Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
1.
Nutrients ; 15(19)2023 Sep 25.
Article in English | MEDLINE | ID: mdl-37836424

ABSTRACT

BACKGROUND: Gestational diabetes is associated with increased risk of obesity, type 2 diabetes and cardiovascular disease. Effective nutritional strategies are needed to reduce BMI and improve long-term maternal cardiometabolic health, but the relative contribution of maternal eating behaviour, a potential barrier to dietary change, has not been explored. We compared eating behaviour in women with gestational diabetes with that of men and non-pregnant women with comparable risk factors, and tested associations between eating behaviour traits and BMI in women with gestational diabetes. We hypothesized that eating behaviour would be unfavourable in gestational diabetes and would be associated with BMI. METHODS: Participants (n = 417) including 53 men, 164 non-pregnant women and 200 women with gestational diabetes (singleton pregnancy; 29 weeks' gestation) were recruited into three prospective studies assessing weight loss interventions, with similar entry criteria. The three-factor eating questionnaire (TFEQ-R18) assessed uncontrolled eating, emotional eating and cognitive restraint at study enrolment. Associations between BMI at study enrolment and TFEQ-R18 (% maximum score) were assessed using linear regression. RESULTS: Women with gestational diabetes had significantly lower uncontrolled eating scores vs. men (53% vs. 65%; p < 0.001) and non-pregnant women (53% vs. 66%; p < 0.001), lower emotional eating scores vs. non-pregnant women (60% vs. 71%; p < 0.001) and higher cognitive restraint (p < 0.001 vs. men and non-pregnant women). In women with gestational diabetes, emotional eating scores were positively associated with BMI at study enrolment (beta coefficient 7.8 (95% CI 3.9 to 11.7), p < 0.001). CONCLUSIONS: Women with gestational diabetes have favourable eating behaviour compared with other population groups. Because BMI at study enrolment was associated with emotional eating, nutritional strategies which reduce emotional eating may provide new opportunities to improve long-term maternal health after gestational diabetes.


Subject(s)
Diabetes Mellitus, Type 2 , Diabetes, Gestational , Pregnancy , Male , Humans , Adult , Female , Diabetes, Gestational/epidemiology , Diabetes Mellitus, Type 2/complications , Prospective Studies , Obesity/epidemiology , Feeding Behavior/psychology , Body Mass Index
2.
PLoS One ; 18(9): e0292094, 2023.
Article in English | MEDLINE | ID: mdl-37756288

ABSTRACT

BACKGROUND: Undiagnosed diabetes in pregnancy is associated with stillbirth and perinatal complications, but standard testing for gestational diabetes using the oral glucose tolerance test (OGTT) is impractical and exacerbates healthcare inequalities. There is an urgent need to improve the accuracy, acceptability and accessibility of glucose testing in pregnancy. We qualitatively assessed the feasibility and acceptability of two alternative home-based methods of glucose testing in pregnant women, using continuous glucose monitoring (CGM), with or without a home-based OGTT. METHODS: We recruited women with a singleton pregnancy at 28 weeks' gestation with ≥1 risk factor for gestational diabetes attending antenatal glucose testing. A Dexcom G6 CGM device was sited and women were asked to take a 75g OGTT solution (Rapilose) on day 4 after an overnight fast. Qualitative interviews were performed with 20 participants using video conferencing according to a semi-structured interview schedule and thematically analysed using NVIVO software. RESULTS: 92 women were recruited; 73 also underwent a home OGTT. Women had an average of 6.9 days of glucose monitoring and found the CGM painless, easy to use with few or no adverse events. During the qualitative study, the main themes identified were reassurance and convenience. All women interviewed would recommend CGM and a home OGTT for diagnosis of gestational diabetes. CONCLUSIONS: CGM with or without a home OGTT is feasible and acceptable to pregnant women for diagnosis of gestational diabetes and offered advantages of convenience and reassurance. Further work is needed to clarify diagnostic thresholds for gestational diabetes using CGM metrics.


Subject(s)
Diabetes, Gestational , Pregnancy , Female , Humans , Diabetes, Gestational/diagnosis , Blood Glucose Self-Monitoring , Feasibility Studies , Prospective Studies , Blood Glucose , Glucose
3.
Acta Diabetol ; 60(12): 1635-1642, 2023 Dec.
Article in English | MEDLINE | ID: mdl-37439859

ABSTRACT

AIMS: Incretin hormones glucagon-like peptide 1 (GLP-1) and gastric inhibitory peptide (GIP) cause increased insulin secretion in non-pregnant adults, but their role in pregnancy, where there are additional metabolically-active hormones from the placenta, is less clear. The aim of the present study was to assess if fasting and post-load incretin concentrations were predictive of pregnancy insulin and glucose concentrations. METHODS: Pregnant women (n = 394) with one or more risk factors for gestational diabetes were recruited at 28 weeks for a 75 g oral glucose tolerance test (OGTT). Glucose, insulin, GLP-1 and GIP were measured in the fasting state and 120 min after glucose ingestion. RESULTS: Fasting plasma GLP-1 concentrations were associated with plasma insulin (standardised ß' 0.393 (0.289-0.498), p = 1.3 × 10-12; n = 306), but not with glucose concentrations (p = 0.3). The association with insulin was still evident when adjusting for BMI (ß' 0.271 (0.180-0.362), p = 1.1 × 10-8; n = 297). Likewise, at 120 min the OGTT GLP-1 concentrations were associated with plasma insulin concentrations (ß' 0.216 (0.100-0.331), p = 2.7 × 10-4; n = 306) even after adjusting for BMI (ß' 0.178 (0.061-0.294), p = 2.9 × 10-3; n = 296), but not with glucose (p = 0.9). GIP concentrations were not associated with insulin or glucose concentrations at either time point (all p > 0.2). In pregnancy plasma GLP-1, but not GIP, concentrations appear to be predictive of circulating insulin concentrations, independently of associations with BMIs. CONCLUSIONS: These results suggest that the relationship between insulin and incretins is preserved in pregnancy, but that other factors, such as placental hormones or counter-regulatory hormones, may be more important determinants of glycaemia and gestational diabetes aetiology.


Subject(s)
Diabetes Mellitus, Type 2 , Diabetes, Gestational , Adult , Female , Humans , Pregnancy , Insulin , Glucagon-Like Peptide 1 , Incretins , Blood Glucose , Placenta , Glucose , Gastric Inhibitory Polypeptide
5.
Int J Mol Sci ; 23(19)2022 Oct 01.
Article in English | MEDLINE | ID: mdl-36232961

ABSTRACT

Male fertility, as manifest by the quantity and progressive motility of spermatozoa, is negatively impacted by obesity, dyslipidaemia and metabolic disease. However, the relative distribution of lipids in spermatozoa and the two compartments which supply lipids for spermatogenesis (seminal fluid and blood serum) has not been studied. We hypothesised that altered availability of lipids in blood serum and seminal fluid may affect the lipid composition and progressive motility of sperm. 60 men of age 35 years (median (range 20-45) and BMI 30.4 kg/m2 (24-36.5) under preliminary investigation for subfertility were recruited at an NHS clinic. Men provided samples of serum and semen, subject to strict acceptance criteria, for analysis of spermatozoa count and motility. Blood serum (n = 60), spermatozoa (n = 26) and seminal fluid (n = 60) were frozen for batch lipidomics analysis. Spermatozoa and seminal fluid had comparable lipid composition but showed marked differences with the serum lipidome. Spermatozoa demonstrated high abundance of ceramides, very-long-chain fatty acids (C20-22), and certain phospholipids (sphingomyelins, plasmalogens, phosphatidylethanolamines) with low abundance of phosphatidylcholines, cholesterol and triglycerides. Men with spermatozoa of low progressive motility had evidence of fewer concentration gradients for many lipid species between blood serum and spermatozoa compartments. Spermatozoa are abundant in multiple lipid species which are likely to contribute to key cellular functions. Lipid metabolism shows reduced regulation between compartments in men with spermatozoa with reduced progressive motility.


