Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 157
Filtrar
1.
Acta Obstet Gynecol Scand ; 103(5): 862-872, 2024 May.
Artigo em Inglês | MEDLINE | ID: mdl-38282287

RESUMO

INTRODUCTION: Maternal obesity, a health condition increasingly prevalent worldwide, has been suggested to be associated with a higher risk of birth defects in offspring, whereas evidence from population-based data from China was largely lacking. Additionally, the role of gestational diabetes in the association between maternal obesity and birth defects remains unclear. We aimed to investigate the association of maternal pre-pregnancy overweight or obesity with any and different types of birth defects in offspring and the interaction between pre-pregnancy overweight or obesity and gestational diabetes. MATERIAL AND METHODS: We conducted a population-based cohort study including 257 107 singletons born between 2015 and 2021 in Longgang District, Shenzhen, China, using data from the Shenzhen Maternal and Child Health Management System. Poisson regression was conducted to estimate the associations of maternal pre-pregnancy overweight or obesity, as well as the interaction between pre-pregnancy overweight or obesity and gestational diabetes, with the risk of birth defects. Models were adjusted for maternal age at delivery, educational level, type of household registration, and gravidity. RESULTS: Maternal pre-pregnancy overweight was associated with a higher risk of any birth defect (risk ratio [RR] 1.21, 95% confidence interval [CI] 1.12 to 1.31) as well as of congenital malformations of the circulatory system (RR 1.26, 95% CI 1.12 to 1.41), eye/ear/face/neck (RR 1.42, 95% CI 1.04 to 1.94), and musculoskeletal system (RR 1.21, 95% CI 1.01 to 1.44). Maternal pre-pregnancy obesity was associated with a higher risk of any birth defect (RR 1.38, 95% CI 1.18 to 1.63) and congenital malformations of the circulatory system (RR 1.61, 95% CI 1.30 to 1.98). Infants born to overweight or obese mothers with gestational diabetes had a higher risk of congenital malformations of the circulatory system than infants born to overweight or obese mothers without gestational diabetes. CONCLUSIONS: Maternal pre-pregnancy overweight or obesity was associated with a higher risk of birth defects, particularly congenital malformations of the circulatory system, in offspring. Gestational diabetes interacts additively with pre-pregnancy overweight or obesity on modifying the risk of congenital malformations of the circulatory system. The importance of improving weight management and assessment of glucose and metabolic functions was emphasized among women planning for pregnancy who are overweight or obese.


Assuntos
Diabetes Gestacional , Obesidade Materna , Lactente , Criança , Feminino , Gravidez , Humanos , Sobrepeso/complicações , Sobrepeso/epidemiologia , Diabetes Gestacional/epidemiologia , Estudos de Coortes , Obesidade Materna/complicações , Índice de Massa Corporal , Obesidade/complicações , Obesidade/epidemiologia , Peso ao Nascer , Parto
2.
Sci Rep ; 14(1): 1466, 2024 01 17.
Artigo em Inglês | MEDLINE | ID: mdl-38233459

RESUMO

Now, no recommendations of gestational weight gain (GWG) after gestational diabetes mellitus (GDM) diagnosis for Chinese women was made. This study aimed to explore the optimal GWG after oral glucose tolerance test (OGTT) for Chinese women with GDM. The GWG status of 11,570 women was retrospectively analyzed. Binary regression model and restricted cubic spline were used to estimate the association between GWG after OGTT and the predicted probability of adverse outcomes. Based on above, the optimal GWG was defined as the range that not exceed 1% increase in the predicted probability from the lowest point. Results shown that every increased one unit GWG after OGTT was associated with higher risks of macrosomia, cesarean section and LGA, and lower risk of preterm birth. According to the WHO and Working Group on Obesity in China (WGOC) recommended pre-pregnancy BMI category, the optimal GWG were proposed: 3.66 to 6.66 kg/3.66 to 6.66 kg in underweight group, 3.07 to 6.50 kg/3.02 to 6.40 kg in normal weight group, 1.06 to 2.73 kg/0 to 1.99 kg in overweight group, and not applicable/- 0.22 to 2.53 kg in obese group, respectively. Therefore, it is necessary to classified Chinese population based on the WGOC recommended pre-pregnancy BMI category, that influenced the contribution of pre-pregnancy BMI groups and the optimal GWG recommendation for GDM women with overweight or obesity.


Assuntos
Diabetes Gestacional , Ganho de Peso na Gestação , Obesidade Materna , Nascimento Prematuro , Gravidez , Recém-Nascido , Humanos , Feminino , Diabetes Gestacional/diagnóstico , Diabetes Gestacional/epidemiologia , Sobrepeso/complicações , Teste de Tolerância a Glucose , Resultado da Gravidez/epidemiologia , Estudos Retrospectivos , Cesárea , Índice de Massa Corporal , Obesidade/complicações , Obesidade Materna/complicações , China
3.
Acta Physiol (Oxf) ; 240(3): e14090, 2024 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-38230587

