Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
1.
Clin Epigenetics ; 16(1): 61, 2024 May 07.
Article in English | MEDLINE | ID: mdl-38715048

ABSTRACT

BACKGROUND: Diabetes in pregnancy is associated with increased risk of long-term metabolic disease in the offspring, potentially mediated by in utero epigenetic variation. Previously, we identified multiple differentially methylated single CpG sites in offspring of women with gestational diabetes mellitus (GDM), but whether stretches of differentially methylated regions (DMRs) can also be identified in adolescent GDM offspring is unknown. Here, we investigate which DNA regions in adolescent offspring are differentially methylated in blood by exposure to diabetes in pregnancy. The secondary aim was to characterize the RNA expression of the identified DMR, which contained the nc886 non-coding RNA. METHODS: To identify DMRs, we employed the bump hunter method in samples from young (9-16 yr, n = 92) offspring of women with GDM (O-GDM) and control offspring (n = 94). Validation by pyrosequencing was performed in an adult offspring cohort (age 28-33 years) consisting of O-GDM (n = 82), offspring exposed to maternal type 1 diabetes (O-T1D, n = 67) and control offspring (O-BP, n = 57). RNA-expression was measured using RT-qPCR in subcutaneous adipose tissue and skeletal muscle. RESULTS: One significant DMR represented by 10 CpGs with a bimodal methylation pattern was identified, located in the nc886/VTRNA2-1 non-coding RNA gene. Low methylation status across all CpGs of the nc886 in the young offspring was associated with maternal GDM. While low methylation degree in adult offspring in blood, adipose tissue, and skeletal muscle was not associated with maternal GDM, adipose tissue nc886 expression was increased in O-GDM compared to O-BP, but not in O-T1D. In addition, adipose tissue nc886 expression levels were positively associated with maternal pre-pregnancy BMI (p = 0.006), but not with the offspring's own adiposity. CONCLUSIONS: Our results highlight that nc886 is a metastable epiallele, whose methylation in young offspring is negatively correlated with maternal obesity and GDM status. The physiological effect of nc886 may be more important in adipose tissue than in skeletal muscle. Further research should aim to investigate how nc886 regulation in adipose tissue by exposure to GDM may contribute to development of metabolic disease.


Subject(s)
Adipose Tissue , DNA Methylation , Diabetes, Gestational , Epigenesis, Genetic , Muscle, Skeletal , Prenatal Exposure Delayed Effects , Humans , Pregnancy , Female , Diabetes, Gestational/genetics , Epigenesis, Genetic/genetics , Adult , DNA Methylation/genetics , Muscle, Skeletal/metabolism , Adolescent , Adipose Tissue/metabolism , Male , Prenatal Exposure Delayed Effects/genetics , Child , Diabetes Mellitus, Type 1/genetics , RNA, Untranslated/genetics , RNA, Untranslated/blood , RNA, Long Noncoding/genetics , CpG Islands/genetics
2.
Sci Rep ; 13(1): 19638, 2023 11 10.
Article in English | MEDLINE | ID: mdl-37949941

ABSTRACT

The fetal insulin hypothesis proposes that low birthweight and type 2 diabetes (T2D) in adulthood may be two phenotypes of the same genotype. In this study we aimed to explore this theory further by testing the effects of GWAS-identified genetic variants related to insulin release and sensitivity on fetal growth and blood flow from week 20 of gestation to birth and on placental weight at birth. We calculated genetic risk scores (GRS) of first phase insulin release (FPIR), fasting insulin (FI), combined insulin resistance and dyslipidaemia (IR + DLD) and insulin sensitivity (IS) in a study population of 665 genotyped newborns. Two-dimensional ultrasound measurements with estimation of fetal weight and blood flow were carried out at week 20, 25, and 32 of gestation in all 665 pregnancies. Birthweight and placental weight were registered at birth. Associations between the GRSs and fetal growth, blood flow and placental weight were investigated using linear mixed models. The FPIR GRS was directly associated with fetal growth from week 20 to birth, and both the FI GRS, IR + DLD GRS, and IS GRS were associated with placental weight at birth. Our findings indicate that insulin-related genetic variants might primarily affect fetal growth via the placenta.


Subject(s)
Diabetes Mellitus, Type 2 , Insulin Resistance , Humans , Pregnancy , Infant, Newborn , Female , Insulin , Placenta/physiology , Birth Weight/genetics , Diabetes Mellitus, Type 2/genetics , Diabetes Mellitus, Type 2/epidemiology , Prospective Studies , Fetal Development/genetics , Insulin Resistance/genetics , Fetal Weight
3.
Diabetes Res Clin Pract ; 203: 110858, 2023 Sep.
Article in English | MEDLINE | ID: mdl-37516412

ABSTRACT

AIM: The aim of this study was to explore the perceptions of women with gestational diabetes mellitus (GDM) in Denmark, with a particular focus on GDM-specific stigma. METHOD: We conducted semi-structured interviews with 20 women with GDM from January to May 2022. All interviews were transcribed and analysed abductively using Braun and Clarke's framework for applied reflexive analysis. RESULTS: Five themes were identified, 1) victim-blaming narrative, 2) identity threat, 3) non-disclosure and anticipated stigma, 4) stigma in a clinical setting, and 5) stigma reduction in a clinical setting. Additionally, intersectionality was identified between GDM-specific stigma, notions of how to be a good mother, and stigma associated with having type 2 diabetes mellitus and overweight. Implications of GDM-specific stigma included suboptimal GDM care and management, i.e., not attending screening for GDM, and not wanting to disclose the diagnosis. CONCLUSION: The impact of GDM-specific stigma on the informants' lives included some informants not accepting all services provided by the healthcare system, and some not wanting to identify with the diagnosis. These findings may help inform both healthcare personnel and future health promotion interventions to minimize the reproduction of a victim-blaming narrative and thereby promote well-being among women with GDM.

4.
Biomedicines ; 10(6)2022 May 26.
Article in English | MEDLINE | ID: mdl-35740266

ABSTRACT

Maternal gestational diabetes and obesity are associated with adverse outcomes in offspring, including increased risk of diabetes and cardiovascular diseases. Previously, we identified a lower DNA methylation degree at genomic sites near the genes ESM1, MS4A3, and TSPAN14 in the blood cells of adolescent offspring exposed to gestational diabetes and/or maternal obesity in utero. In the present study, we aimed to investigate if altered methylation and expression of these genes were detectable in blood, as well in the metabolically relevant subcutaneous adipose tissue, in a separate cohort of adult offspring exposed to gestational diabetes and obesity (O-GDM) or type 1 diabetes (O-T1D) in utero, compared with the offspring of women from the background population (O-BP). We did not replicate the findings of lower methylation of ESM1, MS4A3, and TSPAN14 in blood from adults, either in O-GDM or O-T1D. In contrast, in adipose tissue of O-T1D, we found higher MS4A3 DNA methylation, which will require further validation. The adipose tissue ESM1 expression was lower in O-GDM compared to O-BP, which in turn was not associated with maternal pre-pregnancy BMI nor the offspring's own adiposity. Adipose tissue TSPAN14 expression was slightly lower in O-GDM compared with O-BP, but also positively associated with maternal pre-pregnancy BMI, as well as offspring's own adiposity and HbA1c levels. In conclusion, the lower DNA methylation in blood from adolescent offspring exposed to GDM could not be confirmed in the present cohort of adult offspring, potentially due to methylation remodeling with increased aging. In offspring adipose tissue, ESM1 expression was associated with maternal GDM, and TSPAN14 expression was associated with both maternal GDM, as well as pre-pregnancy BMI. These altered expression patterns are potentially relevant to the concept of developmental programming of cardiometabolic diseases and require further studies.

5.
BMC Med Genomics ; 14(1): 3, 2021 01 06.
Article in English | MEDLINE | ID: mdl-33407418

ABSTRACT

BACKGROUND: In order to explore the pathophysiology underlying type 2 diabetes we examined the impact of gene variants associated with type 2 diabetes on circulating levels of glucagon during an oral glucose tolerance test (OGTT). Furthermore, we performed a genome-wide association study (GWAS) aiming to identify novel genomic loci affecting plasma glucagon levels. METHODS: Plasma levels of glucagon were examined in samples obtained at three time points during an OGTT; 0, 30 and 120 min, in two separate cohorts with a total of up to 1899 individuals. Cross-sectional analyses were performed separately in the two cohorts and the results were combined in a meta-analysis. RESULTS: A known type 2 diabetes variant in EYA2 was significantly associated with higher plasma glucagon level at 30 min during the OGTT (Beta 0.145, SE 0.038, P = 1.2 × 10-4) corresponding to a 7.4% increase in plasma glucagon level per effect allele. In the GWAS, we identified a marker in the MARCH1 locus, which was genome-wide significantly associated with reduced suppression of glucagon during the first 30 min of the OGTT (Beta - 0.210, SE 0.037, P = 1.9 × 10-8), equivalent to 8.2% less suppression per effect allele. Nine additional independent markers, not previously associated with type 2 diabetes, showed suggestive associations with reduced glucagon suppression during the first 30 min of the OGTT (P < 1.0 × 10-5). CONCLUSIONS: A type 2 diabetes risk variant in the EYA2 locus was associated with higher plasma glucagon levels at 30 min. Ten additional variants were suggestively associated with reduced glucagon suppression without conferring increased type 2 diabetes risk.


Subject(s)
Genome-Wide Association Study , Adult , Diabetes Mellitus, Type 2 , Glucagon , Glucose Tolerance Test , Humans , Male , Middle Aged
6.
Ugeskr Laeger ; 181(33)2019 Aug 12.
Article in Danish | MEDLINE | ID: mdl-31495354

ABSTRACT

This review summarises the knowledge of recurrent diploid biparental hydatidiform mole, which is a rare genetic condition. Pathogenic variants in both alleles of NLRP7 or KHDC3L are associated with maternal imprinting defects and can cause the condition. Women with biallelic inactivation of NLRP7 can achieve a normal pregnancy by oocyte donation, and it is highly likely, that this applies to women with biallelic inactivation of KHDCL3 as well. Identifying the cause of the recurrent moles can prevent that couples waist time and possibly reduce medical expenses related to fertility treatment.


Subject(s)
Hydatidiform Mole , Uterine Neoplasms , Adaptor Proteins, Signal Transducing/genetics , Diploidy , Female , Humans , Hydatidiform Mole/genetics , Neoplasm Recurrence, Local , Pregnancy , Uterine Neoplasms/genetics
7.
Ugeskr Laeger ; 180(36)2018 Sep 03.
Article in Danish | MEDLINE | ID: mdl-30187851

ABSTRACT

Harlequin ichthyosis (HI) is a rare and severe form of the autosomal recessive congenital ichthyosis. This is a case report of a 30-year-old healthy woman with a pregnancy resulting in preterm birth of a child with severe HI, who did not survive. At the autopsy, the child was found with HI and a diaphragmatic hernia of the Bochdalek type. Genetic analysis showed, that the child was homozygous for the mutation c.5121_5124del in ABCA12. The parents were related and were found heterozygous of this mutation. This clinical presentation with this new mutation has not been described in the literature before.


Subject(s)
ATP-Binding Cassette Transporters/genetics , Ichthyosis, Lamellar/genetics , Adult , Female , Hernias, Diaphragmatic, Congenital/genetics , Hernias, Diaphragmatic, Congenital/pathology , Humans , Ichthyosis, Lamellar/pathology , Infant, Newborn , Infant, Premature , Male , Mutation , Perinatal Death , Pregnancy , Premature Birth
8.
Hum Mol Genet ; 27(10): 1763-1771, 2018 05 15.
Article in English | MEDLINE | ID: mdl-29528396

ABSTRACT

Offspring of women with diabetes in pregnancy exhibit skeletal muscle insulin resistance and are at increased risk of developing type 2 diabetes, potentially mediated by epigenetic mechanisms or changes in the expression of small non-coding microRNAs. Members of the miR-15 family can alter the expression or function of important proteins in the insulin signalling pathway, affecting insulin sensitivity and secretion. We hypothesized that exposure to maternal diabetes may cause altered expression of these microRNAs in offspring skeletal muscle, representing a potential underlying mechanism by which exposure to maternal diabetes leads to increased risk of cardiometabolic disease in offspring. We measured microRNA expression in skeletal muscle biopsies of 26- to 35-year-old offspring of women with either gestational diabetes (O-GDM, n = 82) or type 1 diabetes (O-T1DM, n = 67) in pregnancy, compared with a control group of offspring from the background population (O-BP, n = 57) from an observational follow-up study. Expression of both miR-15a and miR-15b was increased in skeletal muscle obtained from O-GDM (both P < 0.001) and O-T1DM (P = 0.024, P = 0.005, respectively) compared with O-BP. Maternal 2 h post OGTT glucose levels were positively associated with miR-15a expression (P = 0.041) in O-GDM after adjustment for confounders and mediators. In all groups collectively, miRNA expression was significantly positively associated with fasting plasma glucose, 2 h plasma glucose and HbA1c. We conclude that fetal exposure to maternal diabetes is associated with increased skeletal muscle expression of miR-15a and miR-15b and that this may contribute to development of metabolic disease in these subjects.


