Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 74
Filtrar
Mais filtros

País/Região como assunto
Tipo de documento
País de afiliação
Intervalo de ano de publicação
1.
J Cell Physiol ; 2024 Mar 11.
Artigo em Inglês | MEDLINE | ID: mdl-38465708

RESUMO

Maternal obesity (MO) is a significant cause of increased cardiometabolic risk in offspring, who present endothelial dysfunction at birth. Alterations in physiologic and cellular redox status are strongly associated with altered gene regulation in arterial endothelium. However, specific mechanisms by which the pro-oxidant fetal environment in MO could modulate the vascular gene expression and function during the offspring's postnatal life are elusive. We tested if oxidative stress could reprogram the antioxidant-coding gene's response to a pro-oxidant challenge through an epigenetic transcriptional memory (ETM) mechanism. A pro-oxidant double-hit protocol was applied to human umbilical artery endothelial cells (HUAECs) and EA.hy 926 endothelial cell lines. The ETM acquisition in the HMOX1 gene was analyzed by RT-qPCR. HMOX1 mRNA decay was evaluated by Actinomycin-D treatment and RT-qPCR. To assess the chromatin accessibility and the enrichment of NRF2, RNAP2, and phosphorylation at serin-5 of RNAP2, at HMOX1 gene regulatory regions, were used DNase HS-qPCR and ChIP-qPCR assays, respectively. The CpG methylation pattern at the HMOX1 core promoter was analyzed by DNA bisulfite conversion and Sanger sequencing. Data were analyzed using two-way ANOVA, and p < 0.05 was statistically significant. Using a pro-oxidant double-hit protocol, we found that the Heme Oxygenase gene (HMOX1) presents an ETM response associated with changes in the chromatin structure at the promoter and gene regulatory regions. The ETM response was characterized by a paused-RNA Polymerase 2 and NRF2 enrichment at the transcription start site and Enhancer 2 of the HMOX1 gene, respectively. Changes in DNA methylation pattern at the HMOX1 promoter were not a hallmark of this oxidative stress-induced ETM. These data suggest that a pro-oxidant milieu could trigger an ETM at the vascular level, indicating a potential epigenetic mechanism involved in the increased cardiovascular risk in the offspring of women with obesity.

2.
Artigo em Inglês | MEDLINE | ID: mdl-38922906

RESUMO

BACKGROUND: Previous studies of maternal docosahexaenoic acid (DHA) supplementation during pregnancy have controversial and contrasting results on the short and long-term effects on early child growth. The impact of this nutritional intervention on the postnatal growth patterns in the offspring of women with pregestational overweight/obesity (PGO) also remains controversial. OBJECTIVE: To analyze the postnatal growth patterns during the first 4 months of life in the offspring of women with PGO randomly supplemented with 800 mg/day (PGO-800) compared with normative doses of 200 mg/day (PGO-200) of DHA during pregnancy (<15 weeks of gestation until delivery). METHODS: This study evaluated the growth patterns during the first 4 months of life of 169 infants of the women that participated in the MIGHT study (NCT02574767). We included the infants of women from the PGO-200 (n = 81) and PGO-800 group (n = 88). The growth patterns (weight, length, and head circumference) and change in z-score (WHO charts) were evaluated. RESULTS: Throughout the first 4 months of life, the infants of the PGO-800 group had lower weight-for-length z-score (coef. -0.65, 95% confidence interval [CI] -1.07, -0.22, p = 0.003) and lower body mass index-for-age z-score (coef. -0.56, 95% CI -0.99, -0.12, p = 0.012) compared with the PGO-200 group adjusted by maternal body mass index, gestational weight gain, gestational age, insulin in cord blood and infant feeding (exclusive breastfed, not breastfed, and partially breastfed). CONCLUSIONS: Maternal supplementation with DHA during pregnancy could beneficially limit the offspring's postnatal weight gain during the first 4 months of life.

3.
Adv Exp Med Biol ; 1428: 83-98, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37466770

RESUMO

Fetal growth and development are influenced by maternal nutrition and gestational weight gain. Adequate intake of nutrients such as folate, vitamin B12, and docosahexaenoic acid (DHA) is essential for healthy fetal and placental development. Many countries have a national flour fortification program with folic acid (FA), together with pre-pregnancy supplementation of FA (400 µg/day) during the first trimester of pregnancy. The latter has been recommended by the WHO and adapted to local requirements by perinatal guidelines. On the other hand, in population studies, many women of childbearing age have vitamin B12 deficiency (<148 pmol/L), which can be additionally masked by high FA intake and maternal pregestational obesity. Under these conditions, these patients could be having pregnancies in a folate/vitamin B12 imbalance, which is associated with higher adiposity, insulin resistance, altered lipid metabolism, and low DHA levels in their offspring. However, if these neonatal consequences of maternal pregestational obesity and folate/vitamin B12 imbalance can be reverted by DHA supplementation during pregnancy has not been addressed. This chapter reviews the literature and exposes the current gaps in knowledge and challenges in maternal nutrition with a life-course perspective.


Assuntos
Ácido Fólico , Vitamina B 12 , Recém-Nascido , Feminino , Humanos , Gravidez , Placenta , Obesidade , Ácidos Graxos Insaturados
4.
Int J Obes (Lond) ; 46(5): 977-985, 2022 05.
Artigo em Inglês | MEDLINE | ID: mdl-35058573

RESUMO

BACKGROUND: The relationship between eating behaviour and current body weight has been described. However little is known about the effect of polyunsaturated fatty acids (PUFA) in this relationship. Genetic contribution to a certain condition is derived from a combination of small effects from many genetic variants, and polygenic risk scores (PRS) summarize these effects. A PRS based on a GWAS for plasma docosahexaenoic fatty acid (DHA) has been created, based on SNPs from 9 genes. OBJECTIVE: To analyze the interaction between the PRS for plasma DHA concentration, body composition and eating behaviour (using the Children Eating Behaviour Questionnaire) in childhood. SUBJECTS/METHODS: We analyzed a subsample of children from the Maternal, Adversity, Vulnerability and Neurodevelopment (MAVAN) cohort with PRS and measurements of eating behaviour performed at 4 years of age (n = 210), 6 y (n = 177), and body fat determined by bioelectric impedance at 4 y and 6 y or by air displacement plethysmography and dual-energy X-ray absorptiometry at 8 y (n = 42 and n = 37). PRS was based on the GWAS from Lemaitre et al. 2011 (p threshold = p < 5*10-6), and a median split created low and high PRS groups (high PRS = higher DHA level). RESULTS: In ALSPAC children, we observed an association between PRS and plasma DHA concentration (ß = 0.100, p < 0.01) and proportion (ß = 0.107, p < 0.01). In MAVAN, there were interactions between PRS and body fat on pro-intake scores in childhood, in which low PRS and higher body fat were linked to altered behaviour. There were also interactions between PRS and pro-intake scores early in childhood on body fat later in childhood, suggesting that the genetic profile and eating behaviour influence the development of adiposity at later ages. CONCLUSIONS: A lower PRS (lower plasma PUFA) can be a risk factor for developing higher body fat associated with non-adaptive eating behaviour in childhood; it is possible that the higher PRS (higher plasma PUFA) is a protective feature.


Assuntos
Composição Corporal , Ácidos Graxos , Absorciometria de Fóton , Composição Corporal/genética , Criança , Ácidos Docosa-Hexaenoicos , Ácidos Graxos Insaturados , Comportamento Alimentar , Humanos , Fatores de Risco
5.
Pediatr Res ; 2022 Nov 23.
Artigo em Inglês | MEDLINE | ID: mdl-36418485

RESUMO

BACKGROUND: Severe pulmonary hypoplasia related to congenital diaphragmatic hernia (CDH) continues to be a potentially fatal condition despite advanced postnatal management strategies. OBJECTIVE: To evaluate the effect of the antenatal sildenafil and 2(S)-amino-6-boronohexanoic acid (ABH-Arginase inhibitor) on lung volume, pulmonary vascular development, and nitric oxide (NO) synthesis in a Nitrofen-induced CDH rat model. METHODS: Nitrofen-induced CDH rat model was used. Nitrofen was administrated on embryonic day(E) 9,5. At E14, five intervention groups were treated separately: Nitrofen, Nitrofen+Sildenafil, Nitrofen+ABH, Nitrofen+Sildenafil+ABH and Control. At term, offspring's lungs were weighed, some paraffin-embedded for histology, others snap-frozen to analyze eNOS, Arginase I-II expression, and activity. RESULTS: In CDH-bearing offsprings, ABH or Sildenafil+ABH preserved the total lung/body-weight index (p < 0.001), preventing pulmonary vascular smooth muscle cell hyperproliferation and improving lung morphometry. Sildenafil+ABH increased 1.7-fold the lung nitrite levels (p < 0.01) without changes in eNOS expression. Sildenafil and ABH improved the number of pulmonary vessels. CONCLUSION: These results suggest that in this CDH rat model, the basal activity of Arginase participates in the lung volume and, together with phosphodiesterase-5, regulates NOS activity in the term fetal lung. The combined treatment (Sildenafil+ABH) could revert some of the pulmonary features in CDH by improving the local NO synthesis and preventing smooth muscle cell hyperproliferation. IMPACT: This study presents Arginase inhibition as a new therapeutic target and the importance of the combined antenatal treatment to improve pulmonary vascular development in a congenital diaphragmatic hernia (CDH) rat model. This study shows that the action of an Arginase inhibitor (ABH) enhances the effects already described for sildenafil in this model. These results reinforce the importance of prenatal treatments' synergy in recovering the hypoplastic lung in the Nitrofen-induced CDH rat model.

6.
Am J Obstet Gynecol ; 224(5): 526.e1-526.e25, 2021 05.
Artigo em Inglês | MEDLINE | ID: mdl-33152314

RESUMO

BACKGROUND: Lifestyle interventions have shown limited effectiveness in the prevention of gestational diabetes mellitus. The combination of lifestyle interventions with omega-3 polyunsaturated fatty acid supplementation could have a synergetic effect on maternal and offspring outcomes. OBJECTIVE: We evaluated the effects of docosahexaenoic acid supplementation among obese and overweight pregnant women (independently or combined with a dietary counseling intervention) on metabolic control in mothers and their offspring. STUDY DESIGN: This study was a randomized controlled trial with a 2×2 factorial design. The following inclusion criteria were used: <15 weeks of gestation; body mass index ≥25 kg/m2 at the first prenatal visit; singleton pregnancy; and 18 years of age or older. The recruited women (n=1002) were randomly allocated to 1 of the 4 parallel groups: Group 1: dietary counseling plus 800 mg/day of docosahexaenoic acid (n=250); Group 2: routine counseling plus 800 mg/day docosahexaenoic acid (n=252); Group 3: dietary counseling plus 200 mg/day docosahexaenoic acid (n=249); and Group 4: routine counseling plus 200 mg/day docosahexaenoic acid (n=251), considered as the reference group. The dietary intervention comprised 3 sessions, and it was focused on reducing the consumption of foods that most contributed to daily sugar intake. Primary outcomes were gestational diabetes mellitus defined according to the national guidelines; macrosomia (birthweight >4000 g); and neonatal insulin resistance (cord blood Homeostasis Model Assessment for Insulin Resistance ≥2.60), which was assessed in a subsample of 226 newborns. The analysis was by intention to treat and by efficacy. The trial was registered on ClinicalTrials.gov (NCT02574767). RESULTS: The overall incidence of gestational diabetes mellitus was 20.2% (Group 1, 21.0%; Group 2, 20.1%; Group 3, 18.9%; and Group 4, 20.9%). Mean birthweight was 3403.0 g (standard deviation, 575.3), and the incidence of macrosomia was 11.9% (Group 1, 13.2%; Group 2, 10.8%; Group 3, 11.5%; and Group 4, 12.1%). Median cord blood Homeostasis Model Assessment for Insulin Resistance was 0.9 (interquartile range, 0.6-1.7), and 10.2% showed cord blood insulin resistance (Group 1, 12.0%; Group 2, 12.0%; Group 3, 9.7%; and Group 4, 5.1%). No significant differences were found among groups regarding primary outcomes (P<.05). Glucose concentrations in the cord blood samples were lower in those adherents to the docosahexaenoic acid supplementation (P<.05). CONCLUSION: For women who were overweight or obese at the beginning of pregnancy, this combined intervention did not reduce the risk of gestational diabetes in mothers or macrosomia and insulin resistance in neonates.


Assuntos
Diabetes Gestacional/prevenção & controle , Aconselhamento Diretivo , Ácidos Docosa-Hexaenoicos/uso terapêutico , Macrossomia Fetal/prevenção & controle , Obesidade/terapia , Adulto , Peso ao Nascer , Glicemia/metabolismo , Terapia Combinada , Açúcares da Dieta , Suplementos Nutricionais , Feminino , Sangue Fetal/metabolismo , Humanos , Recém-Nascido , Resistência à Insulina , Obesidade/metabolismo , Sobrepeso/metabolismo , Sobrepeso/terapia , Gravidez , Adulto Jovem
7.
J Pediatr Gastroenterol Nutr ; 73(2): 264-270, 2021 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-34016877

RESUMO

BACKGROUND: Deleterious long-term effects in the offspring from women with pregravid obesity have been described; however, the evidence supporting early metabolic and inflammatory markers in the offspring at birth and gender differences are conflicting. OBJECTIVE: The present study aimed to compare cord blood adipokines and cytokines concentrations and anthropometric characteristics of the offspring of women with maternal obesity (MO) and normal-weight mothers (NWM). Also, maternal and neonatal variables on the association of maternal body mass index (BMI) with cord blood adipokines were evaluated. METHODS: A cross-sectional analysis of a subsample of mother-child dyads participating in a cohort study (n = 221) was assessed. Anthropometrics, cord blood adipokines (leptin and adiponectin) and cytokines (interleukin [IL]-1ß, IL-4, IL-10, IL-12 p40, IL-12p70, IL-13, and tumor necrosis factor α) concentrations in the offspring of normal-weight women (BMI >18.5 and <24.9 kg/m2) and women with pregravid obesity (BMI > 30 kg/m2) without comorbidities was performed. RESULTS: Offspring from mothers with obesity had higher birth weight, a higher proportion of large for gestational age, higher ponderal index, and heavier placentae than offspring from normal-weight mothers (P < 0.05). Within the offspring from women with obesity, males had significantly higher weight, length, the proportion of large-for-gestational-age newborns, higher weight for length ratio. Males had more efficient placentas than females (P < 0.05). Higher adiponectin and leptin in both sexes and higher leptin in female offspring of mothers with obesity after adjusting for birth size (P < 0.05) were found. Higher IL-12p40 in the offspring of women with MO with no other differences in other cytokines among groups were evidenced. CONCLUSIONS: Maternal obesity associates with a higher concentration of adiponectin and leptin in their offspring at birth. There is a relevant effect on anthropometrics in male offspring and on leptin in female newborn. Further studies need to evaluate the extension of these effects in postnatal life. TRAIL IDENTIFICATION NUMBER: NCT02903134.


Assuntos
Adipocinas , Obesidade Materna , Estudos de Coortes , Estudos Transversais , Feminino , Sangue Fetal , Humanos , Masculino , Gravidez
8.
BMC Pediatr ; 20(1): 164, 2020 04 15.
Artigo em Inglês | MEDLINE | ID: mdl-32293348

RESUMO

BACKGROUND: Growing evidence shows that atopic dermatitis (AD), food allergy (FA), allergic rhinitis, and asthma are largely determined during the first 1000 days (time elapsed from conception to the 2nd birthday). The ARIES birth cohort aims to determine prenatal and perinatal conditions, as well as genetic and epigenetic factors, that participate in the early setting of immune responses, and the role of these in the later determination of the risk of allergic diseases and asthma in the offspring. METHODS: We have designed a birth cohort of 250 families with prenatal recruitment (~ 14 weeks). We will genotype relevant allergy/asthma-associated variants in trios and will perform immunophenotyping and evaluation of allergy biomarkers in cord blood. At 1 and 2 years of age we will assess if infants have developed allergic sensitization, AD, FA, as well as biomarkers of asthma including the asthma predictive index. We will also evaluate how maternal conditions modify immune programming through epigenetic modifications and will then depict newborn epigenetic cues of allergy/asthma risk. Next, we will assess composition/diversity of maternal gut, placenta, breastmilk and infant gut microbiome and their association with immunophenotype and biomarkers at birth, and clinical outcomes at age 1 and 2. Finally, we plan to assess how environmental exposures (perinatal outdoor and indoor pollution, allergens and endotoxin) affect the incidence of allergic sensitization, AD, FA, and risk of asthma. DISCUSSION: The in-depth study of the ARIES birth cohort shall provide crucial information to understand the rising incidence of allergies and asthma in developing countries, and hopefully provide cues on how to prevent and treat these diseases. TRIAL REGISTRATION: clinicaltrials.gov NCT04186949, retrospectively registered on December 5, 2019.


Assuntos
Asma/epidemiologia , Dermatite Atópica/epidemiologia , Hipersensibilidade Alimentar/epidemiologia , Rinite Alérgica/epidemiologia , Asma/etiologia , Chile/epidemiologia , Dermatite Atópica/etiologia , Feminino , Hipersensibilidade Alimentar/etiologia , Humanos , Lactente , Recém-Nascido , Gravidez , Estudos Prospectivos , Rinite Alérgica/etiologia
9.
Int J Mol Sci ; 21(13)2020 Jun 30.
Artigo em Inglês | MEDLINE | ID: mdl-32630161

RESUMO

Preeclampsia is a pregnancy complication that appears after 20 weeks of gestation and is characterized by hypertension and proteinuria, affecting both mother and offspring. The cellular and molecular mechanisms that cause the development of preeclampsia are poorly understood. An important feature of preeclampsia is an increase in oxygen and nitrogen derived free radicals (reactive oxygen species/reactive nitrogen species (ROS/RNS), which seem to be central players setting the development and progression of preeclampsia. Cell-to-cell communication may be disrupted as well. Connexins (Cxs), a family of transmembrane proteins that form hemichannels and gap junction channels (GJCs), are essential in paracrine and autocrine cell communication, allowing the movement of signaling molecules between cells as well as between the cytoplasm and the extracellular media. GJCs and hemichannels are fundamental for communication between endothelial and smooth muscle cells and, therefore, in the control of vascular contraction and relaxation. In systemic vasculature, the activity of GJCs and hemichannels is modulated by ROS and RNS. Cxs participate in the development of the placenta and are expressed in placental vasculature. However, it is unknown whether Cxs are modulated by ROS/RNS in the placenta, or whether this potential modulation contributes to the pathogenesis of preeclampsia. Our review addresses the possible role of Cxs in preeclampsia, and the plausible modulation of Cxs-formed channels by ROS and RNS. We suggest these factors may contribute to the development of preeclampsia.


Assuntos
Conexinas/metabolismo , Pré-Eclâmpsia/etiologia , Espécies Reativas de Nitrogênio/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Animais , Vasos Sanguíneos/metabolismo , Feminino , Humanos , Inflamação/metabolismo , Placenta/metabolismo , Pré-Eclâmpsia/metabolismo , Gravidez
10.
J Cell Physiol ; 234(10): 18571-18586, 2019 08.
Artigo em Inglês | MEDLINE | ID: mdl-30937903

RESUMO

Obesity is a public health problem worldwide, and especially in women in reproductive age where more than one in three have obesity. Maternal obesity is associated with an increased maternal, placental, and newborn oxidative stress, which has been proposed as a central factor in vascular dysfunction in large-for-gestational-age (LGA) newborn. However, cellular and molecular mechanisms behind this effect have not been elucidated. Untreated human umbilical artery endothelial cells (HUAEC) from LGA (LGA-HUAEC) presented higher O2- levels, superoxide dismutase activity and heme oxygenase 1 messenger RNA (mRNA) levels, paralleled by reduced GSH:GSSG ratio and NRF2 mRNA levels. In response to an oxidative challenge (hydrogen peroxide), only HUAEC from LGA exhibited an enhanced Glutathione Peroxidase 1 (GPX1) expression, as well as a more efficient antioxidant machinery measured by the biosensor probe, HyPer. An open state of chromatin in the TSS region of GPX1 in LGA-HUAEC was evidenced by the DNase-HS assay. Altogether, our data indicate that LGA-HUAEC have an altered cellular and molecular antioxidant system. We propose that a chronic pro-oxidant intrauterine milieu, as evidenced in pregestational obesity, could induce a more efficient antioxidant system in fetal vascular cells, which could be maintained by epigenetic mechanism during postnatal life.


Assuntos
Antioxidantes/metabolismo , Células Endoteliais/citologia , Regulação da Expressão Gênica , Idade Gestacional , Artérias Umbilicais/citologia , Cromatina/metabolismo , Células Endoteliais/enzimologia , Feminino , Fluorescência , Dissulfeto de Glutationa/metabolismo , Humanos , Peróxido de Hidrogênio/metabolismo , Recém-Nascido , Cinética , Modelos Biológicos , Obesidade/patologia , Oxirredução , Gravidez , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Superóxido Dismutase/metabolismo , Superóxidos/metabolismo , Sítio de Iniciação de Transcrição
11.
J Cell Physiol ; 233(10): 6723-6733, 2018 10.
Artigo em Inglês | MEDLINE | ID: mdl-29377113

RESUMO

Maternal obesity is associated with large-for-gestational-age (LGA) neonates and programming of obesity-related cardiovascular disease in the offspring, however, the mechanisms that lead to the later are unclear. Presently, interpretations of NO-dependent changes in vascular function in LGA newborn from obese mothers are conflicting. Adiponectin improves endothelial function by increasing eNOS activity and NO production. We propose that LGAs from obese mothers present a diminished vascular response to adiponectin; thus, affecting eNOS and AMPK activation. Chorionic arteries, umbilical cord and primary cultures of umbilical artery endothelial cells (HUAEC) were collected at term (>38 weeks) from uncomplicated singleton pregnancies of LGA and adequate-for-gestational (AGA) newborn. Vascular reactivity of chorionic plate arteries was assessed by wire myography. mRNA expression of adiponectin receptors 1 (AdipoR1) and AdipoR2 in HUAEC was determined by qPCR. Protein expression of AdipoR1, AdipoR2, AMPK, phospho-AMPKαThr172 , eNOS, and phospho-eNOSSer1177 after stimulation with AdipoRon was determined by Western Blot. Maximal adiponectin-induced chorionic artery relaxation in LGAs was diminished compared to control. In vitro studies showed no differences in expression of AdipoRs, total AMPK and, eNOS activation between groups; however, higher expression of total eNOS and AMPK activation in HUAEC of LGA relative to AGAs were observed. LGA HUAEC showed diminished NO production and eNOS activity compared to AGA in response to AdipoRon but no changes in AMPK activation. Placental endothelium of LGAs shows a diminished vascular response to adiponectin. Moreover, eNOS activation and adiponectin-dependent NO production is lower in HUAEC of LGA from obese mothers, indicating they present dysfuncional placental-endothelial responses.


Assuntos
Adiponectina/genética , Endotélio Vascular/fisiopatologia , Obesidade/genética , Complicações Cardiovasculares na Gravidez/genética , Quinases Proteína-Quinases Ativadas por AMP , Artérias/metabolismo , Artérias/fisiopatologia , Células Endoteliais/patologia , Endotélio Vascular/diagnóstico por imagem , Endotélio Vascular/metabolismo , Feminino , Regulação da Expressão Gênica/genética , Humanos , Recém-Nascido , Masculino , Miografia , Óxido Nítrico/genética , Óxido Nítrico/metabolismo , Óxido Nítrico Sintase Tipo III/genética , Obesidade/diagnóstico por imagem , Obesidade/metabolismo , Obesidade/fisiopatologia , Placenta/metabolismo , Placenta/fisiopatologia , Circulação Placentária/fisiologia , Gravidez , Complicações Cardiovasculares na Gravidez/metabolismo , Complicações Cardiovasculares na Gravidez/fisiopatologia , Proteínas Quinases/genética , Receptores de Adiponectina/genética , Vasodilatação/genética
12.
J Physiol ; 595(4): 1077-1092, 2017 02 15.
Artigo em Inglês | MEDLINE | ID: mdl-27739590

RESUMO

KEY POINTS: Intrauterine growth restriction (IUGR) is associated with vascular dysfunction, oxidative stress and signs of endothelial epigenetic programming of the umbilical vessels. There is no evidence that this epigenetic programming is occurring on systemic fetal arteries. In IUGR guinea pigs we studied the functional and epigenetic programming of endothelial nitric oxide synthase (eNOS) (Nos3 gene) in umbilical and systemic fetal arteries, addressing the role of oxidative stress in this process by maternal treatment with N-acetylcysteine (NAC) during the second half of gestation. The present study suggests that IUGR endothelial cells have common molecular markers of programming in umbilical and systemic arteries. Notably, maternal treatment with NAC restores fetal growth by increasing placental efficiency and reverting the functional and epigenetic programming of eNOS in arterial endothelium in IUGR guinea pigs. ABSTRACT: In humans, intrauterine growth restriction (IUGR) is associated with vascular dysfunction, oxidative stress and signs of endothelial programming in umbilical vessels. We aimed to determine the effects of maternal antioxidant treatment with N-acetylcysteine (NAC) on fetal endothelial function and endothelial nitric oxide synthase (eNOS) programming in IUGR guinea pigs. IUGR was induced by implanting ameroid constrictors on uterine arteries of pregnant guinea pigs at mid gestation, half of the sows receiving NAC in the drinking water (from day 34 until term). Fetal biometry and placental vascular resistance were followed by ultrasound throughout gestation. At term, umbilical arteries and fetal aortae were isolated to assess endothelial function by wire-myography. Primary cultures of endothelial cells (ECs) from fetal aorta, femoral and umbilical arteries were used to determine eNOS mRNA levels by quantitative PCR and analyse DNA methylation in the Nos3 promoter by pyrosequencing. Doppler ultrasound measurements showed that NAC reduced placental vascular resistance in IUGR (P < 0.05) and recovered fetal weight (P < 0.05), increasing fetal-to-placental ratio at term (∼40%) (P < 0.001). In IUGR, NAC treatment restored eNOS-dependent relaxation in aorta and umbilical arteries (P < 0.05), normalizing eNOS mRNA levels in EC fetal and umbilical arteries (P < 0.05). IUGR-derived ECs had a decreased DNA methylation (∼30%) at CpG -170 (from the transcription start site) and this epigenetic signature was absent in NAC-treated fetuses (P < 0.001). These data show that IUGR-ECs have common molecular markers of eNOS programming in umbilical and systemic arteries and this effect is prevented by maternal treatment with antioxidants.


Assuntos
Acetilcisteína/farmacologia , Antioxidantes/farmacologia , Reprogramação Celular , Células Endoteliais/metabolismo , Epigênese Genética , Retardo do Crescimento Fetal/metabolismo , Acetilcisteína/uso terapêutico , Animais , Antioxidantes/uso terapêutico , Células Cultivadas , Metilação de DNA , Células Endoteliais/citologia , Células Endoteliais/efeitos dos fármacos , Feminino , Retardo do Crescimento Fetal/tratamento farmacológico , Cobaias , Óxido Nítrico Sintase Tipo III/genética , Óxido Nítrico Sintase Tipo III/metabolismo , Regiões Promotoras Genéticas , Artérias Umbilicais/efeitos dos fármacos , Artérias Umbilicais/metabolismo , Artérias Umbilicais/patologia
13.
J Cell Physiol ; 232(12): 3693-3701, 2017 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-28160500

RESUMO

Obese women offspring have a higher risk of developing chronic diseases associated with an altered immune function. We aim to determine, in neonatal monocyte-derived macrophages, whether maternal obesity is associated with an altered expression and DNA methylation of pro- and anti-inflammatory genes, along with a higher pro-inflammatory response. Cord blood from newborns of obese (Ob) and lean (control) women were obtained at delivery. Monocytes were isolated and differentiated into macrophages, in which M1 (LPS/IFNγ) and M2 (IL-4) polarization were assayed. The mRNA levels for TNFα, IL-1ß, IL-12A, IL-12B, IL-10, and IL-4R were quantified by qPCR and the DNA methylation of candidate genes determined by pyrosequencing. RESULTS: Ob-monocytes had decreased levels of mRNA for pro-inflammatory cytokines IL-1ß, IL-10, and IL-12B compared with controls. Conversely, Ob-macrophages showed increased levels of mRNA for TNFα, IL-4R, and IL-10 compared with controls. M1 response was comparable between both groups, characterized by an important induction of TNFα and IL-1ß. In response to an M2 stimulus, control macrophages showed a decreased expression of inflammatory mediators while Ob-macrophages had an additional suppression of the anti-inflammatory mediator IL-10. Changes in IL-1ß (monocytes) and IL-10 (macrophages) in Ob-monocytes were paralleled by changes in their promoter DNA methylation in fetal monocytes. These results suggest that monocyte-derived macrophages from obese newborns show a basal anti-inflammatory phenotype with an unbalanced response to M1 and M2 polarization stimuli. The presence of changes in DNA methylation of key inflammatory genes in neonatal monocytes suggests an intrauterine programing of immune function by maternal obesity.


Assuntos
Mediadores da Inflamação/metabolismo , Interferon gama/farmacologia , Interleucina-10/metabolismo , Interleucina-4/farmacologia , Lipopolissacarídeos/farmacologia , Macrófagos/efeitos dos fármacos , Fenômenos Fisiológicos da Nutrição Materna , Obesidade/metabolismo , Adulto , Estudos de Casos e Controles , Células Cultivadas , Ilhas de CpG , Metilação de DNA , Regulação para Baixo , Epigênese Genética , Feminino , Sangue Fetal/citologia , Humanos , Recém-Nascido , Interleucina-10/genética , Leptina/sangue , Macrófagos/metabolismo , Obesidade/sangue , Obesidade/genética , Fenótipo , Gravidez , Regiões Promotoras Genéticas , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Adulto Jovem
14.
J Physiol ; 594(6): 1553-61, 2016 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-26719023

RESUMO

Intra-uterine growth restriction (IUGR) is associated with short and long-term metabolic and cardiovascular alterations. Mice and rats have been extensively used to study the effects of IUGR, but there are notable differences in fetal and placental physiology relative to those of humans that argue for alternative animal models. This study proposes that gradual occlusion of uterine arteries from mid-gestation in pregnant guinea pigs produces a novel model to better assess human IUGR. Fetal biometry and in vivo placental vascular function were followed by sonography and Doppler of control pregnant guinea pigs and sows submitted to surgical placement of ameroid constrictors in both uterine arteries (IUGR) at mid-gestation (35 days). The ameroid constrictors induced a reduction in the fetal abdominal circumference growth rate (0.205 cm day(-1) ) compared to control (0.241 cm day(-1) , P < 0.001) without affecting biparietal diameter growth. Umbilical artery pulsatility and resistance indexes at 10 and 20 days after surgery were significantly higher in IUGR animals than controls (P < 0.01). These effects were associated with a decrease in the relative luminal area of placental chorionic arteries (21.3 ± 2.2% vs. 33.2 ± 2.7%, P < 0.01) in IUGR sows at near term. Uterine artery intervention reduced fetal (∼30%), placental (∼20%) and liver (∼50%) weights (P < 0.05), with an increased brain to liver ratio (P < 0.001) relative to the control group. These data demonstrate that the ameroid constrictor implantations in uterine arteries in pregnant guinea pigs lead to placental vascular dysfunction and altered fetal growth that induces asymmetric IUGR.


Assuntos
Retardo do Crescimento Fetal/fisiopatologia , Circulação Placentária , Embolização da Artéria Uterina/métodos , Artéria Uterina/cirurgia , Animais , Modelos Animais de Doenças , Feminino , Retardo do Crescimento Fetal/etiologia , Cobaias , Gravidez , Artéria Uterina/patologia , Embolização da Artéria Uterina/efeitos adversos
15.
Rev Chil Pediatr ; 87(2): 88-95, 2016.
Artigo em Espanhol | MEDLINE | ID: mdl-27055949

RESUMO

Allergic diseases and asthma are the result of complex interactions between genetic predisposition and environmental factors. Asthma is one of the most prevalent chronic disease among children. In this article we review some environmental factors like: allergen exposition, tobacco, bacteria, microbial components, diet, obesity and stress, which influences during intrauterine and infancy life in the epigenetic regulation of asthma and allergic diseases. The review has been done in three models: in-vitro, animal and human.


Assuntos
Asma/etiologia , Epigênese Genética , Hipersensibilidade/etiologia , Animais , Asma/genética , Criança , Meio Ambiente , Predisposição Genética para Doença , Humanos , Hipersensibilidade/genética , Fatores de Risco
16.
Rev Chil Pediatr ; 87(1): 4-10, 2016.
Artigo em Espanhol | MEDLINE | ID: mdl-26872716

RESUMO

Current evidence supports the notion that alterations in intrauterine growth and during the first years of life have a substantial effect on the risk for the development of chronic disease, which in some cases is even higher than those due to genetic factors. The persistence and reproducibility of the phenotypes associated with altered early development suggest the participation of mechanisms that would record environmental cues, generating a cellular reprogramming (i.e., epigenetic mechanisms). This review is an introduction to a series of five articles focused on the participation of epigenetic mechanisms in the development of highly prevalent chronic diseases (i.e., cardiovascular, metabolic, asthma/allergies and cancer) and their origins in the foetal and neonatal period. This series of articles aims to show the state of the art in this research area and present the upcoming clues and challenges, in which paediatricians have a prominent role, developing strategies for the prevention, early detection and follow-up.


Assuntos
Epigênese Genética/genética , Desenvolvimento Fetal/genética , Pediatras/organização & administração , Doença Crônica , Feminino , Predisposição Genética para Doença , Humanos , Lactente , Recém-Nascido , Papel do Médico , Gravidez , Reprodutibilidade dos Testes
17.
Rev Chil Pediatr ; 87(3): 154-61, 2016.
Artigo em Espanhol | MEDLINE | ID: mdl-27209119

RESUMO

Intrauterine growth restriction (IUGR) is a perinatal condition affecting foetal growth, with under the 10th percentile of the weight curve expected for gestational age. This condition has been associated with higher cardiovascular and metabolic risk and post-natal obesity. There are also major changes in placental function, and particularly in a key molecule in this regulation, nitric oxide. The synthesis of nitric oxide has numerous control mechanisms and competition with arginase for their common substrate, the amino acid L-arginine. This competition is reflected in various vascular diseases and particularly in the endothelium of the umbilical vessels of babies with IUGR. Along with this, there is regulation at the epigenetic level, where methylation in specific regions of some gene promoters, such as the nitric oxide synthase, regulating their expression. It is currently of great interest to understand the mechanisms by which diseases such as IUGR may be conditioned, particularly by maternal nutritional and metabolic conditions, and epigenetic mechanisms that could eventually be modifiable, and thus a focus of interest for health interventions.


Assuntos
Epigênese Genética , Retardo do Crescimento Fetal/genética , Placenta/metabolismo , Feminino , Retardo do Crescimento Fetal/fisiopatologia , Humanos , Óxido Nítrico/metabolismo , Gravidez , Regiões Promotoras Genéticas
18.
Rev Chil Pediatr ; 87(5): 335-342, 2016.
Artigo em Espanhol | MEDLINE | ID: mdl-27692574

RESUMO

Current evidence supports the notion that exposure to various environmental conditions in early life may induce permanent changes in the epigenome that persist throughout the life-course. This article focuses on early changes associated with obesity in adult life. A review is presented on the factors that induce changes in whole genome (DNA) methylation in early life that are associated with adult onset obesity and related disorders. In contrast, reversal of epigenetic changes associated with weight loss in obese subjects has not been demonstrated. This contrasts with well-established associations found between obesity related DNA methylation patterns at birth and adult onset obesity and diabetes. Epigenetic markers may serve to screen indivuals at risk for obesity and assess the effects of interventions in early life that may delay or prevent obesity in early life. This might contribute to lower the obesity-related burden of death and disability at the population level. The available evidence indicates that epigenetic marks are in fact modifiable, based on modifications in the intrauterine environment and changes in food intake, physical activity and dietary patterns patterns during pregnancy and early years of adult life. This offers the opportunity to intervene before conception, during pregnancy, infancy, childhood, and also in later life. There must be documentation on the best preventive actions in terms of diet and physical activity that will modify or revert the adverse epigenetic markers, thus preventing obesity and diabetes in suceptible individuals and populations.


Assuntos
Diabetes Mellitus Tipo 2/epidemiologia , Epigênese Genética , Obesidade/epidemiologia , Adulto , Metilação de DNA , Diabetes Mellitus Tipo 2/genética , Diabetes Mellitus Tipo 2/prevenção & controle , Dieta , Ingestão de Alimentos/fisiologia , Exercício Físico/fisiologia , Humanos , Obesidade/genética , Obesidade/prevenção & controle , Redução de Peso/fisiologia
19.
BMC Pregnancy Childbirth ; 15: 175, 2015 Aug 18.
Artigo em Inglês | MEDLINE | ID: mdl-26283529

RESUMO

BACKGROUND: Maternal obesity before and during pregnancy predicts maternal and infant risks of obesity and its associated metabolic conditions. Dietary and physical activity recommendations during pregnancy as well as weight monitoring are currently available in the Chilean primary health care system. However some of these recommendations are not updated and most of them are poorly implemented. We seek to assess the effectiveness of an intervention that enhances the implementation of updated nutrition health care standards (diet, physical activity, and breastfeeding promotion) during pregnancy on maternal weight gain and infant growth. DESIGN & SETTING: Cluster randomized controlled trial. The cluster units will be 12 primary health care centers from two counties (La Florida and Puente Alto) from the South-East Area of Santiago randomly allocated to: 1) enhanced nutrition health care standards (intervention group) or 2) routine care (control group). PARTICIPANTS: Women seeking prenatal care before 15 weeks of gestation, residing within a catchment area of selected health centers, and who express that they are not planning to change residence will be invited to participate in the study. Pregnant women classified as high risk according to the Chilean norms (i.e age <16 or >40 years, multiple gestation, pre-gestational medical conditions, previous pregnancy-related issues) and/or underweight will be excluded. INTERVENTION: Pregnant women who attend intervened health care centers starting at their first prenatal visit will receive advice regarding optimal weight gain during pregnancy and diet and physical activity counseling-support. Pregnant women who attend control health clinics will receive routine antenatal care according to national guidelines. We plan to recruit 200 women in each health center. Assuming a 20% loss to follow up, we expect to include 960 women per arm. MAIN OUTCOME MEASURES: 1) Achievement of adequate weight gain based on IOM 2009 recommendations and adequate glycaemic control at 24-28 weeks of pregnancy according to ADA 2011, and 2) healthy infant growth during the first year of age based on WHO standards. DISCUSSION: We expect that the intervention will benefit the participants in achieving adequate weight gain & metabolic control during pregnancy as well as adequate infant growth as a result of an increased impact of standard nutrition and health care practices. Gathered information should contribute to a better understanding of how to develop effective interventions to halt the maternal obesity epidemic and its associated co-morbidities in the Chilean population. TRIAL REGISTRATION: Clinicaltrials.gov Identifier: NCT01916603.


Assuntos
Aleitamento Materno , Desenvolvimento Infantil , Dieta , Atividade Motora , Obesidade/terapia , Cuidado Pós-Natal/métodos , Complicações na Gravidez/terapia , Cuidado Pré-Natal/métodos , Adolescente , Adulto , Chile , Estudos de Coortes , Feminino , Humanos , Lactente , Recém-Nascido , Masculino , Fenômenos Fisiológicos da Nutrição Materna , Estado Nutricional , Sobrepeso/terapia , Gravidez , Avaliação de Programas e Projetos de Saúde , Aumento de Peso , Adulto Jovem
20.
Pediatr Obes ; 19(6): e13120, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38590200

RESUMO

Maternal obesity is a well-known risk factor for developing premature obesity, metabolic syndrome, cardiovascular disease and type 2 diabetes in the progeny. The development of white adipose tissue is a dynamic process that starts during prenatal life: fat depots laid down in utero are associated with the proportion of fat in children later on. How early this programming takes place is still unknown. However, recent evidence shows that mesenchymal stem cells (MSC), the embryonic adipocyte precursor cells, show signatures of the early setting of an adipogenic committed phenotype when exposed to maternal obesity. This review aims to present current findings on the cellular adaptations of MSCs from the offspring of women with obesity and how the metabolic environment of MSCs could affect the early commitment towards adipocytes. In conclusion, maternal obesity can induce early programming of fetal adipose tissue by conditioning MSCs. These cells have higher expression of adipogenic markers, altered insulin signalling and mitochondrial performance, compared to MSCs of neonates from lean pregnancies. Fetal MSCs imprinting by maternal obesity could help explain the increased risk of childhood obesity and development of further noncommunicable diseases.


Assuntos
Células-Tronco Mesenquimais , Obesidade Materna , Efeitos Tardios da Exposição Pré-Natal , Humanos , Feminino , Gravidez , Obesidade Materna/metabolismo , Tecido Adiposo , Obesidade Infantil , Adipogenia/fisiologia , Recém-Nascido , Adipócitos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA