Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 833
Filtrar
2.
BMC Pregnancy Childbirth ; 24(1): 610, 2024 Sep 19.
Artículo en Inglés | MEDLINE | ID: mdl-39300411

RESUMEN

BACKGROUND: Gestational diabetes mellitus (GDM) is the most prevalent metabolic disturbance during pregnancy and is associated with adverse outcomes in offspring, including an elevated risk for developing atopic diseases in early childhood. Research is limited regarding only women at risk of GDM among whom some develop GDM while others do not. Information about adverse health outcomes in the offspring of these women is also lacking. The main aim was to assess whether maternal GDM increases the offspring's risk of atopic dermatitis (AD), asthma and allergic rhinitis at 1, 2 and 5 years of age. The second aim was to analyze the association of other maternal health characteristics on the development of these disorders in offspring. METHODS: The follow-up study group of the Gestational Diabetes Study (GDS), conducted at Tartu University Hospital, Estonia, between 2014 and 2020, comprised 223 mother-child dyads. All women had at least one risk factor for GDM, of whom only some developed GDM. Information about the diagnoses of interest was obtained from Electronic Health Records. Allergen-specific IgE from children's serum was measured using ImmunoCAP™ Phadiatop™ Infant, with results ≥ 0.35 kU/l considered positive. Statistical analysis was performed using the RStudio software (version 4.3.0). RESULTS: According to our results, only the cases of GDM requiring the use of antidiabetic medications were associated with the development of asthma and/or allergic rhinitis at 2 years of age (aOR 4.68, 95%CI 1.08-20.21, p = 0.039). Maternal obesity (BMI > 30) was associated with offspring´s asthma and/or allergic rhinitis diagnosis at 2 years of age (aOR 3.15, 95%CI 1.03-9.63, p = 0.045). Maternal abnormal weight gain during pregnancy was associated with asthma and/or allergic rhinitis at 5 years of age (aOR 2.76, 95%CI 1.04-7.31, p = 0.041). CONCLUSION: Among pregnant women at risk for GDM, maternal weight-related factors significantly influence the development of atopic diseases in their children between 1 and 5 years of age, regardless of the GDM diagnosis. This suggests that, besides women with GDM greater attention should also be paid to women at risk but who do not develop GDM, as their children seem to be at higher risk of atopic diseases.


Asunto(s)
Asma , Dermatitis Atópica , Diabetes Gestacional , Efectos Tardíos de la Exposición Prenatal , Humanos , Diabetes Gestacional/epidemiología , Embarazo , Femenino , Asma/epidemiología , Asma/etiología , Estudios de Seguimiento , Preescolar , Adulto , Dermatitis Atópica/epidemiología , Lactante , Factores de Riesgo , Efectos Tardíos de la Exposición Prenatal/epidemiología , Masculino , Rinitis Alérgica/epidemiología , Madres/estadística & datos numéricos
3.
BMC Womens Health ; 24(1): 520, 2024 Sep 18.
Artículo en Inglés | MEDLINE | ID: mdl-39294652

RESUMEN

Maternal obesity rates are increasing significantly, posing substantial risks to both mothers and their children. This study aims to introduce health policies addressing maternal obesity, identify preventive interventions, and highlight scientific gaps necessitating further research.We identified documents through electronic searches in PubMed, CINAHL Plus, EMBASE, and grey literature sources (ministry of health websites, national gynecology and obstetrics associations) from January 2013 to August 2023, updated in June 2024. The inclusion criteria focused on English-language documents discussing interventions or health policies that promote weight loss through lifestyle changes during pregnancy.A total of 22 documents (10 studies and 12 guidelines) were included. 12 studies (N=1244) identified via databases; included two Clinical Practice Guidelines (CPGs) from Canada and Singapore. Other 10 CPGs sourced from governmental websites and national associations: England (1), Australia (1), New Zealand (1), combined Australia and New Zealand (1), Canada (3), USA (1), Ireland (1), Germany (1). 10 guidelines focused on obesity in pregnancy, two on weight management during pregnancy. Covered interventions across pre-pregnancy, pregnancy, and postpartum periods (9 guidelines); pre-pregnancy and pregnancy (2); exclusively postpartum (1). Seven guidelines offered evidence-based recommendations on maintaining healthy weight in mothers, largely based on expert opinions.Maternal obesity poses significant risks to both mothers and children, underscoring the need for effective health policies and systems. However, few countries have integrated adequate responses into their healthcare policies and guidelines for professionals. Limited evidence exists on optimal practices to improve reproductive health outcomes in obese women. Hence, the crucial need to developing comprehensive guidelines and proactive strategies to manage maternal obesity. These measures can improve outcomes and reduce healthcare costs. Increased focus on research and policymaking is essential to protect the health of mothers and their children.


Asunto(s)
Política de Salud , Humanos , Femenino , Embarazo , Obesidad Materna , Manejo de la Obesidad/métodos , Guías de Práctica Clínica como Asunto , Complicaciones del Embarazo/prevención & control , Complicaciones del Embarazo/terapia
4.
Adv Anat Embryol Cell Biol ; 239: 157-197, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39283486

RESUMEN

Maternal nutrition and metabolic health status during pregnancy are critical factors that shape the life-long health trajectory of offspring. Altered nutrition during specific times of development in utero can lead to functional changes in tissues such as the pancreatic ß-cells, predisposing those tissues to metabolic diseases and Type 2 diabetes that manifest later in life. This chapter will focus on the role of pregnancy complications with altered nutrition during gestation in the maladaptive programming of ß-cell mass and function in the offspring.


Asunto(s)
Células Secretoras de Insulina , Femenino , Embarazo , Células Secretoras de Insulina/metabolismo , Humanos , Fenómenos Fisiologicos Nutricionales Maternos , Animales , Efectos Tardíos de la Exposición Prenatal/metabolismo , Estado Nutricional , Complicaciones del Embarazo , Diabetes Mellitus Tipo 2/metabolismo
5.
Int J Mol Sci ; 25(17)2024 Aug 31.
Artículo en Inglés | MEDLINE | ID: mdl-39273445

RESUMEN

Limb muscle is responsible for physical activities and myogenic cell migration during embryogenesis is indispensable for limb muscle formation. Maternal obesity (MO) impairs prenatal skeletal muscle development, but the effects of MO on myogenic cell migration remain to be examined. C57BL/6 mice embryos were collected at E13.5. The GeoMx DSP platform was used to customize five regions along myogenic cell migration routes (myotome, dorsal/ventral limb, limb stroma, limb tip), and data were analyzed by GeomxTools 3.6.0. A total of 2224 genes were down-regulated in the MO group. The GO enrichment analysis showed that MO inhibited migration-related biological processes. The signaling pathways guiding myogenic migration such as hepatocyte growth factor signaling, fibroblast growth factor signaling, Wnt signaling and GTPase signaling were down-regulated in the MO E13.5 limb tip. Correspondingly, the expression levels of genes involved in myogenic cell migration, such as Pax3, Gab1, Pxn, Tln2 and Arpc, were decreased in the MO group, especially in the dorsal and ventral sides of the limb. Additionally, myogenic differentiation-related genes were down-regulated in the MO limb. MO impedes myogenic cell migration and differentiation in the embryonic limb, providing an explanation for the impairment of fetal muscle development and offspring muscle function due to MO.


Asunto(s)
Diferenciación Celular , Movimiento Celular , Desarrollo de Músculos , Obesidad Materna , Animales , Movimiento Celular/genética , Ratones , Femenino , Desarrollo de Músculos/genética , Diferenciación Celular/genética , Embarazo , Obesidad Materna/metabolismo , Obesidad Materna/genética , Ratones Endogámicos C57BL , Regulación del Desarrollo de la Expresión Génica , Transcriptoma , Desarrollo Embrionario/genética , Extremidades/embriología , Perfilación de la Expresión Génica , Transducción de Señal , Músculo Esquelético/metabolismo , Músculo Esquelético/embriología
6.
Mol Metab ; 88: 102008, 2024 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-39142562

RESUMEN

OBJECTIVE: Maternal obesity is increasingly common and negatively impacts offspring health. Children of mothers with obesity are at higher risk of developing diseases linked to hematopoietic system abnormalities and metabolism such as type 2 diabetes. Interestingly, disease risks are often dependent on the offspring's sex, suggesting sex-specific reprogramming effect of maternal obesity on offspring hematopoietic stem and progenitor cell (HSPC) function. However, the impact of maternal obesity exposure on offspring HSPC function, and the capability of HSPC to regulate offspring metabolic health is largely understudied. This study aims to test the hypothesis that offspring of obese mice exhibit sex-differences in HSPC function that affect offspring's metabolic health. METHODS: We first assessed bone marrow hematopoietic stem and progenitor cell phenotype using postnatal day 21 (P21) and 8-week-old C57BL/6J mice born to control and diet-induced obese dams. We also sorted HSPC (Lineage-, Sca1+, cKit + cells) from P21 mice for competitive primary and secondary transplant, as well as transcriptomic analysis. Body weight, adiposity, insulin tolerance test and glucose tolerance tests were performed in primary and secondary transplant recipient animals. RESULTS: We discovered sex-differences in offspring HSPC function in response to maternal obesity exposure, where male offspring of obese dams (MatOb) showed decreased HSPC numbers and engraftment, while female MatOb offspring remained largely unaffected. RNA-seq revealed immune stimulatory pathways in female MatOb offspring. Finally, only recipients of male MatOb offspring HSPC exhibited glucose intolerance. CONCLUSIONS: This study demonstrated the lasting effect of maternal obesity exposure on offspring HSPC function and implicates HSPC in metabolic regulation.


Asunto(s)
Intolerancia a la Glucosa , Células Madre Hematopoyéticas , Ratones Endogámicos C57BL , Animales , Intolerancia a la Glucosa/metabolismo , Femenino , Ratones , Masculino , Células Madre Hematopoyéticas/metabolismo , Embarazo , Obesidad Materna/metabolismo , Ratones Obesos , Efectos Tardíos de la Exposición Prenatal/metabolismo , Obesidad/metabolismo , Dieta Alta en Grasa/efectos adversos
7.
Nutrients ; 16(16)2024 Aug 22.
Artículo en Inglés | MEDLINE | ID: mdl-39203941

RESUMEN

Both maternal obesity and postnatal consumption of obesogenic diets contribute to the development of metabolic dysfunction-associated steatotic liver disease (MASLD) and hepatocellular carcinoma (HCC). However, there is no consensus as to whether diets that are high in fat or carbohydrates/sugars differentially influence the development of HCC. Moreover, the long-term effects of prenatal HF exposure on HCC and whether this is influenced by postnatal diet has not yet been evaluated. C57BL/6 dams were fed either a low-fat, high-carbohydrate control (C) or low-carbohydrate, high-fat (HF) diet. At weaning, male and female offspring were fed the C or HF diet, generating four diet groups: C/C, C/HF, HF/C and HF/HF. Tissues were collected at 16 months of age and livers were assessed for MASLD and HCC. Glucose regulation and pancreatic morphology were also evaluated. Liver tissues were assessed for markers of glycolysis and fatty acid metabolism and validated using a human HCC bioinformatic database. Both C/HF and HF/HF mice developed obesity, hyperinsulinemia and a greater degree of MASLD than C/C and HF/C offspring. However, despite significant liver and pancreas pathology, C/HF mice had the lowest incidence of HCC while tumour burden was highest in HF/C male offspring. The molecular profile of HCC mouse samples suggested an upregulation of the pentose phosphate pathway and a downregulation of fatty acid synthesis and oxidation, which was largely validated in the human dataset. Both pre-weaning HF diet exposure and post-weaning consumption of a high-carbohydrate diet increased the risk of developing spontaneous HCC in aged mice. However, the influence of pre-weaning HF feeding on HCC development appeared to be stronger in the context of post-weaning obesity. As rates of maternal obesity continue to rise, this has implications for the future incidence of HCC and possible dietary manipulation of offspring carbohydrate intake to counteract this risk.


Asunto(s)
Carcinoma Hepatocelular , Dieta Alta en Grasa , Neoplasias Hepáticas , Ratones Endogámicos C57BL , Destete , Animales , Femenino , Dieta Alta en Grasa/efectos adversos , Carcinoma Hepatocelular/etiología , Carcinoma Hepatocelular/metabolismo , Neoplasias Hepáticas/etiología , Neoplasias Hepáticas/metabolismo , Embarazo , Masculino , Ratones , Efectos Tardíos de la Exposición Prenatal , Fenómenos Fisiologicos Nutricionales Maternos , Hígado/metabolismo , Hígado/patología , Obesidad , Hígado Graso/etiología , Carbohidratos de la Dieta/efectos adversos , Carbohidratos de la Dieta/administración & dosificación
8.
Int J Mol Sci ; 25(16)2024 Aug 21.
Artículo en Inglés | MEDLINE | ID: mdl-39201761

RESUMEN

Maternal obesity during pregnancy is associated with adverse pregnancy outcomes. This might be due to undesired obesity-induced changes in the maternal gut microbiota and related changes in the maternal immune adaptations during pregnancy. The current study examines how obesity affects gut microbiota and immunity in pregnant obese and lean mice during mid-pregnancy (gestational day 12 (GD12)). C57BL/6 mice were fed a high-fat diet or low-fat diet from 8 weeks before mating and during pregnancy. At GD12, we analyzed the gut microbiota composition in the feces and immune responses in the intestine (Peyer's patches, mesenteric lymph nodes) and the peripheral circulation (spleen and peripheral blood). Maternal obesity reduced beneficial bacteria (e.g., Bifidobacterium and Akkermansia) and changed intestinal and peripheral immune responses (e.g., dendritic cells, Th1/Th2/Th17/Treg axis, monocytes). Numerous correlations were found between obesity-associated bacterial genera and intestinal/peripheral immune anomalies. This study shows that maternal obesity impacts the abundance of specific bacterial gut genera as compared to lean mice and deranges maternal intestinal immune responses that subsequently change peripheral maternal immune responses in mid-pregnancy. Our findings underscore the opportunities for early intervention strategies targeting maternal obesity, ideally starting in the periconceptional period, to mitigate these obesity-related pregnancy effects.


Asunto(s)
Dieta Alta en Grasa , Microbioma Gastrointestinal , Ratones Endogámicos C57BL , Obesidad , Animales , Femenino , Embarazo , Microbioma Gastrointestinal/inmunología , Ratones , Dieta Alta en Grasa/efectos adversos , Obesidad/inmunología , Obesidad/microbiología , Obesidad/etiología , Obesidad Materna/inmunología
9.
BMC Womens Health ; 24(1): 467, 2024 Aug 24.
Artículo en Inglés | MEDLINE | ID: mdl-39182118

RESUMEN

BACKGROUND: Gestational diabetes mellitus (GDM) is a common pregnancy complication with long-term health consequences for mothers and their children. The escalating trends of GDM coupled with the growing prevalence of maternal obesity, a significant GDM risk factor projected to approach nearly 60% by 2030 in Kansas, has emerged as a pressing public health issue. METHODS: The aim of this study was to compare GDM and maternal obesity trends in rural and urban areas and investigate maternal demographic characteristics influencing the risk of GDM development over a 15-year period. Trend analyses and a binary logistic regression were employed utilizing 2005 to 2019 de-identified birth record vital statistics from the Kansas Department of Health and Environment (N = 589,605). RESULTS: Over the cumulative 15-year period, a higher prevalence of GDM was observed across age, race/ethnicity, education, and insurance source. Throughout this period, there was an increasing trend in both GDM and obese pre-pregnancy BMI age-adjusted prevalence, with noticeable rural-urban disparities. From 2005 to 2019, women, including Asians (OR: 2.73, 95% CI 2.58%-2.88%), American Indian or Alaskan Natives (OR: 1.58, 95%, CI 1.44-1.73%), Hispanics (OR: 1.42, 95% CI 1.37%-1.48%), women residing in rural areas (OR: 1.09, 95%, CI 1.06-1.12%), with advanced maternal age (35-39 years, OR: 4.83 95% CI 4.47%-5.22%; ≥40 years, OR: 6.36 95%, CI 5.80-6.98%), with lower educational status (less than high school, OR: 1.15, 95% CI 1.10%-1.20%; high school graduate, OR: 1.10, 95% CI 1.06%-1.13%), Medicaid users (OR: 1.10, 95% CI 1.06%-1.13%), or with an overweight (OR: 1.78, 95% CI 1.72%-1.84%) or obese (OR: 3.61, 95% CI 3.50%-3.72%) pre-pregnancy BMI were found to be at an increased risk of developing GDM. CONCLUSIONS: There are persistent rural-urban and racial/ethnic disparities present from 2005 to 2019 among pregnant women in Kansas with or at-risk of GDM. There are several socioeconomic factors that contribute to these health disparities affecting GDM development. These findings, alongside with prominent rising maternal obesity trends, highlight the need to expand GDM services in a predominantly rural state, and implement culturally-responsive interventions for at-risk women.


Asunto(s)
Diabetes Gestacional , Población Rural , Determinantes Sociales de la Salud , Población Urbana , Adolescente , Adulto , Femenino , Humanos , Embarazo , Adulto Joven , Diabetes Gestacional/epidemiología , Etnicidad/estadística & datos numéricos , Kansas/epidemiología , Obesidad Materna/epidemiología , Obesidad Materna/complicaciones , Prevalencia , Factores de Riesgo , Población Rural/estadística & datos numéricos , Determinantes Sociales de la Salud/estadística & datos numéricos , Población Urbana/estadística & datos numéricos , Grupos Raciales/estadística & datos numéricos
10.
Curr Diab Rep ; 24(10): 227-235, 2024 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-39162956

RESUMEN

PURPOSE OF REVIEW: Entering pregnancy with obesity increases the risk of adverse health outcomes for parent and child. As such, research interventions are largely focused on limiting excess gestational weight gain during pregnancy, especially in those with obesity. Yet, while many lifestyle interventions are successful in reducing GWG, few affect pregnancy outcomes. Here we review work targeting the metabolic milieu instead of focusing solely on weight. RECENT FINDINGS: Work done in non-pregnant populations suggests that specifically targeting glucose, triglyceride, and leptin levels or inflammatory makers improves the metabolic milieu and overall health. We posit that precision interventions that include strategies such as time restricted eating, following the 24 h movement guidelines, or reducing sedentary behavior during pregnancy can be successful approaches benefiting the maternal metabolic milieu and minimize the risk of adverse pregnancy outcomes. Personalized tools such as continuous glucose monitors or community-based approaches play an important role in pre-conception health and should be extrapolated to pregnancy interventions to directly benefit the metabolic milieu optimizing health outcomes for both parent and child.


Asunto(s)
Obesidad , Complicaciones del Embarazo , Humanos , Femenino , Embarazo , Obesidad/metabolismo , Obesidad/terapia , Resultado del Embarazo , Ganancia de Peso Gestacional/fisiología , Medicina de Precisión/métodos
11.
Cureus ; 16(7): e64867, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-39156241

RESUMEN

INTRODUCTION:  Maternal overweight and obesity during pregnancy have been shown to have multiple negative effects on the mother's health, which can even affect the infant's growth by increasing weight gain and altering various indicators, such as weight for age, length for age and weight for length. While breast milk on the other hand reduces these risks, and it's the best and most complete food for the newborn. It's a dynamic fluid capable of being modified to meet the needs of each stage of the newborn, but despite this capacity and the fact that maternal body mass index can have an impact on its components, through complex biological mechanisms, it manages to reduce the negative effects accumulated during pregnancy and even promotes a healthy state in the baby. In a country like Mexico, where overweight and obesity affect a large part of the population, it is important to study their causes and which could be the effect of this increased maternal overweight during pregnancy and lactation on newborns. OBJECTIVE: Identify the alterations associated with increased maternal body mass index during pregnancy and breastfeeding on mothers' health and their possible effect on the growth of the newborn during the first six months of life. MATERIAL AND METHODS: This was a prospective cohort study. Forty-two healthy binomials (mother and child), without problems during delivery and without serious illnesses during the breastfeeding period, were included. Maternal body mass index at the beginning of pregnancy allowed us to create two comparison groups between mothers: one with adequate weight, another with overweight or obesity. Follow-up was carried out once a month during the first six months of life, evaluating the somatometric development of mothers and children. All mothers completed the six-month period of exclusive breastfeeding. RESULTS:  There were differences between both groups of women. The one that included overweight and obese women compared to the group of women with adequate weight had a higher number of pregnancies, abortions, plasma glucose levels in the third trimester of pregnancy, and a lower number of prenatal control visits and plasma platelet levels (all with p<0.05). Regarding the baby's growth, there was a difference between the weight for length classification at 60-, 120-, 150- and 180-day follow-ups. The group to which the mother was assigned with respect to her body mass index at the beginning of pregnancy (adequate weight group and overweight/obese group) was the only factor associated with the risk of the baby being overweight according to weight for length indicator at the 180-day follow-up, with an OR = 5.2 (95%CI 1.02-26.59). CONCLUSIONS: Maternal overweight and obesity during pregnancy have a negative effect on the mother's health and baby's weight gain in its weight-for-length classification during the first six months of life. Although breastfeeding has been shown to have a positive effect on the growth of the baby, exposure to a higher maternal body mass index during pregnancy triggers important metabolic alterations that promote the development of diseases. It is important to establish weight control guidelines in women who wish to become pregnant to reduce the negative effects on the mother and offspring.

12.
J Obstet Gynaecol Can ; 46(10): 102643, 2024 Aug 28.
Artículo en Inglés | MEDLINE | ID: mdl-39214249

RESUMEN

OBJECTIVES: To examine the relation between maternal pre-delivery BMI and the accuracy of sonographic estimated fetal weight (EFW) in very preterm infants (<32 weeks gestation). METHODS: This retrospective study included singleton infants born between January 2010 and March 2023, at gestational ages 230 to 316 weeks, at a tertiary university-affiliated hospital. Absolute weight, percentage error, absolute percentage error, and overestimation and underestimation of EFW were compared between women with pre-delivery normal weight (BMI 18.5-24.99 kg/m2), overweight (BMI 25.0-29.99 kg/m2), and obesity (BMI >35.0 kg/m2). Multivariate linear regression analyses adjusted for potential confounders were performed to assess relations of maternal pre-conception and of pre-delivery BMI, with EFW accuracy. RESULTS: Included were 286 pregnancies. The absolute difference, percentage error, absolute percentage error, error within the 10% range, and underestimation or overestimation of EFW were similar between the groups. The multivariate linear regression analyses did not show significant associations of pre-conceptional BMI or of pre-delivery BMI with the percentage error. However, for small for gestational age compared to appropriate for gestational age fetuses, the percentage error was greater (8.9% vs. -0.6%, ß = 0.35, P < 0.001) and the absolute percentage error was greater (11.0% vs. 6.7%, P < 0.001). Small for gestational age fetuses were at risk of fetal weight overestimation (percentage error exceeding 15%); OR 7.20 (95% CI 2.91-17.80). CONCLUSIONS: Maternal pre-delivery BMI was not found to be related to EFW accuracy in very preterm infants. Nevertheless, EFW should be interpreted carefully, as it may underdiagnose poor fetal growth in this population.

13.
J Physiol ; 602(17): 4251-4269, 2024 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-39087821

RESUMEN

The consumption of high fat-high energy diets (HF-HEDs) continues to rise worldwide and parallels the rise in maternal obesity (MO) that predisposes offspring to cardiometabolic disorders. Although the underlying mechanisms are unclear, thyroid hormones (TH) modulate cardiac maturation in utero. Therefore, we aimed to determine the impact of a high fat-high energy diet (HF-HED) on the hormonal, metabolic and contractility profile of the non-human primate (NHP) fetal heart. At ∼9 months preconception, female baboons (Papio hamadryas) were randomly assigned to either a control diet or HF-HED. At 165 days gestational age (term = 184 days), fetuses were delivered by Caesarean section under anaesthesia, humanely killed, and left ventricular cardiac tissue (Control (n = 6 female, 6 male); HF-HED (n = 6 F, 6 M)) was collected. Maternal HF-HED decreased the concentration of active cardiac TH (i.e. triiodothyronine (T3)), and type 1 iodothyronine deiodinase (DIO1) mRNA expression. Maternal HF-HED decreased the abundance of cardiac markers of insulin-mediated glucose uptake phosphorylated insulin receptor substrate 1 (Ser789) and glucose transporter 4, and increased protein abundance of key oxidative phosphorylation complexes (I, III, IV) and mitochondrial abundance in both sexes. Maternal HF-HED alters cardiac TH status, which may induce early signs of cardiac insulin resistance. This may increase the risk of cardiometabolic disorders in later life in offspring born to these pregnancies. KEY POINTS: Babies born to mothers who consume a high fat-high energy diet (HF-HED) prior to and during pregnancy are predisposed to an increased risk of cardiometabolic disorders across the life course. Maternal HF-HED prior to and during pregnancy decreased thyroid hormone triiodothyronine (T3) concentrations and type 1 iodothyronine deiodinase DIO1 mRNA expression in the non-human primate fetal heart. Maternal HF-HED decreased markers of insulin-dependent glucose uptake, phosphorylated insulin receptor substrate 1 and glucose transporter 4 in the fetal heart. Maternal HF-HED increased mitochondrial abundance and mitochondrial OXPHOS complex I, III and IV in the fetal heart. Fetuses from HF-HED pregnancies are predisposed to cardiometabolic disorders that may be mediated by changes in T3, placing them on a poor lifetime cardiovascular health trajectory.


Asunto(s)
Dieta Alta en Grasa , Corazón Fetal , Animales , Femenino , Embarazo , Dieta Alta en Grasa/efectos adversos , Corazón Fetal/metabolismo , Masculino , Hormonas Tiroideas/metabolismo , Hormonas Tiroideas/sangre , Fenómenos Fisiologicos Nutricionales Maternos , Papio hamadryas/metabolismo
14.
Brain Behav Immun ; 121: 317-330, 2024 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-39032541

RESUMEN

The developing central nervous system is highly sensitive to nutrient changes during the perinatal period, emphasising the potential impact of alterations of maternal diet on offspring brain development and behaviour. A growing body of research implicates the gut microbiota in neurodevelopment and behaviour. Maternal overweight and obesity during the perinatal period has been linked to changes in neurodevelopment, plasticity and affective disorders in the offspring, with implications for microbial signals from the maternal gut. Here we investigate the impact of maternal high-fat diet (mHFD)-induced changes in microbial signals on offspring brain development, and neuroimmune signals, and the enduring effects on behaviour into adolescence. We first demonstrate that maternal caecal microbiota composition at term pregnancy (embryonic day 18: E18) differs significantly in response to maternal diet. Moreover, mHFD resulted in the upregulation of microbial genes in the maternal intestinal tissue linked to alterations in quinolinic acid synthesis and elevated kynurenine levels in the maternal plasma, both neuronal plasticity mediators related to glutamate metabolism. Metabolomics of mHFD embryonic brains at E18 also detected molecules linked to glutamate-glutamine cycle, including glutamic acid, glutathione disulphide, and kynurenine. During adolescence, the mHFD offspring exhibited increased locomotor activity and anxiety-like behaviour in a sex-dependent manner, along with upregulation of glutamate-related genes compared to controls. Overall, our results demonstrate that maternal exposure to high-fat diet results in microbiota changes, behavioural imprinting, altered brain metabolism, and glutamate signalling during critical developmental windows during the perinatal period.


Asunto(s)
Encéfalo , Dieta Alta en Grasa , Microbioma Gastrointestinal , Efectos Tardíos de la Exposición Prenatal , Animales , Dieta Alta en Grasa/efectos adversos , Femenino , Embarazo , Encéfalo/metabolismo , Microbioma Gastrointestinal/fisiología , Efectos Tardíos de la Exposición Prenatal/metabolismo , Efectos Tardíos de la Exposición Prenatal/microbiología , Masculino , Conducta Animal/fisiología , Fenómenos Fisiologicos Nutricionales Maternos , Conducta del Adolescente/fisiología , Ratones , Ansiedad/metabolismo , Ansiedad/microbiología
15.
J Neurosci Res ; 102(7): e25368, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-39007363

RESUMEN

Maternal obesity before or during pregnancy has been associated previously in offspring with a wide range of poor neurodevelopmental outcomes and mental health problems. The effects of maternal obesity on offspring brain structure and function that may be responsible for these poor outcomes are not well understood. We, therefore, undertook a systematic review of magnetic resonance imaging (MRI) studies that have assessed the associations of maternal obesity with brain measures in offspring. A systematic search was conducted in PubMed, Web of Science, Scopus, and PsycINFO on August 20, 2023. Of 15 eligible studies, seven employed functional MRI (fMRI), five diffusion tensor imaging (DTI), and four anatomical MRI (one used both DTI and anatomical MRI) in the offspring. The ages of offspring varied widely: one was a study of fetuses in utero, five of neonates, one of infants, five of school-aged children, two of both neonates and infants, and one of both children and adults. Collectively, 12 studies reported significant associations of maternal obesity with structural or functional alterations of the offspring's brain, most frequently in the prefrontal cortex and limbic system. In conclusion, maternal obesity appears to have a profound influence on offspring brain development, particularly within the prefrontal and limbic networks that regulate emotion and behavior. Further studies are needed to identify how changes in brain structure and function mediate the effects of maternal obesity on long-term emotional and behavioral outcomes, as well as the molecular pathways through which maternal obesity alters offspring brain development.


Asunto(s)
Encéfalo , Imagen por Resonancia Magnética , Obesidad Materna , Niño , Femenino , Humanos , Lactante , Recién Nacido , Embarazo , Encéfalo/diagnóstico por imagen , Encéfalo/crecimiento & desarrollo , Encéfalo/patología , Efectos Tardíos de la Exposición Prenatal/fisiopatología , Efectos Tardíos de la Exposición Prenatal/diagnóstico por imagen , Efectos Tardíos de la Exposición Prenatal/patología
16.
Am J Epidemiol ; 2024 Jul 16.
Artículo en Inglés | MEDLINE | ID: mdl-39013792

RESUMEN

The objectives of this study were to examine the total effect of grandmaternal [G0] pre-pregnancy body mass index (BMI) on infant [G2] birthweight z-score and to quantify the mediation role of maternal [G1] pre-pregnancy BMI. Data were extracted from the Nova Scotia 3G Multigenerational Cohort. The association between G0 pre-pregnancy BMI and G2 birthweight z-score and the mediated effect by G1 pre-pregnancy BMI were estimated using g-computation with adjustment for confounders identified using a directed acyclic graph and accounting for intermediate confounding. 20822 G1-G2 dyads from 18450 G0 were included. Relative to G0 normal weight, G0 underweight decreased mean G2 birthweight z-score (-0.11, 95% confidence interval (CI) -0.20, -0.030), while G0 overweight and obesity increased mean G2 birthweight z-score (0.091 [95% CI 0.034, 0.15] and 0.22 [95% CI 0.11, 0.33]). G1 pre-pregnancy BMI partly mediated the association, with the largest effect size observed for G0 obesity (0.11, 95% CI 0.080, 0.14). Estimates of the direct effect were close to the null. In conclusion, grandmaternal pre-pregnancy BMI was associated with infant birthweight z-score. Maternal pre-pregnancy BMI partly mediated the association, suggesting that factors related to BMI may play an important role in the transmission of weight across the maternal line.

17.
Front Immunol ; 15: 1376583, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39072322

RESUMEN

Introduction: Maternal obesity poses risks for both mother and offspring during pregnancy, with underlying mechanisms remaining largely unexplored. Obesity is associated with microbial gut dysbiosis and low-grade inflammation, and also the diet has a major impact on these parameters. This study aimed to investigate how maternal obesity and diet contribute to changes in immune responses, exploring potential associations with gut microbiota dysbiosis and adverse pregnancy outcomes in mice. Methods: Before mating, C57BL/6 mice were assigned to either a high-fat-diet (HFD) or low-fat-diet (LFD) to obtain obese (n=17) and lean (n=10) mice. To distinguish between the effects of obesity and diet, 7 obese mice were switched from the HFD to the LFD from day 7 until day 18 of pregnancy ("switch group"), which was the endpoint of the study. T helper (Th) cell subsets were studied in the spleen, mesenteric lymph nodes (MLN) and Peyer's patches (PP), while monocyte subsets and activation status were determined in maternal blood (flow cytometry). Feces were collected before and during pregnancy (day 7,14,18) for microbiota analysis (16S rRNA sequencing). Pregnancy outcome included determination of fetal and placental weight. Results: Obesity increased splenic Th1 and regulatory T cells, MLN Th1 and PP Th17 cells and enhanced IFN-γ and IL-17A production by splenic Th cells upon ex vivo stimulation. Switching diet decreased splenic and PP Th2 cells and classical monocytes, increased intermediate monocytes and activation of intermediate/nonclassical monocytes. Obesity and diet independently induced changes in the gut microbiota. Various bacterial genera were increased or decreased by obesity or the diet switch. These changes correlated with the immunological changes. Fetal weight was lower in the obese than the lean group, while placental weight was lower in the switch than the obese group. Discussion: This study demonstrates that obesity and diet independently impact peripheral and intestinal immune responses at the end of pregnancy. Simultaneously, both factors affect specific bacterial gut genera and lead to reduced fetal or placental weight. Our data suggest that switching diet during pregnancy to improve maternal health is not advisable and it supports pre/probiotic treatment of maternal obesity-induced gut dysbiosis to improve maternal immune responses and pregnancy outcome.