Subject(s)
Diabetes Mellitus, Type 2/genetics , Diabetes, Gestational/genetics , Epigenesis, Genetic , MicroRNAs/genetics , Adult , Adult Children , Diabetes Mellitus, Type 2/blood , Diabetes Mellitus, Type 2/pathology , Diabetes, Gestational/blood , Diabetes, Gestational/pathology , Female , Gene Expression Regulation , Glycated Hemoglobin/genetics , Humans , Insulin/metabolism , Insulin Resistance/genetics , Male , Muscle, Skeletal/pathology , Pregnancy
9.
PLoS One ; 12(10): e0187038, 2017.
Article in English | MEDLINE | ID: mdl-29077742

ABSTRACT

BACKGROUND: Fetal exposure to maternal diabetes increases the risk of type 2 diabetes (T2DM), possibly mediated by epigenetic mechanisms. Low blood TXNIP DNA methylation has been associated with elevated glucose levels and risk of T2DM, and increased skeletal muscle TXNIP gene expression was reported in subjects with impaired glucose metabolism or T2DM. Subcutaneous adipose tissue (SAT) and skeletal muscle play a key role in the control of whole body glucose metabolism and insulin action. The extent to which TXNIP DNA methylation levels are decreased and/or gene expression levels increased in SAT or skeletal muscle of a developmentally programmed at-risk population is unknown. OBJECTIVE AND METHODS: The objective of this study was to investigate TXNIP DNA methylation and gene expression in SAT and skeletal muscle, and DNA methylation in blood, from adult offspring of women with gestational diabetes (O-GDM, n = 82) or type 1 diabetes (O-T1DM, n = 67) in pregnancy compared with offspring of women from the background population (O-BP, n = 57). RESULTS: SAT TXNIP DNA methylation was increased (p = 0.032) and gene expression decreased (p = 0.001) in O-GDM, but these differences were attenuated after adjustment for confounders. Neither blood/muscle TXNIP DNA methylation nor muscle gene expression differed between groups. CONCLUSION: We found no evidence of decreased TXNIP DNA methylation or increased gene expression in metabolic target tissues of offspring exposed to maternal diabetes. Further studies are needed to confirm and understand the paradoxical SAT TXNIP DNA methylation and gene expression changes in O-GDM subjects.


Subject(s)
Carrier Proteins/genetics , DNA Methylation , Gene Expression , Pregnancy in Diabetics/genetics , Adult , Denmark , Diabetes Mellitus, Type 1/complications , Diabetes Mellitus, Type 1/genetics , Diabetes Mellitus, Type 2/complications , Diabetes Mellitus, Type 2/genetics , Diabetes, Gestational/genetics , Female , Humans , Male , Muscle, Skeletal/metabolism , Pregnancy
10.
Clin Epigenetics ; 9: 37, 2017.
Article in English | MEDLINE | ID: mdl-28413567

ABSTRACT

BACKGROUND: Offspring of women with diabetes in pregnancy are at increased risk of type 2 diabetes mellitus (T2DM), potentially mediated by epigenetic mechanisms. The adipokines leptin, adiponectin, and resistin (genes: LEP, ADIPOQ, RETN) play key roles in the pathophysiology of T2DM. We hypothesized that offspring exposed to maternal diabetes exhibit alterations in epigenetic regulation of subcutaneous adipose tissue (SAT) adipokine transcription. We studied adipokine plasma levels, SAT gene expression, and DNA methylation of LEP, ADIPOQ, and RETN in adult offspring of women with gestational diabetes (O-GDM, N = 82) or type 1 diabetes (O-T1DM, N = 67) in pregnancy, compared to offspring of women from the background population (O-BP, N = 57). RESULTS: Compared to O-BP, we found elevated plasma leptin and resistin levels in O-T1DM, decreased gene expression of all adipokines in O-GDM, decreased RETN expression in O-T1DM, and increased LEP and ADIPOQ methylation in O-GDM. In multivariate regression analysis, O-GDM remained associated with increased ADIPOQ methylation and decreased ADIPOQ and RETN gene expression and O-T1DM remained associated with decreased RETN expression after adjustment for potential confounders and mediators. CONCLUSIONS: In conclusion, offspring of women with diabetes in pregnancy exhibit increased ADIPOQ DNA methylation and decreased ADIPOQ and RETN gene expression in SAT. However, altered methylation and expression levels were not reflected in plasma protein levels, and the functional implications of these findings remain uncertain.


Subject(s)
Adiponectin/genetics , DNA Methylation , Diabetes Mellitus, Type 2/genetics , Leptin/genetics , Maternal Exposure/adverse effects , Resistin/genetics , Subcutaneous Fat/metabolism , Adiponectin/blood , Adiponectin/metabolism , Adult Children , Diabetes Mellitus, Type 1/epidemiology , Diabetes, Gestational/epidemiology , Epigenesis, Genetic , Female , Follow-Up Studies , Gene Expression Regulation , Genetic Predisposition to Disease , Humans , Leptin/blood , Pregnancy , Resistin/blood
11.
J Clin Endocrinol Metab ; 102(4): 1141-1150, 2017 04 01.
Article in English | MEDLINE | ID: mdl-28204515

ABSTRACT

Context: Offspring of women with gestational diabetes (O-GDM) or type 1 diabetes mellitus (O-T1DM) have been exposed to hyperglycemia in utero and have an increased risk of developing metabolic disease in adulthood. Design: In total, we recruited 206 adult offspring comprising the two fetal hyperglycemic groups, O-GDM and O-T1DM, and, as a control group, offspring from the background population (O-BP). Subcutaneous fat biopsies were obtained and preadipocyte cell cultures were established from adult male O-GDM (n = 18, age 30.1 ± 2.5 years), O-T1DM (n = 18, age 31.6 ± 2.2 years), and O-BP (n = 16; age, 31.5 ± 2.7 years) and cultured in vitro. Main Outcome Measures: First, we studied in vivo adipocyte histology. Second, we studied in vitro preadipocyte leptin secretion, gene expression, and LEP DNA methylation. This was studied in combination with in vitro preadipocyte lipogenesis, lipolysis, and mitochondrial respiration. Results: We show that subcutaneous adipocytes from O-GDM are enlarged compared with O-BP adipocytes. Preadipocytes isolated from male O-GDM and O-T1DM and cultured in vitro displayed decreased LEP promoter methylation, increased leptin gene expression, and elevated leptin secretion throughout differentiation, compared with adipocytes established from male O-BP. In addition, the preadipocytes demonstrated functional defects including decreased maximal mitochondrial capacity with increased lipolysis and decreased ability to store fatty acids when challenged with 3 days of extra fatty acid supply. Conclusions: Taken together, these findings show that intrinsic epigenetic and functional changes exist in preadipocyte cultures from individuals exposed to fetal hyperglycemia who are at increased risk of developing metabolic disease.


Subject(s)
Adipocytes/physiology , Adult Children , Diabetes Mellitus, Type 1 , Diabetes, Gestational , Hyperglycemia/congenital , Pregnancy in Diabetics , Prenatal Exposure Delayed Effects/physiopathology , Adult , Case-Control Studies , Cells, Cultured , Diabetes Mellitus, Type 2/etiology , Disease Susceptibility , Female , Fetal Diseases/metabolism , Fetal Diseases/physiopathology , Humans , Hyperglycemia/metabolism , Hyperglycemia/physiopathology , Lipolysis/physiology , Male , Pregnancy , Prenatal Exposure Delayed Effects/metabolism , Risk Factors
12.
Diabetes ; 65(10): 2900-10, 2016 10.
Article in English | MEDLINE | ID: mdl-27388218

ABSTRACT

Prenatal exposure to maternal hyperglycemia is associated with an increased risk of later adverse metabolic health. Changes in the regulation of peroxisome proliferator-activated receptor-γ coactivator-1α (PPARGC1A) in skeletal muscle and subcutaneous adipose tissue (SAT) is suggested to play a role in the developmental programming of dysmetabolism based on studies of human subjects exposed to an abnormal intrauterine environment (e.g., individuals with a low birth weight). We studied 206 adult offspring of women with gestational diabetes mellitus (O-GDM) or type 1 diabetes (O-T1D) and of women from the background population (O-BP) using a clinical examination, oral glucose tolerance test, and gene expression and DNA methylation of PPARGC1A in skeletal muscle and SAT. Plasma glucose was significantly higher for both O-GDM and O-T1D compared with O-BP (P < 0.05). PPARGC1A gene expression in muscle was lower in O-GDM compared with O-BP (P = 0.0003), whereas no differences were found between O-T1D and O-BP in either tissue. PPARGC1A DNA methylation percentages in muscle and SAT were similar among all groups. Decreased PPARGC1A gene expression in muscle has previously been associated with abnormal insulin function and may thus contribute to the increased risk of metabolic disease in O-GDM. The unaltered PPARGC1A gene expression in muscle of O-T1D suggests that factors other than intrauterine hyperglycemia may contribute to the decreased PPARGC1A expression in O-GDM.


Subject(s)
Adipose Tissue/metabolism , DNA Methylation , Diabetes, Gestational/physiopathology , Gene Expression , Muscle, Skeletal/metabolism , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/genetics , Adult , Blood Glucose/metabolism , Diabetes Mellitus, Type 1/genetics , Diabetes Mellitus, Type 1/physiopathology , Diabetes, Gestational/genetics , Female , Glucose Tolerance Test , Humans , Middle Aged , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/metabolism , Pregnancy
13.
Diabetologia ; 59(7): 1396-1399, 2016 07.
Article in English | MEDLINE | ID: mdl-27174368

ABSTRACT

Gestational diabetes mellitus (GDM) is defined as glucose intolerance of varying severity and is present in about 2-6% of all pregnancies in Europe, making it one of the most common pregnancy disorders. Aside from the short-term maternal, fetal and neonatal consequences associated with GDM, there are long-term consequences for both mother and child. Although maternal glucose tolerance often normalises shortly after pregnancy, women with GDM have a substantially increased risk of developing type 2 diabetes later in life. Studies have reported that women are more than seven times as likely to develop diabetes after GDM, and that approximately 50% of mothers with GDM will develop diabetes within 10 years, making GDM one of the strongest predictors of type 2 diabetes. In women with previous GDM, development of type 2 diabetes can be prevented or delayed by lifestyle intervention and/or medical treatment. Systematic follow-up programmes would be ideal to prevent progression of GDM to diabetes, but such programmes are unfortunately lacking in the routine clinical set-up in most countries. Studies have found that the risks of obesity, the metabolic syndrome, type 2 diabetes and impaired insulin sensitivity and secretion in offspring of mothers with GDM are two- to eightfold those in offspring of mothers without GDM. The underlying pathogenic mechanisms behind the abnormal metabolic risk profile in offspring are unknown, but epigenetic changes induced by exposure to maternal hyperglycaemia during fetal life are implicated. Animal studies indicate that treatment can prevent long-term metabolic complications in offspring, but this remains to be confirmed in humans. Thus, diabetes begets diabetes and it is likely that GDM plays a significant role in the global diabetes epidemic. This review summarises a presentation given at the 'Gestational diabetes: what's up?' symposium at the 2015 annual meeting of the EASD. It is accompanied by two other reviews on topics from this symposium (by Marja Vääräsmäki, DOI: 10.1007/s00125-016-3976-6 , and by Cuilin Zhang and colleagues, DOI: 10.1007/s00125-016-3979-3 ) and an overview by the Session Chair, Kerstin Berntorp (DOI: 10.1007/s00125-016-3975-7 ).


Subject(s)
Diabetes Mellitus, Type 2/epidemiology , Diabetes, Gestational/epidemiology , Denmark/epidemiology , Female , Humans , Mothers , Pregnancy , Pregnancy Complications , Risk Factors
14.
Diabetes Res Clin Pract ; 114: 50-4, 2016 Apr.
Article in English | MEDLINE | ID: mdl-27103369

ABSTRACT

Plasma levels of the inflammatory marker YKL-40 were investigated in 597 adult offspring born to women with and without diabetes during pregnancy. No association between fetal exposure to maternal hyperglycemia and levels of YKL-40 was found. However, female sex and increasing BMI in the offspring were associated to YKL-40.


Subject(s)
Biomarkers/blood , Chitinase-3-Like Protein 1/blood , Diabetes Mellitus, Type 1/epidemiology , Diabetes, Gestational/epidemiology , Hyperglycemia/complications , Inflammation/diagnosis , Adult , Adult Children , Denmark/epidemiology , Diabetes Mellitus, Type 1/blood , Diabetes Mellitus, Type 1/immunology , Diabetes, Gestational/blood , Diabetes, Gestational/immunology , Female , Humans , Hyperglycemia/epidemiology , Hyperglycemia/physiopathology , Inflammation/blood , Inflammation/etiology , Male , Pregnancy , Risk Factors , Young Adult
15.
J Clin Endocrinol Metab ; 100(5): 1967-75, 2015 May.
Article in English | MEDLINE | ID: mdl-25781355

ABSTRACT

CONTEXT: Fetal exposure to maternal diabetes is associated with increased risk of type 2 diabetes mellitus (T2DM) later in life. The pathogenesis of T2DM involves dysfunction of the incretin hormones, glucagon-like peptide-1 (GLP-1) and glucose-dependent insulinotropic polypeptide (GIP), as well as hyperglucagonemia. OBJECTIVE: Our aim was to investigate circulating plasma levels of GLP-1, GIP, and glucagon during the oral glucose tolerance test (OGTT) in adult offspring of women with diabetes in pregnancy. DESIGN AND PARTICIPANTS: We conducted a follow-up study of 567 offspring, aged 18-27 years. We included two groups exposed to maternal diabetes in utero: offspring of women with diet-treated gestational diabetes mellitus (O-GDM; n = 163) or type 1 diabetes (O-T1DM; n = 146). Two reference groups were included: offspring of women with risk factors for GDM, but normoglycemia during pregnancy (O-NoGDM; n = 133) and offspring from the background population (O-BP; n = 125). The subjects underwent a 75-g OGTT with venous samples at 0, 30, and 120 minutes. RESULTS: Fasting plasma levels of GLP-1 were lower in the two diabetes-exposed groups compared to O-BP (O-GDM, P = .040; O-T1DM, P = .008). Increasing maternal blood glucose during OGTT in pregnancy was associated with reduced postprandial suppression of glucagon in the offspring. Lower levels of GLP-1 and higher levels of glucagon during the OGTT were present in offspring characterized by overweight or prediabetes/T2DM at follow-up, irrespective of exposure status. CONCLUSION: Lower levels of fasting GLP-1 and impaired glucagon suppression in adult offspring exposed to maternal diabetes during pregnancy are diabetogenic traits that may contribute to glucose intolerance in these persons, but further investigations are needed.


Subject(s)
Diabetes Mellitus, Type 1/blood , Diabetes, Gestational/blood , Gastric Inhibitory Polypeptide/blood , Glucagon-Like Peptide 1/blood , Glucagon/blood , Prenatal Exposure Delayed Effects/blood , Adolescent , Adult , Adult Children , Female , Follow-Up Studies , Glucose Tolerance Test , Humans , Male , Pregnancy , Young Adult
16.
J Clin Endocrinol Metab ; 98(9): 3793-801, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23796568

ABSTRACT

CONTEXT: Offspring of women with diabetes during pregnancy have an increased risk of glucose intolerance in adulthood, but the underlying mechanisms are unknown. OBJECTIVE: We aimed to investigate the effects of intrauterine hyperglycemia on insulin secretion and action in adult offspring of mothers with diabetes. DESIGN, SETTING, AND PARTICIPANTS: A cohort of 587 Caucasian offspring, without known diabetes, was followed up at the age of 18-27 years. We included 2 groups exposed to maternal diabetes in utero: offspring of women with gestational diabetes mellitus (n = 167) or type 1 diabetes (n = 153). Two reference groups were included: offspring of women with risk factors for gestational diabetes mellitus but normoglycemia during pregnancy (n = 139) and offspring from the background population (n = 128). MAIN OUTCOME MEASURES: Indices of insulin sensitivity and insulin release were calculated using insulin and glucose values from a standard oral glucose tolerance test (120 minutes, 75 g glucose). Pancreatic ß-cell function taking the prevailing insulin sensitivity into account was estimated by disposition indices. RESULTS: Both groups of offspring exposed during pregnancy to either maternal gestational diabetes or type 1 diabetes had reduced insulin sensitivity compared with offspring from the background population (both P < .005). We did not find any significant difference in absolute measures of insulin release. However, the disposition index was significantly reduced in both the diabetes-exposed groups (both P < .005). CONCLUSION: Reduced insulin sensitivity as well as impaired pancreatic ß-cell function may contribute to the increased risk of glucose intolerance among adult offspring born to women with diabetes during pregnancy.


Subject(s)
Child of Impaired Parents , Diabetes Mellitus, Type 1/physiopathology , Diabetes, Gestational/physiopathology , Insulin Resistance/physiology , Insulin-Secreting Cells/physiology , Pancreas/physiopathology , Pregnancy in Diabetics/physiopathology , Adolescent , Adult , Adult Children , Female , Glucose Tolerance Test , Humans , Male , Pregnancy , Risk Factors
17.
Diabetes Care ; 36(5): 1102-6, 2013 May.
Article in English | MEDLINE | ID: mdl-23248191

ABSTRACT

OBJECTIVE: Since January 2008, obese women with type 2 diabetes were advised to gain 0-5 kg during pregnancy. The aim with this study was to evaluate fetal growth and perinatal morbidity in relation to gestational weight gain in these women. RESEARCH DESIGN AND METHODS: A retrospective cohort comprised the records of 58 singleton pregnancies in obese women (BMI ≥30 kg/m(2)) with type 2 diabetes giving birth between 2008 and 2011. Birth weight was evaluated by SD z score to adjust for gestational age and sex. RESULTS: Seventeen women (29%) gained ≤5 kg, and the remaining 41 gained >5 kg. The median (range) gestational weight gains were 3.7 kg (-4.7 to 5 kg) and 12.1 kg (5.5-25.5 kg), respectively. Prepregnancy BMI was 33.5 kg/m(2) (30-53 kg/m(2)) vs. 36.8 kg/m(2) (30-48 kg/m(2)), P = 0.037, and median HbA1c was 6.7% at first visit in both groups and decreased to 5.7 and 6.0%, P = 0.620, in late pregnancy, respectively. Gestational weight gain ≤5 kg was associated with lower birth weight z score (P = 0.008), lower rates of large-for-gestational-age (LGA) infants (12 vs. 39%, P = 0.041), delivery closer to term (268 vs. 262 days, P = 0.039), and less perinatal morbidity (35 vs. 71%, P = 0.024) compared with pregnancies with maternal weight gain >5 kg. CONCLUSIONS: In this pilot study in obese women with type 2 diabetes, maternal gestational weight gain ≤5 kg was associated with a more proportionate birth weight and less perinatal morbidity.


Subject(s)
Diabetes Mellitus, Type 2/physiopathology , Fetal Development/physiology , Obesity/physiopathology , Weight Gain/physiology , Adult , Birth Weight/physiology , Female , Humans , Middle Aged , Pregnancy , Retrospective Studies , Young Adult
18.
Nat Rev Endocrinol ; 8(11): 659-67, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22965164

ABSTRACT

Type 1 diabetes mellitus in pregnant women increases the risk of adverse outcomes for mother and offspring. Careful preconception counselling and screening is important, with particular focus on glycaemic control, indications for antihypertensive therapy, screening for diabetic nephropathy, diabetic retinopathy and thyroid dysfunction, as well as review of other medications. Supplementation with folic acid should be initiated before conception in order to minimize the risk of fetal malformations. Obtaining and maintaining tight control of blood glucose and blood pressure before and during pregnancy is crucial for optimizing outcomes; however, the risk of severe hypoglycaemia during pregnancy is a major obstacle. Although pregnancy does not result in deterioration of kidney function in women with diabetic nephropathy and normal serum creatinine levels, pregnancy complications such as pre-eclampsia and preterm delivery are more frequent in these women than in women with T1DM and normal kidney function. Rapid-acting insulin analogues are considered safe to use in pregnancy and studies on long-acting insulin analogues have provided reassuring results. Immediately after delivery the insulin requirement declines to approximately 60% of the prepregnancy dose, and remains 10% lower than before pregnancy during breastfeeding.


Subject(s)
Breast Feeding/methods , Diabetes Mellitus, Type 1/therapy , Pregnancy in Diabetics/therapy , Prenatal Care/methods , Animals , Diabetes Mellitus, Type 1/blood , Diabetes Mellitus, Type 1/diagnosis , Disease Management , Female , Folic Acid/administration & dosage , Humans , Infant, Newborn , Postnatal Care/methods , Pregnancy , Pregnancy in Diabetics/blood , Pregnancy in Diabetics/diagnosis
19.
Diabetes Res Clin Pract ; 97(2): 322-30, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22622155

ABSTRACT

AIM: To investigate associations between fetal exposure to intrauterine hyperglycemia and plasma concentrations of interleukin-6 (IL-6) and high sensitivity C-reactive protein (hs-CRP) in adult offspring. METHOD: We studied 597 offspring, aged 18-27 years, from four different groups concerning exposure to intrauterine hyperglycemia and genetic predisposition to type 2 diabetes (offspring of women with: gestational diabetes mellitus (GDM), risk factors for GDM but normal glucose tolerance, type 1 diabetes and women from the background population, respectively). The participants were characterized by fasting plasma levels of IL-6 and hs-CRP, a 75 g oral glucose tolerance test and anthropometric measurements. RESULTS: No association between intrauterine exposure to hyperglycemia and levels of IL-6 and hs-CRP in the offspring was found. In contrast maternal overweight (body mass index ≥ 25 kg/m(2)) was positively associated with levels of both IL-6 and hs-CRP (p for both=0.003). Offspring who had already developed overweight or conditions of abnormal glucose tolerance were characterized by higher levels of IL-6 and hs-CRP compared with the remaining offspring (all p<0.007). CONCLUSION: Maternal overweight but not exposure to intrauterine hyperglycemia was associated with low-grade inflammation in adult offspring.


Subject(s)
Blood Glucose/metabolism , C-Reactive Protein/metabolism , Diabetes Mellitus, Type 1/epidemiology , Diabetes, Gestational/epidemiology , Hyperglycemia/epidemiology , Interleukin-6/blood , Overweight/epidemiology , Adolescent , Adult , Adult Children , Denmark/epidemiology , Diabetes Mellitus, Type 1/blood , Diabetes Mellitus, Type 1/immunology , Diabetes, Gestational/blood , Diabetes, Gestational/immunology , Female , Glucose Tolerance Test , Humans , Hyperglycemia/blood , Hyperglycemia/immunology , Inflammation/blood , Male , Overweight/blood , Pregnancy , Risk Factors , Young Adult
SELECTION OF CITATIONS
SEARCH DETAIL
...