas en Tándem/métodos , Aumento de Peso
13.
Diabetes ; 65(3): 574-84, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26631738

RESUMEN

Maternal obesity is a worldwide problem associated with increased risk of metabolic diseases in the offspring. Genetic deletion of the gastric inhibitory polypeptide (GIP) receptor (GIPR) prevents high-fat diet (HFD)-induced obesity in mice due to specific changes in energy and fat cell metabolism. We investigated whether GIP-associated pathways may be targeted by fetal programming and mimicked the situation by exposing pregnant mice to control or HFD during pregnancy (intrauterine [IU]) and lactation (L). Male wild-type (WT) and Gipr(-/-) offspring received control chow until 25 weeks of age followed by 20 weeks of HFD. Gipr(-/-) offspring of mice exposed to HFD during IU/L became insulin resistant and obese and exhibited increased adipose tissue inflammation and decreased peripheral tissue substrate utilization after being reintroduced to HFD, similar to WT mice on regular chow during IU/L. They showed decreased hypothalamic insulin sensitivity compared with Gipr(-/-) mice on control diet during IU/L. DNA methylation analysis revealed increased methylation of CpG dinucleotides and differential transcription factor binding of promoter regions of genes involved in lipid oxidation in the muscle of Gipr(-/-) offspring on HFD during IU/L, which were inversely correlated with gene expression levels. Our data identify GIP-regulated metabolic pathways that are targeted by fetal programming.


Asunto(s)
Dieta Alta en Grasa , Desarrollo Fetal/genética , Polipéptido Inhibidor Gástrico/metabolismo , Hipotálamo/metabolismo , Resistencia a la Insulina/genética , Obesidad/genética , Efectos Tardíos de la Exposición Prenatal/genética , ARN Mensajero/metabolismo , Receptores de la Hormona Gastrointestinal/genética , Tejido Adiposo , Tejido Adiposo Blanco/metabolismo , Tejido Adiposo Blanco/patología , Animales , Islas de CpG , Metilación de ADN , Ensayo de Cambio de Movilidad Electroforética , Femenino , Regulación de la Expresión Génica , Prueba de Tolerancia a la Glucosa , Inflamación , Lactancia , Masculino , Redes y Vías Metabólicas , Ratones , Ratones Noqueados , Músculo Esquelético/metabolismo , Obesidad/metabolismo , Embarazo , Complicaciones del Embarazo/metabolismo , Regiones Promotoras Genéticas , Reacción en Cadena en Tiempo Real de la Polimerasa
14.
Ginekol Pol ; 85(7): 494-9, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-25118499

RESUMEN

INTRODUCTION: Important role is attributed to genetic polymorphisms influencing enzymatic activity in folate metabolism. These inherited genetic variants may influence fetal growth and fetal hypotrophy development. The aim of the study was to investigate the connection of 401A>G polymorphism of methyleneterahydrofolate dehydrogenase gene (MTHFD1) with increased risk of fetal hypotrophy. MATERIAL AND METHODS: To the study group 120 women who delivered children with fetal hypotrophy and to the control group 120 healthy women were enrolled. Study group was divided into subgroups according to gestational age at delivery (52 patients < 37 weeks, 68 patients > or = 37 weeks) and to the neonatal weight (31 mothers of newborns with birth weight < 1500 g, 89 mothers of newborns with birth weight > or = 1500 g). The genetic analysis was performed with the use of PCR/RFLP method. RESULTS: We observed statistically higher occurrence of mutated 401A allele in hypotrophy group (401A: 27,1 vs. 18,8%, OR = 1,61, p = 0,02). At mothers who delivered hypotrophic children weighted more than 1500 g the presence of 401A allele was higher (28,7 vs. 18,8%, OR = 1,74, p = 0,01). Additionally in mothers who delivered hypotrophic children before 37 gestational week statistically higher frequency of 401A allele has been noted (31,7 vs. 18,8%, OR = 2,01, p = 0,007). CONCLUSIONS: Our results indicated that mutated 401A allele of MTHFD1 gene is essential risk factor of fetal hypotrophy in population of Polish women. Appropriate folate supplementation could be particularly essential in women carriers the genetic polymorphism influencing the folate metabolism.


Asunto(s)
Peso al Nacer/genética , Retardo del Crecimiento Fetal/genética , Metilenotetrahidrofolato Deshidrogenasa (NADP)/genética , Mutación , Polimorfismo Genético , Adulto , Femenino , Desarrollo Fetal/genética , Ácido Fólico/metabolismo , Frecuencia de los Genes , Genotipo , Edad Gestacional , Humanos , Recién Nacido , Masculino , Metilenotetrahidrofolato Deshidrogenasa (NADP)/metabolismo , Antígenos de Histocompatibilidad Menor , Embarazo
15.
Hypertens Res ; 37(8): 753-8, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24718301

RESUMEN

Our earlier studies of preeclampsia (PE) at delivery have demonstrated the alteration of one carbon cycle, reduced placental omega 3 fatty acids, altered circulating levels of angiogenic factors and differential placental gene-specific methylation patterns of angiogenic factors. This study was undertaken to examine changes in the levels of angiogenic factors and angiotensin II type 1 receptor autoantibodies (AT1-AAs) throughout gestation, from early pregnancy until delivery, in women with PE and to examine their association with cord angiogenic factors, blood pressure and infant weight. A total of 81 pregnant women (46 normotensive and 35 with PE) were followed at three different time points during pregnancy: 16-20 weeks (T1), 26-30 weeks (T2) and at the time of delivery (T3). The plasma levels of angiogenic factors and AT1-AAs were determined in the maternal and cord plasma by commercial enzyme-linked immunosorbent assay kits. Maternal plasma levels of vascular endothelial growth factor (VEGF) and placental growth factor (PlGF) were lower (P<0.05 for both), whereas soluble fms-like tyrosine kinase-1 (sFlt-1; P<0.05) and the sFlt-1/PlGF ratio (P<0.01) were higher in early pregnancy in the PE group. Maternal plasma AT1-AA levels were higher (P<0.05) at T2 in women with PE. Cord plasma VEGF and soluble kinase insert domain receptor (sKDR) levels were lower (P<0.01 and P<0.05, respectively), whereas AT1-AA levels were higher (P<0.05) in the PE group. Maternal plasma VEGF levels in early pregnancy were positively associated with systolic blood pressure, whereas the sFlt-1/PlGF ratio at T2 was negatively associated with infant weight in the PE group. Low levels of proangiogenic factors (VEGF and PlGF) and high levels of AT1-AAs and antiangiogenic factors (sFlt-1 and sFlt-1/PlGF ratio) are present in the maternal circulation during early gestation in women with PE.


Asunto(s)
Proteínas Angiogénicas/metabolismo , Autoanticuerpos/análisis , Preeclampsia/sangre , Receptor de Angiotensina Tipo 1/inmunología , Adulto , Peso al Nacer , Presión Sanguínea/fisiología , Femenino , Desarrollo Fetal/genética , Desarrollo Fetal/fisiología , Humanos , Recién Nacido , Estudios Longitudinales , Factor de Crecimiento Placentario , Embarazo , Proteínas Gestacionales/sangre , Factor A de Crecimiento Endotelial Vascular/sangre , Receptor 1 de Factores de Crecimiento Endotelial Vascular/sangre , Receptor 2 de Factores de Crecimiento Endotelial Vascular/sangre
16.
Clin Obstet Gynecol ; 56(3): 529-36, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23751877

RESUMEN

A newly recognized primary cause of the obesity epidemic is the developmental programming effects of infants born to mothers with obesity or gestational diabetes, intrauterine growth-restricted newborns, and offspring exposed to environmental toxins including bisphenol A. The mechanisms which result in offspring obesity include the programming of the hypothalamic appetite pathway and adipogenic signals regulating lipogenesis. Processes include nutrient sensors, epigenetic modifications, and alterations in stem cell precursors of both appetite/satiety neurons and adipocytes which are modulated to potentiate offspring obesity. Future strategies for the prevention and therapy of obesity must address programming effects of the early life environment.


Asunto(s)
Adipogénesis/fisiología , Apetito/fisiología , Diabetes Gestacional/fisiopatología , Desarrollo Fetal/fisiología , Retardo del Crecimiento Fetal/fisiopatología , Obesidad/embriología , Efectos Tardíos de la Exposición Prenatal/fisiopatología , Adipogénesis/genética , Animales , Apetito/genética , Compuestos de Bencidrilo/efectos adversos , Diabetes Gestacional/genética , Epigénesis Genética , Estrógenos no Esteroides/efectos adversos , Femenino , Desarrollo Fetal/genética , Retardo del Crecimiento Fetal/genética , Humanos , Hipotálamo/fisiología , Hipotálamo/fisiopatología , Lipogénesis/genética , Lipogénesis/fisiología , Fenómenos Fisiologicos Nutricionales Maternos , Obesidad/genética , Fenoles/efectos adversos , Embarazo , Efectos Tardíos de la Exposición Prenatal/genética , Fenómenos Fisiologicos de la Nutrición Prenatal
17.
Am J Chin Med ; 41(3): 515-29, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23711139

RESUMEN

Toki-shakuyaku-san (TSS), an herbal formula based on traditional Chinese medicine, is commonly used in obstetrics. To examine the effects of TSS on the normal mouse fetus and placenta, TSS was administered to normal pregnant mice and their placentas and fetuses were studied. First, the effects of maternal TSS treatment on implantation were investigated. Administration of TSS from gestation day 0.5 (G0.5) to G6.5 showed that litter size was not altered at embryonic day 11.5 (E11.5), but the number of resorbed fetuses was slightly decreased. Then, to investigate effects on fetal and placental growths after implantation, TSS was administered from G5.5. At E14.5, the body weight of fetuses from TSS-treated dams was significantly increased. Gene expression of insulin-like growth factor 2 (Igf2), one of the most important modulators of fetal growth, was significantly increased in the placentas and fetuses of TSS-treated dams. In addition, the expression of particular placental developmental genes and nutrient transporter genes was significantly increased in TSS-treated placentas. At E18.5, after longer-term administration of TSS, fetal and placental weights were not altered, but the expression of the placental developmental and nutrient transporter genes remained elevated compared with controls. These results suggest that maternal TSS treatment in normal mice enhances the expression of Igf2, placental developmental genes and nutrient transporter genes, resulting in increased fetal weight. No obvious changes were observed in the expression of these genes after longer-term maternal TSS treatment.


Asunto(s)
Medicamentos Herbarios Chinos/farmacología , Desarrollo Fetal/efectos de los fármacos , Peso Fetal/efectos de los fármacos , Feto/efectos de los fármacos , Expresión Génica/efectos de los fármacos , Placenta/efectos de los fármacos , Placentación/efectos de los fármacos , Animales , Transporte Biológico/genética , Pérdida del Embrión , Femenino , Desarrollo Fetal/genética , Peso Fetal/genética , Factor II del Crecimiento Similar a la Insulina/genética , Factor II del Crecimiento Similar a la Insulina/metabolismo , Ratones , Ratones Endogámicos C57BL , Placentación/genética , Embarazo , Valores de Referencia
18.
Trends Endocrinol Metab ; 24(6): 279-89, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23474063

RESUMEN

Folate plays a key role in the interactions between nutrition, fetal programming, and epigenomics. Maternal folate status influences DNA methylation, inheritance of the agouti phenotype, expression of imprinting genes, and the effects of mycotoxin FB1 on heterochromatin assembly in rodent offspring. Deficiency in folate and other methyl donors increases birth defects and produces visceral manifestations of fetal programming, including liver and heart steatosis, through imbalanced methylation and acetylation of PGC1-α and decreased SIRT1 expression, and produces persistent cognitive and learning disabilities through impaired plasticity and hippocampal atrophy. Maternal folate supplementation also produces long-term epigenomic effects in offspring, some beneficial and others negative. Deciphering these mechanisms will help understanding the discordances between experimental models and population studies of folate deficiency and supplementation.


Asunto(s)
Desarrollo Fetal/fisiología , Ácido Fólico/metabolismo , Epigénesis Genética/genética , Femenino , Desarrollo Fetal/genética , Humanos , Modelos Biológicos , Coactivador 1-alfa del Receptor Activado por Proliferadores de Peroxisomas gamma , Sirtuina 1/genética , Sirtuina 1/metabolismo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
19.
Placenta ; 33(10): 830-8, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-22819041

RESUMEN

Distal-less 3 (Dlx3)(-/-) mice die at E9.5 presumably due to an abnormal placental phenotype including reduced placental vasculature and secretion of placental growth factor. To examine the role of Dlx3 specifically within the epiblast, Dlx3 conditional knockout mice were generated using an epiblast-specific Meox2(CreSor) allele. Dlx3(-/fl), Meox2(CreSor) animals were born at expected frequencies and survived to weaning providing indirect evidence that loss of Dlx3 within the trophoectoderm plays a critical role in fetal survival in the Dlx3(-/-) mouse. We next examined the hypothesis that loss of a single Dlx3 allele would have a negative impact on placental and fetal fitness. Dlx3(+/-) mice displayed reduced fetal growth beginning at E12.5 compared with Dlx3(+/+) controls. Altered fetal growth trajectory occurred coincident with elevated oxidative stress and apoptosis within Dlx3(+/-) placentas. Oral supplementation with the superoxide dismutase mimetic, Tempol, rescued the fetal growth and placental cell death phenotypes in Dlx3(+/-) mice. To determine the potential mechanisms associated with elevated oxidative stress on the Dlx3(+/-) placentas, we next examined vascular characteristics within the feto-placental unit. Studies revealed reduced maternal spiral artery luminal area in the Dlx3(+/-) mice receiving water; Dlx3(+/-) mice receiving Tempol displayed maternal spiral artery luminal area similar to control Dlx3(+/+) mice. We conclude that reduced Dlx3 gene dose results in diminished fetal fitness associated with elevated placental cell oxidative stress and apoptosis coincident with altered vascular remodeling. Administration of antioxidant therapy ameliorated this feto-placental phenotype, suggesting that Dlx3 may be required for adaptation to oxidative stresses within the intrauterine environment.


Asunto(s)
Haploinsuficiencia/fisiología , Proteínas de Homeodominio/genética , Estrés Oxidativo/genética , Placenta/metabolismo , Factores de Transcripción/genética , Animales , Antioxidantes/uso terapéutico , Apoptosis/genética , Óxidos N-Cíclicos/uso terapéutico , Femenino , Desarrollo Fetal/efectos de los fármacos , Desarrollo Fetal/genética , Masculino , Ratones , Ratones Noqueados , Embarazo , Marcadores de Spin , Superóxido Dismutasa/genética
20.
Bull Acad Natl Med ; 196(9): 1829-42, 2012 Dec.
Artículo en Francés | MEDLINE | ID: mdl-24552105

RESUMEN

Folates are needed for synthesis of methionine, the precursor of S-adenosyl methionine (SAM). They play therefore a key role in nutrition and epigenomics by fluxing monocarbons towards synthesis or methylation of DNA and RNA, and methylation of gene transregulators, respectively. The deficiency produces intrauterine growth retardation and birth dejects. Folate deficiency deregulates epigenomic mechanisms related to fetal programming through decreased cellular availability of SAM. Epigenetic mechanisms of folate deficiency are illustrated by inheritance of coat colour of agouti mice model and altered expression of Igf2/H19 imprinting genes. Dietary exposure to fumonisin FB1 acts synergistically with folate deficiency on alterations of heterochromatin assembly. Deficiency in folate and vitamin B12 produces impaired fatty acid oxidation in liver and heart through imbalanced methylation and acetylation of PGC1-alpha and decreased expression of SIRT1, and long-lasting cognitive disabilities through impaired hippocampal cell proliferation, differentiation and plasticity and atrophy of hippocampal CA1. Deciphering these mechanisms will help understand the discordances between experimental models and population studies on folate supplementation.


Asunto(s)
Epigénesis Genética/fisiología , Desarrollo Fetal/genética , Deficiencia de Ácido Fólico/genética , Animales , Epigenómica , Femenino , Deficiencia de Ácido Fólico/complicaciones , Humanos , Metilenotetrahidrofolato Reductasa (NADPH2)/genética , Ratones , Estado Nutricional/genética , Embarazo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA