Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 63
Filter
Add more filters

Publication year range
1.
PLoS Med ; 17(11): e1003229, 2020 11.
Article in English | MEDLINE | ID: mdl-33151971

ABSTRACT

BACKGROUND: Higher maternal plasma glucose (PG) concentrations, even below gestational diabetes mellitus (GDM) thresholds, are associated with adverse offspring outcomes, with DNA methylation proposed as a mediating mechanism. Here, we examined the relationships between maternal dysglycaemia at 24 to 28 weeks' gestation and DNA methylation in neonates and whether a dietary and physical activity intervention in pregnant women with obesity modified the methylation signatures associated with maternal dysglycaemia. METHODS AND FINDINGS: We investigated 557 women, recruited between 2009 and 2014 from the UK Pregnancies Better Eating and Activity Trial (UPBEAT), a randomised controlled trial (RCT), of a lifestyle intervention (low glycaemic index (GI) diet plus physical activity) in pregnant women with obesity (294 contol, 263 intervention). Between 27 and 28 weeks of pregnancy, participants had an oral glucose (75 g) tolerance test (OGTT), and GDM diagnosis was based on diagnostic criteria recommended by the International Association of Diabetes and Pregnancy Study Groups (IADPSG), with 159 women having a diagnosis of GDM. Cord blood DNA samples from the infants were interrogated for genome-wide DNA methylation levels using the Infinium Human MethylationEPIC BeadChip array. Robust regression was carried out, adjusting for maternal age, smoking, parity, ethnicity, neonate sex, and predicted cell-type composition. Maternal GDM, fasting glucose, 1-h, and 2-h glucose concentrations following an OGTT were associated with 242, 1, 592, and 17 differentially methylated cytosine-phosphate-guanine (dmCpG) sites (false discovery rate (FDR) ≤ 0.05), respectively, in the infant's cord blood DNA. The most significantly GDM-associated CpG was cg03566881 located within the leucine-rich repeat-containing G-protein coupled receptor 6 (LGR6) (FDR = 0.0002). Moreover, we show that the GDM and 1-h glucose-associated methylation signatures in the cord blood of the infant appeared to be attenuated by the dietary and physical activity intervention during pregnancy; in the intervention arm, there were no GDM and two 1-h glucose-associated dmCpGs, whereas in the standard care arm, there were 41 GDM and 160 1-h glucose-associated dmCpGs. A total of 87% of the GDM and 77% of the 1-h glucose-associated dmCpGs had smaller effect sizes in the intervention compared to the standard care arm; the adjusted r2 for the association of LGR6 cg03566881 with GDM was 0.317 (95% confidence interval (CI) 0.012, 0.022) in the standard care and 0.240 (95% CI 0.001, 0.015) in the intervention arm. Limitations included measurement of DNA methylation in cord blood, where the functional significance of such changes are unclear, and because of the strong collinearity between treatment modality and severity of hyperglycaemia, we cannot exclude that treatment-related differences are potential confounders. CONCLUSIONS: Maternal dysglycaemia was associated with significant changes in the epigenome of the infants. Moreover, we found that the epigenetic impact of a dysglycaemic prenatal maternal environment appeared to be modified by a lifestyle intervention in pregnancy. Further research will be needed to investigate possible medical implications of the findings. TRIAL REGISTRATION: ISRCTN89971375.


Subject(s)
Diabetes, Gestational/epidemiology , Diet , Epigenome , Life Style , Adult , Diet/adverse effects , Epigenome/drug effects , Epigenome/physiology , Exercise/physiology , Female , Gestational Age , Humans , Infant , Infant, Newborn , Obesity/epidemiology , Obesity/therapy , Pregnancy
2.
Int J Obes (Lond) ; 43(5): 974-988, 2019 05.
Article in English | MEDLINE | ID: mdl-30622309

ABSTRACT

BACKGROUND: The early life environment may influence susceptibility to obesity and metabolic disease in later life through epigenetic processes. SLC6A4 is an important mediator of serotonin bioavailability, and has a key role in energy balance. We tested the hypothesis that methylation of the SLC6A4 gene predicts adiposity across the life course. METHODS: DNA methylation at 5 CpGs within the SLC6A4 gene identified from a previous methyl binding domain array was measured by pyrosequencing. We measured DNA methylation in umbilical cord (UC) from children in the Southampton Women's Survey cohort (n = 680), in peripheral blood from adolescents in the Western Australian Pregnancy Cohort Study (n = 812), and in adipose tissue from lean and obese adults from the UK BIOCLAIMS cohort (n = 81). Real-time PCR was performed to assess whether there were corresponding alterations in gene expression in the adipose tissue. RESULTS: Lower UC methylation of CpG5 was associated with higher total fat mass at 4 years (p = 0.031), total fat mass at 6-7 years (p = 0.0001) and % fat mass at 6-7 years (p = 0.004). Lower UC methylation of CpG5 was also associated with higher triceps skinfold thickness at birth (p = 0.013), 6 months (p = 0.038), 12 months (p = 0.062), 2 years (p = 0.0003), 3 years (p = 0.00004) and 6-7 years (p = 0.013). Higher maternal pregnancy weight gain (p = 0.046) and lower parity (p = 0.029) were both associated with lower SLC6A4 CpG5 methylation. In adolescents, lower methylation of CpG5 in peripheral blood was associated with greater concurrent measures of adiposity including BMI (p ≤ 0.001), waist circumference (p = 0.011), subcutaneous fat (p ≤ 0.001) and subscapular, abdominal and suprailiac skinfold thicknesses (p = 0.002, p = 0.008, p = 0.004, respectively). In adipose tissue, methylation of both SLC6A4 CpG5 (p = 0.019) and expression of SLC6A4 (p = 0.008) was lower in obese compared with lean adults. CONCLUSIONS: These data suggest that altered methylation of CpG loci within SLC6A4 may provide a robust marker of adiposity across the life course.


Subject(s)
Adiposity/genetics , DNA Methylation/physiology , Epigenesis, Genetic/physiology , Metabolic Diseases/genetics , Obesity/genetics , Serotonin Plasma Membrane Transport Proteins/metabolism , Absorptiometry, Photon , Adolescent , Adult , Australia/epidemiology , Biomarkers/metabolism , Child , Child, Preschool , Cohort Studies , DNA Methylation/genetics , Female , Gene-Environment Interaction , Genetic Predisposition to Disease , Humans , Infant, Newborn , Male , Metabolic Diseases/epidemiology , Obesity/epidemiology , Promoter Regions, Genetic/genetics
3.
Trends Genet ; 31(1): 2-4, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25547197

ABSTRACT

Some of our fate is predetermined, by genetics and by the environment in the womb. Gestational environments are reflected in the DNA methylomes of newborns, in a manner that is often influenced by genotype. Therefore, DNA methylation serves as molecular mechanism linking the interplay of early life environments and genetics to later life health. As such, methylation marks are potential biomarkers of suboptimal developmental trajectories. Can DNA methylation also be used to construct an escape route from biological fate?


Subject(s)
Epigenesis, Genetic , Biomarkers/metabolism , DNA Methylation/genetics , Genetic Determinism , Humans , Models, Genetic
4.
Cereb Cortex ; 27(5): 3080-3092, 2017 05 01.
Article in English | MEDLINE | ID: mdl-28334351

ABSTRACT

This study included 168 and 85 mother-infant dyads from Asian and United States of America cohorts to examine whether a genomic profile risk score for major depressive disorder (GPRSMDD) moderates the association between antenatal maternal depressive symptoms (or socio-economic status, SES) and fetal neurodevelopment, and to identify candidate biological processes underlying such association. Both cohorts showed a significant interaction between antenatal maternal depressive symptoms and infant GPRSMDD on the right amygdala volume. The Asian cohort also showed such interaction on the right hippocampal volume and shape, thickness of the orbitofrontal and ventromedial prefrontal cortex. Likewise, a significant interaction between SES and infant GPRSMDD was on the right amygdala and hippocampal volumes and shapes. After controlling for each other, the interaction effect of antenatal maternal depressive symptoms and GPRSMDD was mainly shown on the right amygdala, while the interaction effect of SES and GPRSMDD was mainly shown on the right hippocampus. Bioinformatic analyses suggested neurotransmitter/neurotrophic signaling, SNAp REceptor complex, and glutamate receptor activity as common biological processes underlying the influence of antenatal maternal depressive symptoms on fetal cortico-limbic development. These findings suggest gene-environment interdependence in the fetal development of brain regions implicated in cognitive-emotional function. Candidate biological mechanisms involve a range of brain region-specific signaling pathways that converge on common processes of synaptic development.


Subject(s)
Brain Mapping , Brain/growth & development , Brain/pathology , Depressive Disorder, Major/pathology , Maternal-Fetal Relations , Social Class , Asian People , Brain/diagnostic imaging , Cohort Studies , Computational Biology , Depressive Disorder, Major/diagnostic imaging , Depressive Disorder, Major/genetics , Depressive Disorder, Major/psychology , Female , Fetal Development/genetics , Gene Regulatory Networks/physiology , Genotype , Gestational Age , Humans , Image Processing, Computer-Assisted , Infant, Newborn , Magnetic Resonance Imaging , Male , Polymorphism, Single Nucleotide/genetics , Pregnancy , Prenatal Exposure Delayed Effects
5.
Dev Psychopathol ; 30(3): 891-903, 2018 08.
Article in English | MEDLINE | ID: mdl-30068421