RESUMO

AIM: Offspring of obese mothers are at high risk of developing metabolic syndrome and cognitive disabilities. Impaired metabolism has also been reported in the offspring of obese fathers. However, whether brain function can also be affected by paternal obesity has barely been examined. This study aimed to characterize the learning deficits resulting from paternal obesity versus those induced by maternal obesity and to identify the underlying mechanisms. METHODS: Founder control and obese female and male Wistar rats were mated to constitute three first-generation (F1) experimental groups: control mother/control father, obese mother/control father, and obese father/control mother. All F1 animals were weaned onto standard chow and underwent a learning test at 4 months of age, after which several markers of glutamate-mediated synaptic plasticity together with the expression of miRNAs targeting glutamate receptors and the concentration of kynurenic and quinolinic acids were quantified in the hippocampus and frontal cortex. RESULTS: Maternal obesity induced a severe learning deficit by impairing memory encoding and memory consolidation. The offspring of obese fathers also showed reduced memory encoding but not impaired long-term memory formation. Memory deficits in offspring of obese fathers and obese mothers were associated with a down-regulation of genes encoding NMDA glutamate receptors subunits and several learning-related genes along with impaired expression of miR-296 and miR-146b and increased concentration of kynurenic acid. CONCLUSION: Paternal and maternal obesity impair offspring's learning abilities by affecting different processes of memory formation. These cognitive deficits are associated with epigenetic and neurochemical alterations leading to impaired glutamate-mediated synaptic plasticity.


Assuntos
MicroRNAs , Obesidade Materna , Humanos , Adulto , Ratos , Feminino , Masculino , Gravidez , Animais , Obesidade Materna/complicações , Obesidade Materna/genética , Ratos Wistar , Obesidade , Pai , Encéfalo , Receptores de Glutamato/genética , Glutamatos/genética , Epigênese Genética
5.
Andrology ; 12(2): 327-337, 2024 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-37424437

RESUMO

BACKGROUND: Low testosterone (T) levels in men associate with increased risks of obesity, type 2 diabetes, metabolic syndrome, and cardiovascular diseases. However, most studies are cross-sectional with follow-up-time < 10 years, and data on early growth are limited. OBJECTIVE: To compare prenatal factors and body mass index (BMI) development from birth to age 46 in relation to low T at age 31. MATERIALS AND METHODS: Men with low T (T < 12.1 nmol/L, n = 132) and men with normal T at age 31 (n = 2561) were derived from the Northern Finland Birth Cohort 1966. Prenatal factors, longitudinal weight and height data from birth to age 14, and cross-sectional weight and height data at ages 31 and 46, and waist-hip-ratio (WHR) and T levels at age 31 were analyzed. Longitudinal modeling and timing of adiposity rebound (AR, second BMI rise at age 5-7 years) were calculated from fitted BMI curves. Results were adjusted for mother's pre-pregnancy BMI and smoking status, birth weight for gestational age, alcohol consumption, education level, smoking status, and WHR at age 31. RESULTS: Neither gestational age nor birth weight was associated with low T at age 31; however, maternal obesity during gestation was more prevalent among men with low T (9.8% vs. 3.5%, adjusted aOR: 2.43 [1.19-4.98]). Men with low T had earlier AR (5.28 vs. 5.82, aOR: 0.73 [0.56-0.94]) and higher BMI (p < 0.001) from AR onward until age 46. Men with both early AR and low T had the highest BMI from AR onward. CONCLUSIONS: In men, maternal obesity and early weight gain associate with lower T levels at age 31, independently of adulthood abdominal obesity. Given the well-known health risks related to obesity, and the rising prevalence of maternal obesity, the results of the present study emphasize the importance of preventing obesity that may also affect the later reproductive health of the offspring.


Assuntos
Diabetes Mellitus Tipo 2 , Obesidade Materna , Masculino , Humanos , Criança , Feminino , Gravidez , Adulto , Pré-Escolar , Pessoa de Meia-Idade , Adolescente , Índice de Massa Corporal , Estudos de Coortes , Peso ao Nascer , Obesidade Materna/complicações , Diabetes Mellitus Tipo 2/complicações , Estudos Transversais , Obesidade/epidemiologia , Obesidade/complicações , Testosterona , Fatores de Risco
6.
Curr Probl Cardiol ; 49(1 Pt B): 102062, 2024 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-37652110

RESUMO

Maternal obesity may affect offspring's cardiovascular health. Our literature search using PubMed, Web of Sciences included original English research and Google Scholar articles published over the past ten years, culminating in 96 articles in this topic. A mother's obesity during pregnancy has a negative impact on the cardiovascular risk for their offspring. Dependence was observed in relation to hypertension, coronary artery disease, stroke, and heart failure. The adverse impact of an abnormal diet in pregnant mice on heart hypertrophy was observed, and was also confirmed in human research. Pregnant women with obesity were at greater risk of having a child with innate heart disease than pregnant women with normal mass. To conclude: mother's obesity has a negative impact on the long-term cardiovascular consequences for their offspring, increasing their risk of high blood pressure, coronary heart disease, stroke and heart failure. It also increases the probability of heart hypertrophy and innate heart defects.


Assuntos
Insuficiência Cardíaca , Obesidade Materna , Acidente Vascular Cerebral , Animais , Criança , Feminino , Humanos , Camundongos , Gravidez , Cardiomegalia , Obesidade/complicações , Obesidade/epidemiologia , Obesidade Materna/complicações , Obesidade Materna/epidemiologia
7.
Nutrients ; 15(21)2023 Oct 31.
Artigo em Inglês | MEDLINE | ID: mdl-37960276

RESUMO

Chronic diseases represent one of the major causes of death worldwide. It has been suggested that pregnancy-related conditions, such as gestational diabetes mellitus (GDM), maternal obesity (MO), and intra-uterine growth restriction (IUGR) induce an adverse intrauterine environment, increasing the offspring's predisposition to chronic diseases later in life. Research has suggested that mitochondrial function and oxidative stress may play a role in the developmental programming of chronic diseases. Having this in mind, in this review, we include evidence that mitochondrial dysfunction and oxidative stress are mechanisms by which GDM, MO, and IUGR program the offspring to chronic diseases. In this specific context, we explore the promising advantages of maternal antioxidant supplementation using compounds such as resveratrol, curcumin, N-acetylcysteine (NAC), and Mitoquinone (MitoQ) in addressing the metabolic dysfunction and oxidative stress associated with GDM, MO, and IUGR in fetoplacental and offspring metabolic health. This approach holds potential to mitigate developmental programming-related risk of chronic diseases, serving as a probable intervention for disease prevention.


Assuntos
Diabetes Gestacional , Obesidade Materna , Complicações na Gravidez , Efeitos Tardios da Exposição Pré-Natal , Gravidez , Feminino , Humanos , Antioxidantes/farmacologia , Efeitos Tardios da Exposição Pré-Natal/prevenção & controle , Efeitos Tardios da Exposição Pré-Natal/etiologia , Resveratrol/farmacologia , Diabetes Gestacional/prevenção & controle , Complicações na Gravidez/prevenção & controle , Dieta , Obesidade Materna/complicações , Retardo do Crescimento Fetal/prevenção & controle , Doença Crônica
8.
Eur J Neurosci ; 58(11): 4393-4422, 2023 12.
Artigo em Inglês | MEDLINE | ID: mdl-37974556

RESUMO

Obesity, affecting one in three pregnant women worldwide, is not only a major obstetric risk factor. The resulting low-grade inflammation may have a long-term impact on the offspring's HPA axis through dysregulation of maternal, placental and fetal corticosteroid metabolism, and children born of obese mothers have increased risk of diabetes and cardiovascular disease. The long-term effects of maternal obesity on offspring neurodevelopment are, however, undetermined and could depend on the specific effects on placental and fetal cortisol metabolism. This systematic review evaluates how maternal obesity affects placental cortisol metabolism and the offspring's HPA axis. Pubmed, Embase and Scopus were searched for original studies on maternal BMI, obesity, and cortisol metabolism and transfer. Fifteen studies were included after the screening of 4556 identified records. Studies were small with heterogeneous exposures and outcomes. Two studies found that maternal obesity reduced placental HSD11ß2 activity. In one study, umbilical cord blood cortisol levels were affected by maternal BMI. In three studies, an altered cortisol response was consistently seen among offspring in childhood (n = 2) or adulthood (n = 1). Maternal BMI was not associated with placental HSD11ß1 or HSD11ß2 mRNA expression, or placental HSD11ß2 methylation. In conclusion, high maternal BMI is associated with reduced placental HSD11ß2 activity and a dampened cortisol level among offspring, but the data is sparse. Further investigations are needed to clarify whether the HPA axis is affected by prenatal factors including maternal obesity and investigate if adverse effects can be ameliorated by optimising the intrauterine environment.


Assuntos
Obesidade Materna , Efeitos Tardios da Exposição Pré-Natal , Criança , Humanos , Feminino , Gravidez , Adulto , Placenta/metabolismo , Hidrocortisona/metabolismo , Obesidade Materna/complicações , Obesidade Materna/metabolismo , Sistema Hipotálamo-Hipofisário/metabolismo , Efeitos Tardios da Exposição Pré-Natal/metabolismo , Sistema Hipófise-Suprarrenal/metabolismo , Obesidade/metabolismo
9.
Pediatr Obes ; 18(12): e13081, 2023 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-37859518

RESUMO

BACKGROUND: Leptin resistance occurs with obesity, but it is unknown if individuals at risk for obesity develop leptin resistance prior to obesity. OBJECTIVE: Investigate whether leptin resistance is independent of weight status in children at risk for obesity due to intrauterine exposure to maternal obesity or gestational diabetes mellitus (GDM). METHODS: Mother-child dyads (N = 179) were grouped by maternal pregnancy weight and GDM status: (1) normal weight, no GDM; (2) overweight/obesity, no GDM; (3) overweight/obesity with GDM. Children (4-10 years) were further stratified by current body mass index (BMI) <85th or ≥85th percentile. Leptin resistance of children and mothers was calculated as fasting leptin/fat mass index. Two-way ANOVA was used to assess whether leptin concentrations and leptin resistance differed by current weight status or in utero exposure group, after adjusting for race, sex and Tanner stage. RESULTS: Children with a BMI ≥85th percentile had more leptin resistance than those with a BMI <85th percentile (p < 0.001), but leptin resistance did not differ by in utero exposure. Similarly, leptin resistance in women was associated with weight status and not prior GDM. CONCLUSIONS: Results suggest that leptin concentrations are associated with obesity but not risk for obesity based on in utero exposure to maternal obesity or GDM.


Assuntos
Diabetes Gestacional , Obesidade Materna , Feminino , Humanos , Gravidez , Peso ao Nascer , Índice de Massa Corporal , Diabetes Gestacional/epidemiologia , Leptina , Obesidade/epidemiologia , Obesidade/complicações , Obesidade Materna/complicações , Sobrepeso/complicações , Fatores de Risco , Pré-Escolar , Criança
10.
Am J Physiol Lung Cell Mol Physiol ; 325(5): L662-L674, 2023 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-37786934

RESUMO

Early life over-nutrition, as experienced in maternal obesity, is a risk factor for developing cardiorespiratory and metabolic diseases. Here we investigated the effects of high-fat diet (HFD) consumption on the breathing pattern and sympathetic discharge to blood vessels in juvenile offspring from dams fed with HFD (O-HFD). Adult female Holtzman rats were given a standard diet (SD) or HFD from 6 wk before gestation to weaning. At weaning (P21), the male offspring from SD dams (O-SD) and O-HFD received SD until the experimental day (P28-P45). Nerve recordings performed in decerebrated in situ preparations demonstrated that O-HFD animals presented abdominal expiratory hyperactivity under resting conditions and higher vasoconstrictor sympathetic activity levels. The latter was associated with blunted respiratory-related oscillations in sympathetic activity, especially in control animals. When exposed to elevated hypercapnia or hypoxia levels, the O-HFD animals mounted similar ventilatory and respiratory motor responses as the control animals. Hypercapnia and hypoxia exposure also increased sympathetic activity in both groups but did not reinstate the respiratory-sympathetic coupling in the O-HFD rats. In freely behaving conditions, O-HFD animals exhibited higher resting pulmonary ventilation and larger variability of arterial pressure levels than the O-SD animals due to augmented sympathetic modulation of blood vessel diameter. Maternal obesity modified the functioning of cardiorespiratory systems in offspring at a young age, inducing active expiration and sympathetic overactivity under resting conditions. These observations represent new evidence about pregnancy-related complications that lead to the development of respiratory distress and hypertension in children of obese mothers.NEW & NOTEWORTHY Maternal obesity is a risk factor for developing cardiorespiratory and metabolic diseases. This study highlights the changes on the breathing pattern and sympathetic discharge to blood vessels in juvenile offspring from dams fed with HFD. Maternal obesity modified the functioning of cardiorespiratory systems in offspring, inducing active expiration and sympathetic overactivity. These observations represent new evidence about pregnancy-related complications that lead to the development of respiratory distress and hypertension in children of obese mothers.


Assuntos
Hipertensão , Doenças Metabólicas , Obesidade Materna , Efeitos Tardios da Exposição Pré-Natal , Síndrome do Desconforto Respiratório , Humanos , Criança , Ratos , Animais , Masculino , Feminino , Gravidez , Dieta Hiperlipídica/efeitos adversos , Obesidade Materna/complicações , Hipercapnia , Respiração , Obesidade , Ratos Sprague-Dawley , Hipóxia/complicações , Doenças Metabólicas/complicações , Síndrome do Desconforto Respiratório/complicações , Efeitos Tardios da Exposição Pré-Natal/metabolismo
11.
Horm Metab Res ; 55(10): 665-676, 2023 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-37813098

RESUMO

Maternal obesity is associated with fetal complications predisposing later to the development of metabolic syndrome during childhood and adult stages. High-fat diet seems to influence individuals and their subsequent generations in mediating weight gain, insulin resistance, obesity, high cholesterol, diabetes, and cardiovascular disorder. Research evidence strongly suggests that epigenetic alteration is the major contributor to the development of metabolic syndrome through DNA methylation, histone modifications, and microRNA expression. In this review, we have discussed the outcome of recent studies on the adverse and beneficial effects of nutrients and vitamins through epigenetics during pregnancy. We have further discussed about the miRNAs altered during maternal obesity. Identification of new epigenetic modifiers such as mesenchymal stem cells condition media (MSCs-CM)/exosomes for accelerating the reversal of epigenetic abnormalities for the development of new treatments is yet another aspect of the present review.


Assuntos
Doenças Metabólicas , Síndrome Metabólica , Obesidade Materna , Adulto , Feminino , Gravidez , Humanos , Síndrome Metabólica/metabolismo , Obesidade Materna/complicações , Obesidade Materna/genética , Obesidade/metabolismo , Doenças Metabólicas/genética , Epigênese Genética
12.
Nutrients ; 15(17)2023 Aug 31.
Artigo em Inglês | MEDLINE | ID: mdl-37686855

RESUMO

Maternal obesity affects the risk of cardiovascular disease and inflammatory response in offspring. However, the impact of maternal obesity on offspring with Kawasaki disease (KD), the leading cause of childhood acquired heart disease, is still an understudied area. This study aimed to elucidate the impact of maternal obesity on offspring in KD-like vasculitis and the underlying mechanisms. Offspring of obese female mice and normal diet dams were randomly divided into two subgroups. The pups were injected intraperitoneally with either Candida albicans water-soluble fraction (CAWS) or phosphate buffered saline (PBS) to establish the obesity (OB)-CAWS group, OB group, wild type (WT)-CAWS group, and WT group. Their weight was monitored during the study. After four weeks, echocardiography was applied to obtain the alternation of cardiac structures. Mouse cytokine panel, Hematoxylin-Eosin (HE) staining, western blot, and real-time qPCR were used to study the pathological changes and protein and RNA expression alternations. Based on the study of pathology, serology and molecular biology, maternal obesity lead to more severe vasculitis and induced altered cardiac structure in the offspring mice and promoted the expression of pro-inflammatory cytokines through activating the NF-κB signaling pathway. Maternal obesity aggravated the inflammatory response of offspring mice in KD-like vasculitis.


Assuntos
Doenças Cardiovasculares , Síndrome de Linfonodos Mucocutâneos , Obesidade Materna , Vasculite , Animais , Feminino , Camundongos , Camundongos Obesos , Obesidade Materna/complicações , Candida albicans
13.
Biol Sex Differ ; 14(1): 66, 2023 09 28.
Artigo em Inglês | MEDLINE | ID: mdl-37770949

RESUMO

BACKGROUND: We have previously reported that maternal obesity reduces placental transport capacity for lysophosphatidylcholine-docosahexaenoic acid (LPC-DHA), a preferred form for transfer of DHA (omega 3) to the fetal brain, but only in male fetuses. Phosphatidylethanolamine (PE) and phosphatidylcholine (PC), have either sn-1 ester, ether or vinyl ether (plasmalogen) linkages to primarily unsaturated and monounsaturated fatty acids and DHA or arachidonic acid (ARA, omega 6) in the sn-2 position. Whether ether and plasmalogen PC and PE metabolism in placenta impacts transfer to the fetus is unexplored. We hypothesized that ether and plasmalogen PC and PE containing DHA and ARA are reduced in maternal-fetal unit in pregnancies complicated by obesity and these differences are dependent on fetal sex. METHODS: In maternal, umbilical cord plasma and placentas from obese women (11 female/5 male infants) and normal weight women (9 female/7 male infants), all PC and PE species containing DHA and ARA were analyzed by LC-MS/MS. Placental protein expression of enzymes involved in phospholipid synthesis, were determined by immunoblotting. All variables were compared between control vs obese groups and separated by fetal sex, in each sample using the Benjamini-Hochberg false discovery rate adjustment to account for multiple testing. RESULTS: Levels of ester PC containing DHA and ARA were profoundly reduced by 60-92% in male placentas of obese mothers, while levels of ether and plasmalogen PE containing DHA and ARA were decreased by 51-84% in female placentas. PLA2G4C abundance was lower in male placentas and LPCAT4 abundance was lower solely in females in obesity. In umbilical cord, levels of ester, ether and plasmalogen PC and PE with DHA were reduced by 43-61% in male, but not female, fetuses of obese mothers. CONCLUSIONS: We found a fetal sex effect in placental PE and PC ester, ether and plasmalogen PE and PC containing DHA in response to maternal obesity which appears to reflect an ability of female placentas to adapt to maintain optimal fetal DHA transfer in maternal obesity.


Docosahexaenoic acid (DHA) is a critical omega 3 long chain polyunsaturated fatty acid (LCPUFA) for fetal brain development. We have recently reported that maternal obesity reduces placental transport capacity for LysophosPhatidylCholine-DHA (LPC-DHA), a preferred form for transfer of DHA to the fetal brain, but only in male fetuses. Other important lipids, the plasmalogen phosphatidylcholine (PC) and phosphatidylethanolamine (PE) are considered DHA reservoirs, but its roles in the maternal­fetal unit are largely unexplored. We examined these lipid species in maternal and fetal circulation and in placental tissue to uncover potential novel roles for ether and plasmalogen lipids in the regulation of placenta delivery of these vital nutrients in pregnancies complicated by obesity depending of fetal sex. We demonstrated for the first time, that female fetuses of obese mothers decrease placental ether and plasmalogen PE containing DHA and arachidonic acid (ARA, omega 6), and show a high fetal­placental adaptability and placental reserve capacity that can maintain the PC-LCPUFA synthesis and the transfer of these crucial species to the fetus to preserve brain development. Our study also demonstrated that male fetuses, in response to maternal obesity, reduce the placental ester PC species containing DHA and ARA and reduce the ether and plasmalogen PE reservoir of DHA and ARA in fetal circulation. Our findings support a fetal sex effect in placental ester, ether and plasmalogen PE and PC containing DHA in response to maternal obesity which appears to reflect an ability of female placentas to adapt to maintain optimal fetal DHA transfer in maternal obesity.


Assuntos
Obesidade Materna , Placenta , Lactente , Feminino , Humanos , Masculino , Gravidez , Placenta/metabolismo , Fosfatidilcolinas/metabolismo , Fosfatidiletanolaminas/metabolismo , Plasmalogênios/metabolismo , Éter , Obesidade Materna/complicações , Obesidade Materna/metabolismo , Caracteres Sexuais , Cromatografia Líquida , Espectrometria de Massas em Tandem , Obesidade/metabolismo , Etil-Éteres/metabolismo , Éteres/metabolismo
14.
Artigo em Inglês | MEDLINE | ID: mdl-37738759

RESUMO

Obesity is a significant health concern worldwide and is associated with numerous health complications, including placental dysfunction during pregnancy. Placental dysfunction can lead to adverse outcomes for both the mother and the foetus, such as preeclampsia, gestational diabetes, preterm birth, and foetal growth restriction. Studies have shown that maternal obesity can lead to placental dysfunction through various mechanisms, including chronic inflammation, oxidative stress, and dysregulation of metabolic pathways. These factors can contribute to changes in the placenta's structure and function, impairing nutrient and oxygen exchange between the mother and foetus. Recent research has also suggested that alteration of gene expression in the placenta due to epigenetic changes, such as DNA methylation, may play a role in placental dysfunction associated with maternal obesity. These changes can affect altering foetal growth and development. Prevention and management of maternal obesity are crucial in reducing the risk of placental dysfunction and associated adverse outcomes during pregnancy. This can be achieved through lifestyle modifications, such as diet and exercise, and early detection and management of underlying health conditions. In conclusion, maternal obesity is a significant risk factor for placental dysfunction during pregnancy, which can lead to adverse outcomes for both the mother and the foetus. Further research is needed to understand the relationship and mechanisms to develop effective interventions to prevent and manage placental dysfunction in obese pregnant women.


Assuntos
Obesidade Materna , Nascimento Prematuro , Feminino , Gravidez , Recém-Nascido , Humanos , Placenta , Obesidade Materna/complicações , Obesidade Materna/metabolismo , Nascimento Prematuro/metabolismo , Obesidade/complicações , Gravidez de Alto Risco
15.
J Matern Fetal Neonatal Med ; 36(2): 2245102, 2023 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-37574213

RESUMO

OBJECTIVE: The aim of the present study was to assess the impact of different maternal Body Mass Index (BMI) classes on the risk of postpartum endometritis, wound infection, and breast abscess after different modes of delivery. Secondly to estimate how the risk of postpartum infection varies with different maternal BMI groups after induction of labor and after obstetric anal sphincter injuries. METHODS: A population-based observational study including women who gave birth during eight years (N = 841,780). Data were collected from three Swedish Medical Health Registers, the Swedish Medical Birth Register, the Swedish National Patient Register, and the Swedish Prescribed Drug Register. Outcomes were defined by ICD-10 codes given within eight weeks postpartum. The reference population was uninfected women. Odds ratios were determined using Mantel-Haenszel technique. Year of delivery, maternal age, parity and smoking in early pregnancy were considered as confounders. RESULTS: There was a dose-dependent relationship between an increasing maternal BMI and a higher risk for postpartum infections. Women in obesity class II and III had an increased risk for endometritis after normal vaginal delivery aOR 1.45 (95% CI: 1.29-1.63) and for wound infections after cesarean section aOR 3.83 (95% CI: 3.39-4.32). There was no difference in how maternal BMI affected the association between cesarean section and wound infection, regardless of whether it was planned or emergent. Women in obesity class II and III had a lower risk of breast abscess compared with normal-weight women, aOR 0.47 (95% CI: 0.38-0.58). The risk of endometritis after labor induction decreased with increasing maternal BMI. The risk of wound infection among women with an obstetrical sphincter injury decreased with increasing BMI. CONCLUSION: This study provides new knowledge about the impact of maternal BMI on the risk of postpartum infections after different modes of delivery. There was no difference in how BMI affected the association between cesarean section and wound infections, regardless of whether it was a planned cesarean section or an emergency cesarean section.


Assuntos
Endometrite , Obesidade Materna , Infecção dos Ferimentos , Gravidez , Feminino , Humanos , Cesárea/efeitos adversos , Obesidade Materna/complicações , Endometrite/etiologia , Endometrite/complicações , Abscesso/complicações , Parto , Obesidade/complicações , Obesidade/epidemiologia , Período Pós-Parto , Infecção dos Ferimentos/complicações
16.
Diabetes Res Clin Pract ; 203: 110889, 2023 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-37633339

RESUMO

AIM: To evaluate the association between maternal obesity, gestational diabetes (GDM), and birth size with infant fat-mass (FM) accretion from 1 to 6 months (M). METHODS: Healthy pregnant women and their term babies from the OBESO cohort were studied (1 M-3 M, n = 122; 1 M-6 M, n = 90). Registered maternal data was: pregestational body-mass-index (preBMI), GDM (2hOGTT), medications, gestational weight gain. Macrosomia (>4000 g), large/small for gestational age (LGA/SGA)(weight/age > 90° and < 90°, respectively-WHO) were recorded at birth. Infant FM (air-displacement plethysmography) was measured (1 M, 3 M, 6 M) and FM accretion computed (ΔkgFM from 1 M-3 M and 1 M-6 M). Exclusive breastfeeding (EBF) was assessed. Adjusted-multiple linear regression models were performed. RESULTS: PreBMI was 27.4 ± 5.2 kg/m2. GDM was present in9%(n = 11) of women; 12.3%(n = 15) of them received metformin/insulin. One newborn was LGA; 20.7%(n = 25) were SGA. From 1 M-3 M, SGA was a predictor of higher FM accretion (B:0.28, 95%CI:0.14-0.43); GDM was not associated. From 1 M-6 M, higher FM accretion was observed in SGA newborns (B:0.43, 95%CI:0.19-0.67) and GDM infants (B:0.48, 95%CI:0.06-0.89). In all models (R2 ≥ 0.48, p < 0.001), infant weight and being female were positively associated, while maternal obesity, metformin/insulin, and EBF were not. CONCLUSIONS: GDM appears to program early higher adiposity accretion, independently of excessive fetal growth. SGA was associated with higher FM accretion in early infancy.


Assuntos
Diabetes Gestacional , Insulinas , Metformina , Obesidade Materna , Lactente , Feminino , Recém-Nascido , Humanos , Gravidez , Masculino , Peso ao Nascer , Adiposidade , Obesidade Materna/complicações , Obesidade/complicações , Macrossomia Fetal/etiologia , Macrossomia Fetal/complicações , Aumento de Peso , Índice de Massa Corporal , Metformina/uso terapêutico
17.
Biochim Biophys Acta Mol Basis Dis ; 1869(8): 166834, 2023 12.
Artigo em Inglês | MEDLINE | ID: mdl-37541330

RESUMO

Gestational diabetes mellitus (GDM) and maternal obesity (MO) increase the risk of adverse fetal outcomes, and the incidence of cardiovascular disease later in life. Extensive research has been conducted to elucidate the underlying mechanisms by which GDM and MO program the offspring to disease. This review focuses on the role of fetoplacental endothelial dysfunction in programming the offspring for cardiovascular disease in GDM and MO pregnancies. We discuss how pre-existing maternal health conditions can lead to vascular dysfunction in the fetoplacental unit and the fetus. We also examine the role of fetoplacental endothelial dysfunction in impairing fetal cardiovascular system development and the involvement of nitric oxide and hydrogen sulfide in mediating fetoplacental vascular dysfunction. Furthermore, we suggest that the L-Arginine-Nitric Oxide and the Adenosine-L-Arginine-Nitric Oxide (ALANO) signaling pathways are pertinent targets for research. Despite significant progress in this area, there are still knowledge gaps that need to be addressed in future research.


Assuntos
Doenças Cardiovasculares , Diabetes Gestacional , Obesidade Materna , Gravidez , Feminino , Humanos , Diabetes Gestacional/metabolismo , Placenta/metabolismo , Óxido Nítrico/metabolismo , Doenças Cardiovasculares/metabolismo , Obesidade Materna/complicações , Obesidade Materna/metabolismo , Arginina/metabolismo
18.
Placenta ; 140: 100-108, 2023 09 07.
Artigo em Inglês | MEDLINE | ID: mdl-37566941

RESUMO

INTRODUCTION: Maternal obesity is associated with increased risk of offspring obesity and cardiometabolic disease. Altered fetoplacental immune programming is a potential candidate mechanism. Differences in fetal placental macrophages, or Hofbauer cells (HBCs), have been observed in maternal obesity, and lipid metabolism is a key function of resident macrophages that may be deranged in inflammation/immune activation. We sought to test the following hypotheses: 1) maternal obesity is associated with altered HBC density and phenotype in the term placenta and 2) obesity-associated HBC changes are associated with altered placental lipid transport to the fetus. The impact of fetal sex was evaluated in all experiments. METHODS: We quantified the density and morphology of CD163-and CD68-positive HBCs in placental villi in 34 full-term pregnancies undergoing cesarean delivery (N = 15, maternal BMI ≥30 kg/m2; N = 19, BMI <30 kg/m2). Antibody-positive cells in terminal villi were detected and cell size and circularity analyzed using a semi-automated method for thresholding of bright-field microscopy images (ImageJ). Placental expression of lipid transporter genes was quantified using RTqPCR, and cord plasma triglycerides (TGs) were profiled using modified Wahlefeld method. The impact of maternal obesity and fetal sex on HBC features, lipid transporters, and cord TGs were evaluated by two-way ANOVA. Spearman correlations of cord TGs, HBC metrics and gene expression levels were calculated. RESULTS: Maternal obesity was associated with significantly increased density of HBCs, with male placentas most affected (fetal sex by maternal obesity interaction p = 0.04). CD163+ HBCs were larger and rounder in obesity-exposed male placentas. Sexually dimorphic expression of placental FATP4, FATP6, FABPPM, AMPKB1 and AMPKG and cord TGs was noted in maternal obesity, such that levels were higher in males and lower in females relative to sex-matched controls. Cord TGs were positively correlated with HBC density and FATP1 expression. DISCUSSION: Maternal obesity is associated with sex-specific alterations in HBC density and placental lipid transporter expression, which may impact umbilical cord blood TG levels and offspring cardiometabolic programming.


Assuntos
Obesidade Materna , Placenta , Humanos , Gravidez , Feminino , Masculino , Placenta/metabolismo , Obesidade Materna/complicações , Obesidade Materna/metabolismo , Sangue Fetal/metabolismo , Macrófagos/metabolismo , Obesidade/complicações , Obesidade/metabolismo , Lipídeos
19.
Am J Clin Nutr ; 118(1): 255-263, 2023 07.
Artigo em Inglês | MEDLINE | ID: mdl-37407164

RESUMO

BACKGROUND: Maternal obesity has been associated with shorter breastfeeding duration, but little is known about mediating factors explaining this association. It is important to assess these relationships across diverse populations because breastfeeding is culturally patterned. OBJECTIVES: We investigated the association of prepregnancy maternal body mass index (BMI) with breastfeeding outcomes and potential mediators of this relationship in 3 culturally diverse international cohorts. METHODS: We analyzed 5120 singleton pregnancies from mother-child cohorts in Spain (INfancia y Medio Ambiente), Greece (Rhea), and the United States (Project Viva). Outcome variables were duration of any and exclusive breastfeeding. A priori hypothesized mediators in the association of maternal prepregnancy BMI with breastfeeding were birthweight (BW), maternal prenatal C-reactive protein (CRP), cesarean delivery, maternal dietary inflammatory index (DII) during pregnancy, gestational age at delivery, and gestational diabetes mellitus (GDM). We estimated the association between BMI and breastfeeding duration using linear regression adjusting for confounders. Mediation analysis estimated direct and indirect effects of maternal overweight/obesity on breastfeeding for each mediator. RESULTS: Women with overweight and obesity had shorter duration of any and exclusive breastfeeding compared with normal-weight women (any: overweight ß = -0.79 mo, 95% CI: -1.17, -0.40; obese ß = -1.75 mo 95% CI: -2.25, -1.25; exclusive: overweight ß = -0.30 mo, 95% CI: -0.42, -0.16; obese ß = -0.73 mo, 95% CI: -0.90, -0.55). Significant mediators (% change in effect estimate) of this association were higher CRP (exclusive: 5.12%), cesarean delivery (any: 6.54%; exclusive: 7.69%), and higher DII (any: 6.48%; exclusive: 7.69%). GDM, gestational age, and BW did not mediate the association of maternal weight status with breastfeeding. CONCLUSIONS: Higher prepregnancy BMI is associated with shorter duration of any and exclusive breastfeeding. Maternal dietary inflammation, systemic inflammation, and mode of delivery may be key modifiable mediators of this association. Identification of mediators provides potential targets for interventions to improve breastfeeding outcomes.


Assuntos
Diabetes Gestacional , Obesidade Materna , Feminino , Gravidez , Humanos , Estados Unidos , Aleitamento Materno , Sobrepeso/complicações , Índice de Massa Corporal , Obesidade/complicações , Obesidade Materna/complicações , Inflamação/complicações , Peso ao Nascer , Proteína C-Reativa
20.
BMC Pregnancy Childbirth ; 23(1): 449, 2023 Jun 16.
Artigo em Inglês | MEDLINE | ID: mdl-37328759

RESUMO

BACKGROUND: Previous studies have suggested that maternal overweight/obesity is asscociated with macrosomia. The present study aimed to investigate the mediation effects of fasting plasma glucose (FPG) and maternal triglyceride (mTG) in the relationship between maternal overweight/obesity and large for gestational age (LGA) among non-diabetes pregnant women. METHODS: This prospective cohort study was conducted in Shenzhen from 2017 to 2021. A total of 19,104 singleton term non-diabetic pregnancies were enrolled form a birth cohort study. FPG and mTG were measured at 24-28 weeks. We analyzed the association of maternal prepregancy overweight/obesity with LGA and mediation effects of FPG and mTG. Multivariable logistic regression analysis and serial multiple mediation analysis were performed. The odds ratio (OR) and 95% confidence intervals (CIs) were calculated. RESULTS: Mothers who were overweight or obese had higher odds of giving birth to LGA after adjusting potential confounders (OR:1.88, 95%CI: 1.60-2.21; OR:2.72, 95%CI: 1.93-3.84, respectively). The serial multiple mediation analysis found prepregnancy overweight can not only have a direct positive effect on LGA (effect = 0.043, 95% CI: 0.028-0.058), but also have an indirect effect on the LGA through two paths: the independent mediating role of FPG (effect = 0.004, 95% CI: 0.002-0.005); the independent mediating role of mTG (effect = 0.003,95% CI: 0.002-0.005). The chain mediating role of FPG and mTG has no indirect effect. The estimated proportions mediated by FPG and mTG were 7.8% and 5.9%. Besides, the prepregnancy obesity also has a direct effect on LGA (effect = 0.076; 95%CI: 0.037-0.118) and an indirect effect on LGA through three paths: the independent mediating role of FPG (effect = 0.006; 95%CI: 0.004-0.009); the independent mediating role of mTG (effect = 0.006; 95%CI: 0.003-0.008), and the chain mediating role of FPG and mTG (effect = 0.001; 95%CI: 0.000-0.001). The estimated proportions were 6.7%, 6.7%, and 1.1%, respectively. CONCLUSION: This study found that in nondiabetic women, maternal overweight/obesity was associated with the occurence of LGA, and this positive association was partly mediated by FPG and mTG, suggesting that FPG and mTG in overweight/obese nondiabetic mothers deserve the attention of clinicians.


Assuntos
Diabetes Gestacional , Obesidade Materna , Feminino , Humanos , Gravidez , Peso ao Nascer , Índice de Massa Corporal , Estudos de Coortes , Jejum , Desenvolvimento Fetal , Macrossomia Fetal/etiologia , Macrossomia Fetal/complicações , Mães , Obesidade Materna/complicações , Sobrepeso/complicações , Sobrepeso/epidemiologia , Estudos Prospectivos , Triglicerídeos/sangue , Glicemia , Ganho de Peso na Gestação , Adulto
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...