Subject(s)
Semen , Sperm Motility , Adult , Ceramides/metabolism , Cholesterol/metabolism , Fatty Acids/metabolism , Humans , Male , Middle Aged , Phosphatidylcholines/metabolism , Phosphatidylethanolamines/metabolism , Plasmalogens , Semen/metabolism , Sperm Count , Sperm Motility/physiology , Spermatozoa/metabolism , Sphingomyelins/metabolism , Triglycerides/metabolism , Young Adult
6.
Nutrients ; 13(4)2021 Mar 31.
Article in English | MEDLINE | ID: mdl-33807319

ABSTRACT

Childhood obesity is an area of intense concern internationally and is influenced by events during antenatal and postnatal life. Although pregnancy complications, such as gestational diabetes and large-for-gestational-age birthweight have been associated with increased obesity risk in offspring, very few successful interventions in pregnancy have been identified. We describe a study protocol to identify if a reduced calorie diet in pregnancy can reduce adiposity in children to 3 years of age. The dietary intervention in gestational diabetes (DiGest) study is a randomised, controlled trial of a reduced calorie diet provided by a whole-diet replacement in pregnant women with gestational diabetes. Women receive a weekly dietbox intervention from enrolment until delivery and are blinded to calorie allocation. This follow-up study will assess associations between a reduced calorie diet in pregnancy with offspring adiposity and maternal weight and glycaemia. Anthropometry will be performed in infants and mothers at 3 months, 1, 2 and 3 years post-birth. Glycaemia will be assessed using bloodspot C-peptide in infants and continuous glucose monitoring with HbA1c in mothers. Data regarding maternal glycaemia in pregnancy, maternal nutrition, infant birthweight, offspring feeding behaviour and milk composition will also be collected. The DiGest follow-up study is expected to take 5 years, with recruitment finishing in 2026.


Subject(s)
Clinical Protocols , Pediatric Obesity , Adult , Blood Glucose , Body Weight , Child , Diabetes, Gestational , Female , Follow-Up Studies , Humans , Life Style , Maternal Nutritional Physiological Phenomena , Pregnancy , Prenatal Care , Risk Factors
7.
BMC Pregnancy Childbirth ; 21(1): 96, 2021 Jan 29.
Article in English | MEDLINE | ID: mdl-33514342

ABSTRACT

BACKGROUND: Offspring of women with type 1 diabetes are at increased risk of fetal growth patterns which are associated with perinatal morbidity. Our aim was to compare rates of large- and small-for-gestational age (LGA; SGA) defined according to different criteria, using data from the Continuous Glucose Monitoring in Type 1 Diabetes Pregnancy Trial (CONCEPTT). METHODS: This was a pre-specified analysis of CONCEPTT involving 225 pregnant women and liveborn infants from 31 international centres ( ClinicalTrials.gov NCT01788527; registered 11/2/2013). Infants were weighed immediately at birth and GROW, INTERGROWTH and WHO centiles were calculated. Relative risk ratios, sensitivity and specificity were used to assess the different growth standards with respect to perinatal outcomes, including neonatal hypoglycaemia, hyperbilirubinaemia, respiratory distress, neonatal intensive care unit (NICU) admission and a composite neonatal outcome. RESULTS: Accelerated fetal growth was common, with mean birthweight percentiles of 82.1, 85.7 and 63.9 and LGA rates of 62, 67 and 30% using GROW, INTERGROWTH and WHO standards respectively. Corresponding rates of SGA were 2.2, 1.3 and 8.9% respectively. LGA defined according to GROW centiles showed stronger associations with preterm delivery, neonatal hypoglycaemia, hyperbilirubinaemia and NICU admission. Infants born > 97.7th centile were at highest risk of complications. SGA defined according to INTERGROWTH centiles showed slightly stronger associations with perinatal outcomes. CONCLUSIONS: GROW and INTERGROWTH standards performed similarly and identified similar numbers of neonates with LGA and SGA. GROW-defined LGA and INTERGROWTH-defined SGA had slightly stronger associations with neonatal complications. WHO standards underestimated size in preterm infants and are less applicable for use in type 1 diabetes. TRIAL REGISTRATION: This trial is registered with ClinicalTrials.gov . number NCT01788527 . Trial registered 11/2/2013.


Subject(s)
Diabetes Mellitus, Type 1/physiopathology , Fetal Growth Retardation/etiology , Fetal Macrosomia/etiology , Growth Charts , Neonatology/standards , Adult , Birth Weight , Diabetes Mellitus, Type 1/therapy , Female , Fetal Development , Fetal Growth Retardation/epidemiology , Fetal Macrosomia/epidemiology , Gestational Age , Humans , Infant, Newborn , Infant, Small for Gestational Age/growth & development , Logistic Models , Male , Pregnancy , Premature Birth , United Kingdom
8.
Diabetologia ; 64(4): 890-902, 2021 04.
Article in English | MEDLINE | ID: mdl-33501603

ABSTRACT

AIMS/HYPOTHESIS: Levels of the microRNA (miRNA) miR-126-3p are programmed cell-autonomously in visceral adipose tissue of adult offspring born to obese female C57BL/6J mice. The spectrum of miR-126-3p targets and thus the consequences of its dysregulation for adipocyte metabolism are unknown. Therefore, the aim of the current study was to identify novel targets of miR-126-3p in vitro and then establish the outcomes of their dysregulation on adipocyte metabolism in vivo using a well-established maternal obesity mouse model. METHODS: miR-126-3p overexpression in 3T3-L1 pre-adipocytes followed by pulsed stable isotope labelling by amino acids in culture (pSILAC) was performed to identify novel targets of the miRNA. Well-established bioinformatics algorithms and luciferase assays were then employed to confirm those that were direct targets of miR-126-3p. Selected knockdown experiments were performed in vitro to define the consequences of target dysregulation. Quantitative real-time PCR, immunoblotting, histology, euglycaemic-hyperinsulinaemic clamps and glucose tolerance tests were performed to determine the phenotypic and functional outcomes of maternal programmed miR-126-3p levels in offspring adipose tissue. RESULTS: The proteomic approach confirmed the identity of known targets of miR-126-3p (including IRS-1) and identified Lunapark, an endoplasmic reticulum (ER) protein, as a novel one. We confirmed by luciferase assay that Lunapark was a direct target of miR-126-3p. Overexpression of miR-126-3p in vitro led to a reduction in Lunapark protein levels and increased Perk (also known as Eif2ak3) mRNA levels and small interference-RNA mediated knockdown of Lunapark led to increased Xbp1, spliced Xbp1, Chop (also known as Ddit3) and Perk mRNA levels and an ER stress transcriptional response in 3T3-L1 pre-adipocytes. Consistent with the results found in vitro, increased miR-126-3p expression in adipose tissue from adult mouse offspring born to obese dams was accompanied by decreased Lunapark and IRS-1 protein levels and increased markers of ER stress. At the whole-body level the animals displayed glucose intolerance. CONCLUSIONS/INTERPRETATION: Concurrently targeting IRS-1 and Lunapark, a nutritionally programmed increase in miR-126-3p causes adipose tissue insulin resistance and an ER stress response, both of which may contribute to impaired glucose tolerance. These findings provide a novel mechanism by which obesity during pregnancy leads to increased risk of type 2 diabetes in the offspring and therefore identify miR-126-3p as a potential therapeutic target.


Subject(s)
Adipocytes/metabolism , Adipose Tissue/metabolism , Endoplasmic Reticulum Stress , Homeodomain Proteins/metabolism , MicroRNAs/metabolism , Obesity, Maternal/metabolism , Prenatal Exposure Delayed Effects , 3T3-L1 Cells , Adipocytes/pathology , Adipose Tissue/pathology , Animals , Blood Glucose/metabolism , Disease Models, Animal , Down-Regulation , Female , Homeodomain Proteins/genetics , Insulin Receptor Substrate Proteins/genetics , Insulin Receptor Substrate Proteins/metabolism , Insulin Resistance , Mice , Mice, Inbred C57BL , MicroRNAs/genetics , Obesity, Maternal/genetics , Obesity, Maternal/pathology , Phenotype , Pregnancy , Signal Transduction
10.
Nutrients ; 12(4)2020 Apr 22.
Article in English | MEDLINE | ID: mdl-32331244

ABSTRACT

Gestational diabetes mellitus (GDM) annually affects 35,000 pregnancies in the United Kingdom, causing suboptimal health outcomes to the mother and child. Obesity and excessive gestational weight gain are risk factors for GDM. The Institute of Medicine recommends weight targets for women that are overweight and obese, however, there are no clear guidelines for women with GDM. Observational data suggest that modest weight loss (0.6-2 kg) after 28 weeks may reduce risk of caesarean section, large-for-gestational-age (LGA), and maternal postnatal glycaemia. This protocol for a multicentre randomised double-blind controlled trial aims to identify if a fully controlled reduced energy diet in GDM pregnancy improves infant birthweight and reduces maternal weight gain (primary outcomes). A total of 500 women with GDM (National Institute of Health and Care Excellence (NICE) 2015 criteria) and body mass index (BMI) ≥25 kg/m2 will be randomised to receive a standard (2000 kcal/day) or reduced energy (1200 kcal/day) diet box containing all meals and snacks from 28 weeks to delivery. Women and caregivers will be blinded to the allocations. Food diaries, continuous glucose monitoring, and anthropometry will measure dietary compliance, glucose levels, and weight changes. Women will receive standard antenatal GDM management (insulin/metformin) according to NICE guidelines. The secondary endpoints include caesarean section rates, LGA, and maternal postnatal glucose concentrations.


Subject(s)
Diabetes, Gestational/diet therapy , Diet, Reducing , Pregnant Women , Blood Glucose/metabolism , Cesarean Section/statistics & numerical data , Diabetes, Gestational/etiology , Diabetes, Gestational/metabolism , Diabetes, Gestational/prevention & control , Female , Gestational Weight Gain , Humans , Obesity/complications , Pregnancy , Pregnancy Complications , Risk Factors , Weight Reduction Programs
11.
Int J Obes (Lond) ; 44(5): 1087-1096, 2020 05.
Article in English | MEDLINE | ID: mdl-32203108

ABSTRACT

BACKGROUND: In utero exposure to obesity is consistently associated with increased risk of metabolic disease, obesity and cardiovascular dysfunction in later life despite the divergence of birth weight outcomes. The placenta plays a critical role in offspring development and long-term health, as it mediates the crosstalk between the maternal and fetal environments. However, its phenotypic and molecular modifications in the context of maternal obesity associated with fetal growth restriction (FGR) remain poorly understood. METHODS: Using a mouse model of maternal diet-induced obesity, we investigated changes in the placental transcriptome through RNA sequencing (RNA-seq) and Ingenuity Pathway Analysis (IPA) at embryonic day (E) 19. The most differentially expressed genes (FDR < 0.05) were validated by Quantitative real-time PCR (qPCR) in male and female placentae at E19. The expression of these targets and related genes was also determined by qPCR at E13 to examine whether the observed alterations had an earlier onset at mid-gestation. Structural analyses were performed using immunofluorescent staining against Ki67 and CD31 to investigate phenotypic outcomes at both timepoints. RESULTS: RNA-seq and IPA analyses revealed differential expression of transcripts and pathway interactions related to placental vascular development and tissue morphology in obese placentae at term, including downregulation of Muc15, Cnn1, and Acta2. Pdgfb, which is implicated in labyrinthine layer development, was downregulated in obese placentae at E13. This was consistent with the morphological evidence of reduced labyrinth zone (LZ) size, as well as lower fetal weight at both timepoints irrespective of offspring sex. CONCLUSIONS: Maternal obesity results in abnormal placental LZ development and impaired vascularization, which may mediate the observed FGR through reduced transfer of nutrients across the placenta.


Subject(s)
Fetal Growth Retardation , Obesity, Maternal , Placenta , Transcriptome/genetics , Animals , Disease Models, Animal , Female , Fetal Growth Retardation/genetics , Fetal Growth Retardation/metabolism , Male , Mice , Mice, Inbred C57BL , Obesity, Maternal/genetics , Obesity, Maternal/metabolism , Placenta/metabolism , Placenta/pathology , Pregnancy
12.
Diabetologia ; 63(2): 324-337, 2020 02.
Article in English | MEDLINE | ID: mdl-31773193

ABSTRACT

AIMS/HYPOTHESIS: Obesity during pregnancy increases offspring type 2 diabetes risk. Given that nearly half of women of child-bearing age in many populations are currently overweight/obese, it is key that we improve our understanding of the impact of the in utero/early life environment on offspring islet function. Whilst a number of experimental studies have examined the effect of maternal obesity on offspring islet architecture and/or function, it has not previously been delineated whether these changes are independent of other confounding risk factors such as obesity, postnatal high-fat-feeding and ageing. Thus, we aimed to study the impact of exposure to maternal obesity on offspring islets in young, glucose-tolerant male and female offspring. METHODS: Female C57BL/6J mice were fed ad libitum either chow or obesogenic diet prior to and throughout pregnancy and lactation. Offspring were weaned onto a chow diet and remained on this diet until the end of the study. An IPGTT was performed on male and female offspring at 7 weeks of age. At 8 weeks of age, pancreatic islets were isolated from offspring for measurement of insulin secretion and content, mitochondrial respiration, ATP content, reactive oxygen species levels, beta and alpha cell mass, granule and mitochondrial density (by transmission electron microscopy), and mRNA and protein expression by real-time RT-PCR and Western blotting, respectively. RESULTS: Glucose tolerance was similar irrespective of maternal diet and offspring sex. However, blood glucose was lower (p < 0.001) and plasma insulin higher (p < 0.05) in female offspring of obese dams 15 min after glucose administration. This was associated with higher glucose- (p < 0.01) and leucine/glutamine-stimulated (p < 0.05) insulin secretion in these offspring. Furthermore, there was increased mitochondrial respiration (p < 0.01) and density (p < 0.05) in female offspring of obese dams compared with same-sex controls. Expression of mitochondrial and nuclear-encoded components of the electron transport chain, L-type Ca2+ channel subtypes that play a key role in stimulus-secretion coupling [Cacna1d (p < 0.05)], and oestrogen receptor α (p < 0.05) was also increased in islets from these female offspring of obese dams. Moreover, cleaved caspase-3 expression and BAX:Bcl-2 were decreased (p < 0.05) reflecting reduced susceptibility to apoptosis. In contrast, in male offspring, glucose and leucine/glutamine-stimulated insulin secretion was comparable between treatment groups. There was, however, compromised mitochondrial respiration characterised by decreased ATP synthesis-driven respiration (p < 0.05) and increased uncoupled respiration (p < 0.01), reduced docked insulin granules (p < 0.001), decreased Cacna1c (p < 0.001) and Cacna1d (p < 0.001) and increased cleaved caspase-3 expression (p < 0.05). CONCLUSIONS/INTERPRETATION: Maternal obesity programs sex differences in offspring islet function. Islets of female but not male offspring appear to be primed to cope with a nutritionally-rich postnatal environment, which may reflect differences in future type 2 diabetes risk.


Subject(s)
Diabetes Mellitus, Type 2/metabolism , Islets of Langerhans/metabolism , Obesity, Maternal/metabolism , Adenosine Triphosphate/metabolism , Animals , Apoptosis/physiology , Blotting, Western , DNA, Mitochondrial/metabolism , Female , Fluorescent Antibody Technique , Male , Mice , Mice, Inbred C57BL , Microscopy, Electron, Transmission , Oxygen Consumption/physiology , Pregnancy , Reverse Transcriptase Polymerase Chain Reaction , Sex Characteristics
13.
Elife ; 82019 06 26.
Article in English | MEDLINE | ID: mdl-31241463

ABSTRACT

Studies suggest that placental nutrient supply adapts according to fetal demands. However, signaling events underlying placental adaptations remain unknown. Here we demonstrate that phosphoinositide 3-kinase p110α in the fetus and the trophoblast interplay to regulate placental nutrient supply and fetal growth. Complete loss of fetal p110α caused embryonic death, whilst heterozygous loss resulted in fetal growth restriction and impaired placental formation and nutrient transport. Loss of trophoblast p110α resulted in viable fetuses, abnormal placental development and a failure of the placenta to transport sufficient nutrients to match fetal demands for growth. Using RNA-seq we identified genes downstream of p110α in the trophoblast that are important in adapting placental phenotype. Using CRISPR/Cas9 we showed loss of p110α differentially affects gene expression in trophoblast and embryonic stem cells. Our findings reveal important, but distinct roles for p110α in the different compartments of the conceptus, which control fetal resource acquisition and growth.


Subject(s)
Class I Phosphatidylinositol 3-Kinases/metabolism , Embryonic Stem Cells/enzymology , Energy Metabolism , Fetal Development , Placentation , Trophoblasts/enzymology , Animals , Female , Fetus , Mice , Pregnancy , Signal Transduction
14.
Mol Metab ; 16: 35-44, 2018 10.
Article in English | MEDLINE | ID: mdl-30293577

ABSTRACT

OBJECTIVE: Obesity during pregnancy is associated with an elevated risk of cardiovascular disease in the offspring. With increased numbers of women entering pregnancy overweight or obese, there is a requirement for targeted interventions to reduce disease risk in future generations. Using an established murine model of maternal obesity during pregnancy, we investigated if a treadmill exercise intervention in the mother could improve offspring cardiac health and explored potential underlying mechanisms. METHODS: A 20-minute treadmill exercise intervention protocol was performed 5 days a week in diet-induced obese female C57BL/6 mice 1 week prior to, and up to E17 of pregnancy. All male offspring were weaned onto a control diet and studied at 8 weeks of age when their cardiovascular physiology was assessed by in vivo echocardiography and non-invasive tail cuff plethysmography. Cardiomyocyte cell area, re-expression of fetal genes and the expression of calcium handling and sympathetic activation proteins were determined. RESULTS: At 8 weeks, there was no difference in bodyweight or fat mass between groups. Offspring of obese dams developed pathologic cardiac hypertrophy, hypertension and cardiac dysfunction characterized by reduced ejection fraction (p < 0.001). Maternal exercise prevented cardiac hypertrophy and dysfunction but failed to prevent hypertension. These offspring of exercised dams also had enhanced (p < 0.001) levels of calcium handling proteins and a sympathetic-activated inotropic response. CONCLUSIONS: Exercise in obese pregnancy was beneficial to offspring cardiac function and structure but did not influence hypertension suggesting they are programmed by separate mechanistic pathways. These data suggest combination interventions in obese pregnancies will be required to improve all aspects of the cardiovascular health of the next generation.


Subject(s)
Obesity/physiopathology , Obesity/therapy , Physical Conditioning, Animal/physiology , Adiposity , Animals , Body Composition , Body Weight/physiology , Cardiovascular Physiological Phenomena , Diet , Exercise Therapy/methods , Female , Lactation , Male , Mice , Mice, Inbred C57BL , Overweight/metabolism , Physical Exertion/physiology , Pregnancy , Pregnancy Complications , Prenatal Care , Weaning
15.
Endocrinology ; 157(11): 4246-4256, 2016 Nov.
Article in English | MEDLINE | ID: mdl-27583789

ABSTRACT

Obesity during pregnancy has a long-term effect on the health of the offspring including risk of developing the metabolic syndrome. Using a mouse model of maternal diet-induced obesity, we employed a genome-wide approach to investigate the microRNA (miRNA) and miRNA transcription profile in adipose tissue to understand mechanisms through which this occurs. Male offspring of diet-induced obese mothers, fed a control diet from weaning, showed no differences in body weight or adiposity at 8 weeks of age. However, offspring from the obese dams had up-regulated cytokine (Tnfα; P < .05) and chemokine (Ccl2 and Ccl7; P < .05) signaling in their adipose tissue. This was accompanied by reduced expression of miR-706, which we showed can directly regulate translation of the inflammatory proteins IL-33 (41% up-regulated; P < .05) and calcium/calmodulin-dependent protein kinase 1D (30% up-regulated; P < .01). We conclude that exposure to obesity during development primes an inflammatory environment in adipose tissue that is independent of offspring adiposity. Programming of adipose tissue miRNAs that regulate expression of inflammatory signaling molecules may be a contributing mechanism.


Subject(s)
Adipose Tissue/metabolism , Maternal-Fetal Exchange/physiology , Obesity/metabolism , Obesity/physiopathology , Adipose Tissue/immunology , Adiposity/genetics , Adiposity/physiology , Animals , Body Weight/physiology , Chemokines/metabolism , Cytokines/metabolism , Female , Interleukin-33/metabolism , Male , Maternal Nutritional Physiological Phenomena , Maternal-Fetal Exchange/genetics , Mice , MicroRNAs , Pregnancy
16.
Am J Physiol Regul Integr Comp Physiol ; 303(1): R86-93, 2012 Jul 01.
Article in English | MEDLINE | ID: mdl-22552791

ABSTRACT

Fetal growth restriction (FGR) is the inability of a fetus to reach its genetically predetermined growth potential. In the absence of a genetic anomaly or maternal undernutrition, FGR is attributable to "placental insufficiency": inappropriate maternal/fetal blood flow, reduced nutrient transport or morphological abnormalities of the placenta (e.g., altered barrier thickness). It is not known whether these diverse factors act singly, or in combination, having additive effects that may lead to greater FGR severity. We suggest that multiplicity of such dysfunction might underlie the diverse FGR phenotypes seen in humans. Pregnant endothelial nitric oxide synthase knockout (eNOS(-/-)) dams exhibit dysregulated vascular adaptations to pregnancy, and eNOS(-/-) fetuses of such dams display FGR. We investigated the hypothesis that both altered vascular function and placental nutrient transport contribute to the FGR phenotype. eNOS(-/-) dams were hypertensive prior to and during pregnancy and at embryonic day (E) 18.5 were proteinuric. Isolated uterine artery constriction was significantly increased, and endothelium-dependent relaxation significantly reduced, compared with wild-type (WT) mice. eNOS(-/-) fetal weight and abdominal circumference were significantly reduced compared with WT. Unidirectional maternofetal (14)C-methylaminoisobutyric acid (MeAIB) clearance and sodium-dependent (14)C-MeAIB uptake into mouse placental vesicles were both significantly lower in eNOS(-/-) fetuses, indicating diminished placental nutrient transport. eNOS(-/-) mouse placentas demonstrated increased hypoxia at E17.5, with elevated superoxide compared with WT. We propose that aberrant uterine artery reactivity in eNOS(-/-) mice promotes placental hypoxia with free radical formation, reducing placental nutrient transport capacity and fetal growth. We further postulate that this mouse model demonstrates "uteroplacental hypoxia," providing a new framework for understanding the etiology of FGR in human pregnancy.


Subject(s)
Fetal Growth Retardation/physiopathology , Models, Animal , Nitric Oxide Synthase Type III/deficiency , Phenotype , Placenta/physiopathology , Uterine Artery/physiopathology , Amino Acid Transport System A/metabolism , Animals , Biological Transport/physiology , Blood Pressure/physiology , Female , Fetal Growth Retardation/metabolism , Fetal Weight/physiology , Mice , Mice, Inbred C57BL , Mice, Knockout , Nitric Oxide Synthase Type III/genetics , Placenta/metabolism , Pregnancy , Proteinuria/metabolism , Proteinuria/physiopathology , Superoxides/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...