ABSTRACT

Prenatal adversity shapes child neurodevelopment and risk for later mental health problems. The quality of the early care environment can buffer some of the negative effects of prenatal adversity on child development. Retrospective studies, in adult samples, highlight epigenetic modifications as sentinel markers of the quality of the early care environment; however, comparable data from pediatric cohorts are lacking. Participants were drawn from the Maternal Adversity Vulnerability and Neurodevelopment (MAVAN) study, a longitudinal cohort with measures of infant attachment, infant development, and child mental health. Children provided buccal epithelial samples (mean age = 6.99, SD = 1.33 years, n = 226), which were used for analyses of genome-wide DNA methylation and genetic variation. We used a series of linear models to describe the association between infant attachment and (a) measures of child outcome and (b) DNA methylation across the genome. Paired genetic data was used to determine the genetic contribution to DNA methylation at attachment-associated sites. Infant attachment style was associated with infant cognitive development (Mental Development Index) and behavior (Behavior Rating Scale) assessed with the Bayley Scales of Infant Development at 36 months. Infant attachment style moderated the effects of prenatal adversity on Behavior Rating Scale scores at 36 months. Infant attachment was also significantly associated with a principal component that accounted for 11.9% of the variation in genome-wide DNA methylation. These effects were most apparent when comparing children with a secure versus a disorganized attachment style and most pronounced in females. The availability of paired genetic data revealed that DNA methylation at approximately half of all infant attachment-associated sites was best explained by considering both infant attachment and child genetic variation. This study provides further evidence that infant attachment can buffer some of the negative effects of early adversity on measures of infant behavior. We also highlight the interplay between infant attachment and child genotype in shaping variation in DNA methylation. Such findings provide preliminary evidence for a molecular signature of infant attachment and may help inform attachment-focused early intervention programs.


Subject(s)
Child Development/physiology , DNA Methylation , Object Attachment , Social Environment , Child , Child, Preschool , Cognition , Family , Female , Genotype , Humans , Longitudinal Studies , Male , Retrospective Studies
6.
Attach Hum Dev ; 20(1): 24-42, 2018 Feb.
Article in English | MEDLINE | ID: mdl-28840781

ABSTRACT

Past research indicates that socioeconomic status (SES) accounts for differences in sensitivity across ethnic groups. However, comparatively little work has been conducted in Asia, with none examining whether ethnicity moderates the relation between SES and sensitivity. We assessed parenting behavior in 293 Singaporean citizen mothers of 6-month olds (153 Chinese, 108 Malay, 32 Indian) via the Maternal Behavioral Q-Sort for video interactions. When entered into the same model, SES (F(1,288) = 17.777, p < .001), but not ethnicity, predicted maternal sensitivity (F(2,288) = .542, p = .582). However, this positive relation between SES and sensitivity was marginally moderated by ethnicity. SES significantly positively predicted sensitivity in Chinese, but not Malay dyads. Within Indian dyads, SES marginally positively predicted sensitivity only when permanent residents were included in analyses. We discuss the importance of culture on perceived SES-associated stress. However, because few university-educated Malays participated, we also consider whether university education, specifically, positively influences sensitivity.


Subject(s)
Mother-Child Relations/ethnology , Mothers , Parenting/ethnology , Socioeconomic Factors , Adult , China/ethnology , Cultural Characteristics , Educational Status , Female , Humans , Income , India/ethnology , Infant , Malaysia/ethnology , Male , Object Attachment , Singapore/epidemiology
7.
Genome Res ; 24(7): 1064-74, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24709820

ABSTRACT

Integrating the genotype with epigenetic marks holds the promise of better understanding the biology that underlies the complex interactions of inherited and environmental components that define the developmental origins of a range of disorders. The quality of the in utero environment significantly influences health over the lifecourse. Epigenetics, and in particular DNA methylation marks, have been postulated as a mechanism for the enduring effects of the prenatal environment. Accordingly, neonate methylomes contain molecular memory of the individual in utero experience. However, interindividual variation in methylation can also be a consequence of DNA sequence polymorphisms that result in methylation quantitative trait loci (methQTLs) and, potentially, the interaction between fixed genetic variation and environmental influences. We surveyed the genotypes and DNA methylomes of 237 neonates and found 1423 punctuate regions of the methylome that were highly variable across individuals, termed variably methylated regions (VMRs), against a backdrop of homogeneity. MethQTLs were readily detected in neonatal methylomes, and genotype alone best explained ∼25% of the VMRs. We found that the best explanation for 75% of VMRs was the interaction of genotype with different in utero environments, including maternal smoking, maternal depression, maternal BMI, infant birth weight, gestational age, and birth order. Our study sheds new light on the complex relationship between biological inheritance as represented by genotype and individual prenatal experience and suggests the importance of considering both fixed genetic variation and environmental factors in interpreting epigenetic variation.


Subject(s)
DNA Methylation , Environment , Epigenesis, Genetic , Gene-Environment Interaction , Genetic Heterogeneity , Genotype , Transcriptome , Computational Biology/methods , CpG Islands , Epigenomics/methods , Female , Humans , Infant, Newborn , Male , Polymorphism, Single Nucleotide , Pregnancy , Quantitative Trait Loci , Risk Factors
8.
BMC Med ; 15(1): 50, 2017 03 07.
Article in English | MEDLINE | ID: mdl-28264723

ABSTRACT

BACKGROUND: Obesity is an escalating health problem worldwide, and hence the causes underlying its development are of primary importance to public health. There is growing evidence that suboptimal intrauterine environment can perturb the metabolic programing of the growing fetus, thereby increasing the risk of developing obesity in later life. However, the link between early exposures in the womb, genetic susceptibility, and perturbed epigenome on metabolic health is not well understood. In this study, we shed more light on this aspect by performing a comprehensive analysis on the effects of variation in prenatal environment, neonatal methylome, and genotype on birth weight and adiposity in early childhood. METHODS: In a prospective mother-offspring cohort (N = 987), we interrogated the effects of 30 variables that influence the prenatal environment, umbilical cord DNA methylation, and genotype on offspring weight and adiposity, over the period from birth to 48 months. This is an interim analysis on an ongoing cohort study. RESULTS: Eleven of 30 prenatal environments, including maternal adiposity, smoking, blood glucose and plasma unsaturated fatty acid levels, were associated with birth weight. Polygenic risk scores derived from genetic association studies on adult adiposity were also associated with birth weight and child adiposity, indicating an overlap between the genetic pathways influencing metabolic health in early and later life. Neonatal methylation markers from seven gene loci (ANK3, CDKN2B, CACNA1G, IGDCC4, P4HA3, ZNF423 and MIRLET7BHG) were significantly associated with birth weight, with a subset of these in genes previously implicated in metabolic pathways in humans and in animal models. Methylation levels at three of seven birth weight-linked loci showed significant association with prenatal environment, but none were affected by polygenic risk score. Six of these birth weight-linked loci continued to show a longitudinal association with offspring size and/or adiposity in early childhood. CONCLUSIONS: This study provides further evidence that developmental pathways to adiposity begin before birth and are influenced by environmental, genetic and epigenetic factors. These pathways can have a lasting effect on offspring size, adiposity and future metabolic outcomes, and offer new opportunities for risk stratification and prevention of obesity. CLINICAL TRIAL REGISTRATION: This birth cohort is a prospective observational study, designed to study the developmental origins of health and disease, and was retrospectively registered on 1 July 2010 under the identifier NCT01174875 .


Subject(s)
Adiposity/genetics , DNA Methylation , Obesity/genetics , Birth Weight , Environment , Epigenomics , Female , Genome-Wide Association Study , Genotype , Humans , Infant, Newborn , Obesity/complications , Prospective Studies
9.
BMC Med ; 15(1): 211, 2017 12 05.
Article in English | MEDLINE | ID: mdl-29202839

ABSTRACT

BACKGROUND: Epigenomes are tissue specific and thus the choice of surrogate tissue can play a critical role in interpreting neonatal epigenome-wide association studies (EWAS) and in their extrapolation to target tissue. To develop a better understanding of the link between tissue specificity and neonatal EWAS, and the contributions of genotype and prenatal factors, we compared genome-wide DNA methylation of cord tissue and cord blood, two of the most accessible surrogate tissues at birth. METHODS: In 295 neonates, DNA methylation was profiled using Infinium HumanMethylation450 beadchip arrays. Sites of inter-individual variability in DNA methylation were mapped and compared across the two surrogate tissues at birth, i.e., cord tissue and cord blood. To ascertain the similarity to target tissues, DNA methylation profiles of surrogate tissues were compared to 25 primary tissues/cell types mapped under the Epigenome Roadmap project. Tissue-specific influences of genotype on the variable CpGs were also analyzed. Finally, to interrogate the impact of the in utero environment, EWAS on 45 prenatal factors were performed and compared across the surrogate tissues. RESULTS: Neonatal EWAS results were tissue specific. In comparison to cord blood, cord tissue showed higher inter-individual variability in the epigenome, with a lower proportion of CpGs influenced by genotype. Both neonatal tissues were good surrogates for target tissues of mesodermal origin. They also showed distinct phenotypic associations, with effect sizes of the overlapping CpGs being in the same order of magnitude. CONCLUSIONS: The inter-relationship between genetics, prenatal factors and epigenetics is tissue specific, and requires careful consideration in designing and interpreting future neonatal EWAS. TRIAL REGISTRATION: This birth cohort is a prospective observational study, designed to study the developmental origins of health and disease, and was retrospectively registered on 1 July 2010 under the identifier NCT01174875 .


Subject(s)
DNA Methylation , Fetal Blood , Genome-Wide Association Study , Umbilical Cord , CpG Islands , DNA , Epigenesis, Genetic , Female , Genotype , Gestational Age , Humans , Infant, Newborn , Male , Pregnancy , Prospective Studies , Young Adult
10.
Brain Cogn ; 116: 17-28, 2017 08.
Article in English | MEDLINE | ID: mdl-28582665

ABSTRACT

Despite claims concerning biological mechanisms sub-serving infant attention, little experimental work examines its underpinnings. This study examines how candidate polymorphisms from the cholinergic (CHRNA4 rs1044396) and dopaminergic (COMT rs4680) systems, respectively indicative of parietal and prefrontal/anterior cingulate involvement, are related to 6-month-olds' (n=217) performance during a visual expectation eye-tracking paradigm. As previous studies suggest that both cholinergic and dopaminergic genes may influence susceptibility to the influence of other genetic and environmental factors, we further examined whether these candidate genes interact with one another and/or with early caregiving experience in predicting infants' visual attention. We detected an interaction between CHRNA4 genotype and observed maternal sensitivity upon infants' orienting to random stimuli and a CHRNA4-COMT interaction effect upon infants' orienting to patterned stimuli. Consistent with adult research, we observed a direct effect of COMT genotype on anticipatory looking to patterned stimuli. Findings suggest that CHRNA4 genotype may influence susceptibility to other attention-related factors in infancy. These interactions may account for the inability to establish a link between CHRNA4 and orienting in infant research to date, despite developmental theorizing suggesting otherwise. Moreover, findings suggest that by 6months, dopamine, and relatedly, the prefrontal cortex/anterior cingulate, may be important to infant attention.


Subject(s)
Attention/physiology , Catechol O-Methyltransferase/genetics , Child Development/physiology , Executive Function/physiology , Gene-Environment Interaction , Maternal Behavior/physiology , Orientation, Spatial/physiology , Receptors, Nicotinic/genetics , Adult , Female , Humans , Infant , Male
11.
BMC Bioinformatics ; 17: 299, 2016 Aug 02.
Article in English | MEDLINE | ID: mdl-27480116

ABSTRACT

BACKGROUND: The interplay among genetic, environment and epigenetic variation is not fully understood. Advances in high-throughput genotyping methods, high-density DNA methylation detection and well-characterized sample collections, enable epigenetic association studies at the genomic and population levels (EWAS). The field has extended to interrogate the interaction of environmental and genetic (GxE) influences on epigenetic variation. Also, the detection of methylation quantitative trait loci (methQTLs) and their association with health status has enhanced our knowledge of epigenetic mechanisms in disease trajectory. However analysis of this type of data brings computational challenges and there are few practical solutions to enable large scale studies in standard computational environments. RESULTS: GEM is a highly efficient R tool suite for performing epigenome wide association studies (EWAS). GEM provides three major functions named GEM_Emodel, GEM_Gmodel and GEM_GxEmodel to study the interplay of Gene, Environment and Methylation (GEM). Within GEM, the pre-existing "Matrix eQTL" package is utilized and extended to study methylation quantitative trait loci (methQTL) and the interaction of genotype and environment (GxE) to determine DNA methylation variation, using matrix based iterative correlation and memory-efficient data analysis. Benchmarking presented here on a publicly available dataset, demonstrated that GEM can facilitate reliable genome-wide methQTL and GxE analysis on a standard laptop computer within minutes. CONCLUSIONS: The GEM package facilitates efficient EWAS study in large cohorts. It is written in R code and can be freely downloaded from Bioconductor at https://www.bioconductor.org/packages/GEM/ .


Subject(s)
DNA Methylation , Epigenomics , Genetic Association Studies , Algorithms , Genomics , Genotype , Internet , Quantitative Trait Loci , User-Computer Interface
12.
PLoS Genet ; 9(6): e1003515, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23754950

ABSTRACT

Down syndrome (DS), commonly caused by an extra copy of chromosome 21 (chr21), occurs in approximately one out of 700 live births. Precisely how an extra chr21 causes over 80 clinically defined phenotypes is not yet clear. Reduced representation bisulfite sequencing (RRBS) analysis at single base resolution revealed DNA hypermethylation in all autosomes in DS samples. We hypothesize that such global hypermethylation may be mediated by down-regulation of TET family genes involved in DNA demethylation, and down-regulation of REST/NRSF involved in transcriptional and epigenetic regulation. Genes located on chr21 were up-regulated by an average of 53% in DS compared to normal villi, while genes with promoter hypermethylation were modestly down-regulated. DNA methylation perturbation was conserved in DS placenta villi and in adult DS peripheral blood leukocytes, and enriched for genes known to be causally associated with DS phenotypes. Our data suggest that global epigenetic changes may occur early in development and contribute to DS phenotypes.


Subject(s)
DNA Methylation/genetics , Down Syndrome/genetics , Epigenesis, Genetic/genetics , Placenta/metabolism , Chromosomes, Human, Pair 21/genetics , CpG Islands/genetics , DNA-Binding Proteins/genetics , Dioxygenases , Down Syndrome/metabolism , Female , Gene Expression Regulation , Humans , Male , Mixed Function Oxygenases , Placenta/cytology , Pregnancy , Promoter Regions, Genetic , Proto-Oncogene Proteins/genetics , Repressor Proteins/genetics , Sequence Analysis, DNA
13.
Dev Psychopathol ; 27(1): 137-50, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25640836

ABSTRACT

Early life environments interact with genotype to determine stable phenotypic outcomes. Here we examined the influence of a variant in the brain-derived neurotropic factor (BDNF) gene (Val66Met), which underlies synaptic plasticity throughout the central nervous system, on the degree to which antenatal maternal anxiety associated with neonatal DNA methylation. We also examined the association between neonatal DNA methylation and brain substructure volume, as a function of BDNF genotype. Infant, but not maternal, BDNF genotype dramatically influences the association of antenatal anxiety on the epigenome at birth as well as that between the epigenome and neonatal brain structure. There was a greater impact of antenatal maternal anxiety on the DNA methylation of infants with the methionine (Met)/Met compared to both Met/valine (Val) and Val/Val genotypes. There were significantly more cytosine-phosphate-guanine sites where methylation levels covaried with right amygdala volume among Met/Met compared with both Met/Val and Val/Val carriers. In contrast, more cytosine-phosphate-guanine sites covaried with left hippocampus volume in Val/Val infants compared with infants of the Met/Val or Met/Met genotype. Thus, antenatal Maternal Anxiety × BDNF Val66Met Polymorphism interactions at the level of the epigenome are reflected differently in the structure of the amygdala and the hippocampus. These findings suggest that BDNF genotype regulates the sensitivity of the methylome to early environment and that differential susceptibility to specific environmental conditions may be both tissue and function specific.


Subject(s)
Amygdala/anatomy & histology , Anxiety/metabolism , Brain-Derived Neurotrophic Factor/genetics , DNA Methylation/genetics , Epigenesis, Genetic/genetics , Gene-Environment Interaction , Hippocampus/anatomy & histology , Adult , Amygdala/pathology , Anxiety/complications , Female , Genotype , Hippocampus/pathology , Humans , Infant, Newborn , Magnetic Resonance Imaging , Male , Polymorphism, Genetic , Pregnancy , Singapore
14.
J Hum Genet ; 59(9): 504-11, 2014 Sep.
Article in English | MEDLINE | ID: mdl-25102097

ABSTRACT

CXCL14 is a chemokine that has previously been implicated in insulin resistance in mice. In humans, the role of CXCL14 in metabolic processes is not well established, and we sought to determine whether CXCL14 is a risk susceptibility gene important in fetal programming of metabolic disease. For this purpose, we investigated whether CXCL14 is differentially regulated in human umbilical cords of infants with varying birth weights. We found an elevated expression of CXCL14 in human low birth weight (LBW) cords, as well as in cords from nutritionally restricted Macaca fascicularis macaques. To further analyze the regulatory mechanisms underlying the expression of CXCL14, we examined CXCL14 in umbilical cord-derived mesenchymal stem cells (MSCs) that provide an in vitro cell-based system amenable to experimental manipulation. Using both whole frozen cords and MSCs, we determined that site-specific CpG methylation in the CXCL14 promoter is associated with altered expression, and that changes in methylation are evident in LBW infant-derived umbilical cords that may indicate future metabolic compromise through CXCL14.


Subject(s)
Chemokines, CXC/genetics , DNA Methylation , Gene Expression Profiling , Infant, Low Birth Weight/metabolism , Adult , Animals , Caloric Restriction , Cells, Cultured , CpG Islands/genetics , Female , Humans , Infant, Newborn , Macaca fascicularis/genetics , Male , Maternal Age , Mesenchymal Stem Cells/metabolism , Pregnancy , Promoter Regions, Genetic/genetics , Reverse Transcriptase Polymerase Chain Reaction , Umbilical Cord/cytology , Umbilical Cord/metabolism
15.
Hum Reprod ; 29(10): 2287-301, 2014 Oct 10.
Article in English | MEDLINE | ID: mdl-25129543

ABSTRACT

STUDY QUESTION: Are molecular pathways reflecting the biology of small for gestational age (SGA) neonates preserved in umbilical cord-derived mesenchymal stem cells (MSCs)? SUMMARY ANSWER: MSCs from SGA newborns were found to express an altered EGR-1-dependent gene network involved in the regulation of cell proliferation and oxidative stress. WHAT IS KNOWN ALREADY: Individuals with suboptimal intrauterine development are at greater risk of metabolic diseases such as type II diabetes, obesity and cardiovascular disease. STUDY DESIGN, SIZE, DURATION: Umbilical cords (n = 283) from the GUSTO (growing up in Singapore towards healthy outcomes) birth cohort study, and primary MSC isolates established from SGA and matched control cases (n = 6 per group), were subjected to gene expression analysis and candidate genes were studied for functional validation. PARTICIPANTS/MATERIALS, SETTING, METHODS: Umbilical cord specimens were derived from babies born at the National University Hospital (NUH) in Singapore. Local ethical approval was obtained. MSC isolates were established in Wharton's jelly and molecular analysis was conducted by gene expression microarrays and RT-PCR. Cells from SGA and control groups were compared in the presence and absence of insulin and candidate gene function was studied via siRNA-mediated gene knockdown and over-expression experiments in MSCs. MAIN RESULTS AND THE ROLE OF CHANCE: Using repeated measure ANOVAs, proliferation rates of MSCs isolated from SGA neonates were found to be significantly increased (P < 0.01). In the absence of insulin, EGR-1 levels were found to be significantly reduced in the group of SGA-derived MSCs, whereas EGR-1 expression was found to be up-regulated in the same group in the presence of insulin (P < 0.01). EGR-1 was found to induce expression of COX-2 in the SGA group (P < 0.01) and both, EGR-1 and COX-2 stimulated glucose uptake in MSCs (P < 0.01). EGR-1 and COX-2 levels were associated in whole umbilical cords (n = 283, P < 0.01) and EGR-1 positively correlated with abdominal circumference and birthweight (n = 91, P < 0.01 and n = 91, P < 0.01). LIMITATIONS, REASONS FOR CAUTION: Cell models may not entirely reflect the physiology of the host and patient follow-up studies will be necessary for further clinical validation. WIDER IMPLICATIONS OF THE FINDINGS: Our study suggests that Wharton's jelly-derived MSCs are useful in identifying pathways specific for fetal growth restriction. STUDY FUNDING/COMPETING INTERESTS: This work is supported by the Translational Clinical Research (TCR) Flagship Program on Developmental Pathways to Metabolic Disease funded by the National Research Foundation (NRF) and administered by the National Medical Research Council (NMRC), Singapore- NMRC/TCR/004-NUS/2008'. SICS Investigators are supported through the Agency for Science Technology and Research (A*STAR) funding. No potential conflicts of interest relevant to this article were reported.


Subject(s)
Fetal Development , Infant, Small for Gestational Age/metabolism , Mesenchymal Stem Cells/metabolism , Wharton Jelly/cytology , Cell Proliferation/genetics , Cyclooxygenase 2/genetics , Cyclooxygenase 2/metabolism , Early Growth Response Protein 1/genetics , Early Growth Response Protein 1/metabolism , Gene Expression Profiling , Gene Expression Regulation, Developmental , Gene Regulatory Networks , Glucose/metabolism , Humans , Infant, Newborn , Oxidative Stress/genetics , Umbilical Cord/cytology , Umbilical Cord/metabolism , Wharton Jelly/metabolism
16.
BMC Pregnancy Childbirth ; 14: 345, 2014 Oct 02.
Article in English | MEDLINE | ID: mdl-25273851

ABSTRACT

BACKGROUND: Universal and high-risk screening for gestational diabetes mellitus (GDM) has been widely studied and debated. Few studies have assessed GDM screening in Asian populations and even fewer have compared Asian ethnic groups in a single multi-ethnic population. METHODS: 1136 pregnant women (56.7% Chinese, 25.5% Malay and 17.8% Indian) from the Growing Up in Singapore Towards healthy Outcomes (GUSTO) birth cohort study were screened for GDM by 75-g oral glucose tolerance test (OGTT) at 26-28 weeks of gestation. GDM was defined using the World Health Organization (WHO) criteria. High-risk screening is based on the guidelines of the UK National Institute for Health and Clinical Excellence. RESULTS: Universal screening detected significantly more cases than high-risk screening [crude OR 2.2 (95% CI 1.7-2.8)], particularly for Chinese women [crude OR = 3.5 (95% CI 2.5-5.0)]. Pre-pregnancy BMI > 30 kg/m2 (adjusted OR = 3.4, 95% CI 1.5-7.9) and previous GDM history (adjusted OR = 6.6, 95% CI 1.2-37.3) were associated with increased risk of GDM in Malay women while GDM history was the only significant risk factor for GDM in Chinese women (adjusted OR = 4.7, 95% CI 2.0-11.0). CONCLUSION: Risk factors used in high-risk screening do not sufficiently predict GDM risk and failed to detect half the GDM cases in Asian women. Asian women, particularly Chinese, should be screened to avoid under-diagnosis of GDM and thereby optimize maternal and fetal outcomes.


Subject(s)
Diabetes, Gestational/diagnosis , Diabetes, Gestational/ethnology , Mass Screening , Adult , Body Mass Index , China/ethnology , Cohort Studies , Diabetes, Gestational/genetics , Female , Glucose Tolerance Test , Humans , India/ethnology , Malaysia/ethnology , Practice Guidelines as Topic , Pregnancy , Prevalence , Reproductive History , Risk Factors , Sensitivity and Specificity , Singapore/epidemiology , Young Adult
17.
Dev Psychopathol ; 26(4 Pt 2): 1255-66, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25422959

ABSTRACT

Multiple behavioral and health outcomes, including internalizing symptoms, may be predicted from prenatal maternal anxiety, depression, or stress. However, not all children are affected, and those that are can be affected in different ways. Here we test the hypothesis that the effects of prenatal anxiety are moderated by genetic variation in the child's brain-derived neurotrophic factor (BDNF) gene, using the Avon Longitudinal Study of Parents and Children population cohort. Internalizing symptoms were assessed from 4 to 13 years of age using the Strengths and Difficulties Questionnaire (n = 8,584); a clinical interview with the adolescents was conducted at age 15 years (n = 4,704). Obstetric and psychosocial risk and postnatal maternal symptoms were included as covariates. Results show that prenatal maternal anxiety predicted internalizing symptoms, including with the diagnostic assessment at 15 years. There was a main effect of two BDNF polymorphisms (rs6265 [val66met] and rs11030104) on internalizing symptoms up to age 13. There was also genetic moderation of the prenatal anxiety effect by different BDNF polymorphisms (rs11030121 and rs7124442), although significant effects were limited to preadolescence. The findings suggest a role for BDNF gene-environment interactions in individual vulnerability to the effects of prenatal anxiety on child internalizing symptoms.


Subject(s)
Anxiety/epidemiology , Behavioral Symptoms , Brain-Derived Neurotrophic Factor/genetics , Gene-Environment Interaction , Pregnancy Complications/epidemiology , Adolescent , Behavioral Symptoms/epidemiology , Behavioral Symptoms/etiology , Behavioral Symptoms/genetics , Child , Child, Preschool , England/epidemiology , Female , Genotype , Humans , Longitudinal Studies , Pregnancy
18.
Ann Nutr Metab ; 64(3-4): 218-25, 2014.
Article in English | MEDLINE | ID: mdl-25300263

ABSTRACT

BACKGROUND: The dramatic emergence of noncommunicable diseases (NCD) in Asia, albeit with ethnic variation, has coincided with the rapid socioeconomic and nutritional transition taking place in the region, with the prevalence of diabetes rising 5-fold in Singapore in less than 4 decades. The Growing Up in Singapore Towards healthy Outcomes (GUSTO) cohort study recruited 1,247 expectant mothers of Chinese, Malay, or Indian ethnicity in their first trimester, with detailed longitudinal tracking--through the antenatal period, birth, and the child's first 4 years of life--to examine the potential roles of fetal, developmental, and epigenetic factors in early pathways to metabolic and neurodevelopmental outcomes. KEY MESSAGES: A number of findings with a translational and clinical focus have already emerged. In the mothers, we found that changes and differences in food consumption varied across ethnic groups, with persistence of traditional beliefs, during pregnancy and the postpartum period. During pregnancy, higher maternal glucose levels, even in the absence of gestational diabetes mellitus, had graded relations with infant adiposity. Relations between maternal emotional health and birth outcomes and neurodevelopment have been identified. Genotype (25%) and in particular gene × environment interactions (75%) shape interindividual variations in the DNA methylome at birth. The complex effects of fixed genetic variations and different in utero environments can influence the epigenetic status at birth and the later-life phenotype. CONCLUSIONS: The richness of the clinical data in 3 ethnicities, the extent of the biospecimen collection, and the extensive infancy and preschool follow-up have allowed us to study the biological pathways that link fetal development to health outcomes. In the coming years, more sophisticated analyses of epigenotype-phenotype relationships will become possible as the children grow and develop. Our studies will lead to the development of clinical and population-based interventions to reduce the burden of NCD.


Subject(s)
Child Development , Child Nutritional Physiological Phenomena , Diet , Fetal Development , Health Promotion , Health Status , Maternal Nutritional Physiological Phenomena , Child Nutritional Physiological Phenomena/ethnology , Child, Preschool , China/ethnology , Cohort Studies , Diet/adverse effects , Diet/ethnology , Epigenesis, Genetic , Female , Humans , India/ethnology , Infant , Infant Nutritional Physiological Phenomena/ethnology , Infant, Newborn , Longitudinal Studies , Malaysia/ethnology , Male , Maternal Nutritional Physiological Phenomena/ethnology , Pregnancy , Singapore
19.
Nat Biotechnol ; 41(10): 1457-1464, 2023 Oct.
Article in English | MEDLINE | ID: mdl-36747096

ABSTRACT

DNA comprises molecular information stored in genetic and epigenetic bases, both of which are vital to our understanding of biology. Most DNA sequencing approaches address either genetics or epigenetics and thus capture incomplete information. Methods widely used to detect epigenetic DNA bases fail to capture common C-to-T mutations or distinguish 5-methylcytosine from 5-hydroxymethylcytosine. We present a single base-resolution sequencing methodology that sequences complete genetics and the two most common cytosine modifications in a single workflow. DNA is copied and bases are enzymatically converted. Coupled decoding of bases across the original and copy strand provides a phased digital readout. Methods are demonstrated on human genomic DNA and cell-free DNA from a blood sample of a patient with cancer. The approach is accurate, requires low DNA input and has a simple workflow and analysis pipeline. Simultaneous, phased reading of genetic and epigenetic bases provides a more complete picture of the information stored in genomes and has applications throughout biomedicine.

20.
Epigenetics ; 17(6): 653-664, 2022 06.
Article in English | MEDLINE | ID: mdl-34461806

ABSTRACT

DNA methylation (DNAm) in mammals is mostly examined within the context of CpG dinucleotides. Non-CpG DNAm is also widespread across the human genome, but the functional relevance, tissue-specific disposition, and inter-individual variability has not been widely studied. Our aim was to examine non-CpG DNAm in the wider methylome across multiple tissues from the same individuals to better understand non-CpG DNAm distribution within different tissues and individuals and in relation to known genomic regulatory features.DNA methylation in umbilical cord and cord blood at birth, and peripheral venous blood at age 12-13 y from 20 individuals from the Southampton Women's Survey cohort was assessed by Agilent SureSelect methyl-seq. Hierarchical cluster analysis (HCA) was performed on CpG and non-CpG sites and stratified by specific cytosine environment. Analysis of tissue and inter-individual variation was then conducted in a second dataset of 12 samples: eight muscle tissues, and four aliquots of cord blood pooled from two individuals.HCA using methylated non-CpG sites showed different clustering patterns specific to the three base-pair triplicate (CNN) sequence. Analysis of CAC sites with non-zero methylation showed that samples clustered first by tissue type, then by individual (as observed for CpG methylation), while analysis using non-zero methylation at CAT sites showed samples grouped predominantly by individual. These clustering patterns were validated in an independent dataset using cord blood and muscle tissue.This research suggests that CAC methylation can have tissue-specific patterns, and that individual effects, either genetic or unmeasured environmental factors, can influence CAT methylation.


Subject(s)
DNA Methylation , Individuality , Animals , CpG Islands , Cytosine , DNA , Female , Genome, Human , Humans , Mammals/genetics
SELECTION OF CITATIONS
SEARCH DETAIL