Asunto(s)
Dieta Alta en Grasa , Microbioma Gastrointestinal , Ratones Endogámicos C57BL , Obesidad , Resultado del Embarazo , Animales , Embarazo , Femenino , Microbioma Gastrointestinal/inmunología , Ratones , Dieta Alta en Grasa/efectos adversos , Obesidad/inmunología , Obesidad/microbiología , Disbiosis/inmunología , Obesidad Materna/inmunología
18.
AJOG Glob Rep ; 4(3): 100361, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-39072339

RESUMEN

BACKGROUND: Preterm birth from intrauterine infection is a leading cause of neonatal neurologic morbidity. Likewise, maternal obesity is associated with intra-amniotic infection and inflammation. Whether maternal obesity is a risk factor for fetal brain injury that occurs with premature birth remains unknown. This study hypothesized that maternal obesity intensifies fetal neuroinflammation in the setting of premature delivery. OBJECTIVE: This study aimed to examine the influence of maternal obesity on perinatal neuroinflammatory responses that arise with preterm birth using a murine model. STUDY DESIGN: Dams with obesity were generated via a high-fat diet that was maintained throughout pregnancy. In parallel, dams without obesity (normal) received a control diet. All dams were paired with males on normal diet. Pregnant dams were randomized to receive an intrauterine administration of bacterial endotoxin (lipopolysaccharide) or the vehicle (phosphate-buffered saline) on embryo day 15.5 of what is typically a 19- to 21-day gestation. Fetal brains were harvested 6 hours after intrauterine administrations, and the expressions of key inflammatory cytokines (Il1b, Il6, and Tnf) and panels of metabolic, immune, and inflammatory genes were analyzed. RESULTS: With the phosphate-buffered saline, there was no difference in gene expression related to maternal obesity. There were substantial differences in Il6 and immune/inflammatory expression profiles in fetal brains from dams with obesity vs normal dams that received lipopolysaccharide. Few differences were observed among the metabolic genes examined under these conditions. The gene expression pattern associated with maternal obesity correlated with pathways related to white matter injury. CONCLUSION: The expression of neuroinflammatory markers instigated by bacterial endotoxin via intrauterine lipopolysaccharide was greater in embryo brains obtained from dams with obesity. Expression profiles suggest that in combination with intrauterine inflammation, maternal obesity may increase the risk of fetal white matter injury. Further investigation is warranted to understand the relationship between maternal health and neurologic outcomes associated with prematurity.

19.
Nutrients ; 16(13)2024 Jul 08.
Artículo en Inglés | MEDLINE | ID: mdl-38999919

RESUMEN

This study examines relationships between breastfeeding practices and postpartum weight retention (PPWR) at 6 and 12 months postpartum among 379 first-time mothers participating in a clinical trial in Singapore. We categorized feeding modes at 6 months into exclusive breastfeeding, mixed feeding, and exclusive formula feeding. Participants were analyzed in two groups based on their PPWR assessment at 6 and 12 months postpartum, with complete datasets available for each assessment. We calculated PPWR by subtracting pre-pregnancy weight from self-reported weight at 6 and 12 months postpartum, defining substantial PPWR as ≥5 kg retention. Modified Poisson regression models adjusted for potential confounders were performed. At 6 and 12 months, 35% (n = 132/379) and 31% (n = 109/347) of women experienced substantial PPWR, respectively. Compared to exclusive breastfeeding, mixed feeding (risk ratio 1.85; 95% confidence interval 1.15, 2.99) and exclusive formula feeding (2.11; 1.32, 3.28) were associated with a higher risk of substantial PPWR at 6 months. These associations were slightly attenuated at 12 months and appeared stronger in women with pre-pregnancy overweight or obesity. This study suggests that breastfeeding by 6 months postpartum may help mitigate PPWR, particularly with exclusive breastfeeding. It also draws attention to targeted interventions to promote breastfeeding among women with overweight or obesity.


Asunto(s)
Lactancia Materna , Periodo Posparto , Humanos , Lactancia Materna/estadística & datos numéricos , Femenino , Adulto , Singapur , Embarazo , Estudios de Cohortes , Pueblo Asiatico , Sobrepeso , Peso Corporal , Adulto Joven , Obesidad , Aumento de Peso , Índice de Masa Corporal
20.
World J Clin Pediatr ; 13(2): 91255, 2024 Jun 09.
Artículo en Inglés | MEDLINE | ID: mdl-38947991

RESUMEN

BACKGROUND: Childhood obesity is a growing global concern with far-reaching health implications. This study focuses on evaluating the knowledge and practices of physicians in Morocco regarding the link between maternal obesity and childhood obesity. Despite the increasing prevalence of childhood obesity worldwide, this issue remains inadequately addressed in the Moroccan context. AIM: To assess the awareness and practices of physicians in Morocco concerning the connection between maternal obesity and childhood obesity. METHODS: The research encompasses a comprehensive survey of practicing physicians, revealing significant gaps in awareness and practices related to maternal obesity. RESULTS: Notably, a significant portion of doctors do not provide adequate guidance to overweight pregnant women, highlighting the urgency for targeted educational programs. CONCLUSION: In conclusion, this research illuminates critical areas for improvement in tackling childhood obesity in Morocco. By addressing these gaps, fostering awareness, and enhancing medical practices, the healthcare system can contribute significantly to preventing childhood obesity and improving the overall health of future generations.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA