Home > Research > Publications & Outputs > Childhood DNA methylation as a marker of early ...

Links

Text available via DOI:

View graph of relations

Childhood DNA methylation as a marker of early life rapid weight gain and subsequent overweight

Research output: Contribution to Journal/MagazineJournal articlepeer-review

Published

Standard

Childhood DNA methylation as a marker of early life rapid weight gain and subsequent overweight. / Robinson, N.; Brown, H.; Antoun, Elie et al.
In: Clinical Epigenetics, Vol. 13, No. 1, 8, 31.12.2021.

Research output: Contribution to Journal/MagazineJournal articlepeer-review

Harvard

Robinson, N, Brown, H, Antoun, E, Godfrey, KM, Hanson, MA, Lillycrop, KA, Crozier, SR, Murray, R, Pearce, MS, Relton, CL, Albani, V & McKay, JA 2021, 'Childhood DNA methylation as a marker of early life rapid weight gain and subsequent overweight', Clinical Epigenetics, vol. 13, no. 1, 8. https://doi.org/10.1186/s13148-020-00952-z

APA

Robinson, N., Brown, H., Antoun, E., Godfrey, K. M., Hanson, M. A., Lillycrop, K. A., Crozier, S. R., Murray, R., Pearce, M. S., Relton, C. L., Albani, V., & McKay, J. A. (2021). Childhood DNA methylation as a marker of early life rapid weight gain and subsequent overweight. Clinical Epigenetics, 13(1), Article 8. https://doi.org/10.1186/s13148-020-00952-z

Vancouver

Robinson N, Brown H, Antoun E, Godfrey KM, Hanson MA, Lillycrop KA et al. Childhood DNA methylation as a marker of early life rapid weight gain and subsequent overweight. Clinical Epigenetics. 2021 Dec 31;13(1):8. Epub 2021 Jan 12. doi: 10.1186/s13148-020-00952-z

Author

Robinson, N. ; Brown, H. ; Antoun, Elie et al. / Childhood DNA methylation as a marker of early life rapid weight gain and subsequent overweight. In: Clinical Epigenetics. 2021 ; Vol. 13, No. 1.

Bibtex

@article{568a8f6e0620455697d082974416756c,
title = "Childhood DNA methylation as a marker of early life rapid weight gain and subsequent overweight",
abstract = "Background: High early postnatal weight gain has been associated with childhood adiposity; however, the mechanism remains unknown. DNA methylation is a hypothesised mechanism linking early life exposures and subsequent disease. However, epigenetic changes associated with high early weight gain have not previously been investigated. Our aim was to investigate the associations between early weight gain, peripheral blood DNA methylation, and subsequent overweight/obese. Data from the UK Avon Longitudinal study of Parents and Children (ALSPAC) cohort were used to estimate associations between early postnatal weight gain and epigenome-wide DNA CpG site methylation (Illumina 450 K Methylation Beadchip) in blood in childhood (n = 125) and late adolescence (n = 96). High weight gain in the first year (a change in weight z‐scores > 0.67), both unconditional (rapid weight gain) and conditional on birthweight (rapid thrive), was related to individual CpG site methylation and across regions using the meffil pipeline, with and without adjustment for cell type proportions, and with 5% false discovery rate correction. Variation in methylation at high weight gain-associated CpG sites was then examined with regard to body composition measures in childhood and adolescence. Replication of the differentially methylated CpG sites was sought using whole-blood DNA samples from 104 children from the UK Southampton Women{\textquoteright}s Survey. Results: Rapid infant weight gain was associated with small (+ 1% change) increases in childhood methylation (age 7) for two distinct CpG sites (cg01379158 (NT5M) and cg11531579 (CHFR)). Childhood methylation at one of these CpGs (cg11531579) was also higher in those who experienced rapid weight gain and were subsequently overweight/obese in adolescence (age 17). Rapid weight gain was not associated with differential DNA methylation in adolescence. Childhood methylation at the cg11531579 site was also suggestively associated with rapid weight gain in the replication cohort. Conclusions: This study identified associations between rapid weight gain in infancy and small increases in childhood methylation at two CpG sites, one of which was replicated and was also associated with subsequent overweight/obese. It will be important to determine whether loci are markers of early rapid weight gain across different, larger populations. The mechanistic relevance of these differentially methylated sites requires further investigation.",
keywords = "ALSPAC, Conditional weight gain, DNA methylation, DOHAD, Epigenetics, EWAS, Rapid weight gain, SWS",
author = "N. Robinson and H. Brown and Elie Antoun and Godfrey, {Keith M.} and Hanson, {Mark A.} and Lillycrop, {Karen A.} and Crozier, {Sarah R.} and Robert Murray and Pearce, {M. S.} and Relton, {C. L.} and V. Albani and McKay, {J. A.}",
year = "2021",
month = dec,
day = "31",
doi = "10.1186/s13148-020-00952-z",
language = "English",
volume = "13",
journal = "Clinical Epigenetics",
issn = "1868-7075",
publisher = "BioMed Central Ltd.",
number = "1",

}

RIS

TY - JOUR

T1 - Childhood DNA methylation as a marker of early life rapid weight gain and subsequent overweight

AU - Robinson, N.

AU - Brown, H.

AU - Antoun, Elie

AU - Godfrey, Keith M.

AU - Hanson, Mark A.

AU - Lillycrop, Karen A.

AU - Crozier, Sarah R.

AU - Murray, Robert

AU - Pearce, M. S.

AU - Relton, C. L.

AU - Albani, V.

AU - McKay, J. A.

PY - 2021/12/31

Y1 - 2021/12/31

N2 - Background: High early postnatal weight gain has been associated with childhood adiposity; however, the mechanism remains unknown. DNA methylation is a hypothesised mechanism linking early life exposures and subsequent disease. However, epigenetic changes associated with high early weight gain have not previously been investigated. Our aim was to investigate the associations between early weight gain, peripheral blood DNA methylation, and subsequent overweight/obese. Data from the UK Avon Longitudinal study of Parents and Children (ALSPAC) cohort were used to estimate associations between early postnatal weight gain and epigenome-wide DNA CpG site methylation (Illumina 450 K Methylation Beadchip) in blood in childhood (n = 125) and late adolescence (n = 96). High weight gain in the first year (a change in weight z‐scores > 0.67), both unconditional (rapid weight gain) and conditional on birthweight (rapid thrive), was related to individual CpG site methylation and across regions using the meffil pipeline, with and without adjustment for cell type proportions, and with 5% false discovery rate correction. Variation in methylation at high weight gain-associated CpG sites was then examined with regard to body composition measures in childhood and adolescence. Replication of the differentially methylated CpG sites was sought using whole-blood DNA samples from 104 children from the UK Southampton Women’s Survey. Results: Rapid infant weight gain was associated with small (+ 1% change) increases in childhood methylation (age 7) for two distinct CpG sites (cg01379158 (NT5M) and cg11531579 (CHFR)). Childhood methylation at one of these CpGs (cg11531579) was also higher in those who experienced rapid weight gain and were subsequently overweight/obese in adolescence (age 17). Rapid weight gain was not associated with differential DNA methylation in adolescence. Childhood methylation at the cg11531579 site was also suggestively associated with rapid weight gain in the replication cohort. Conclusions: This study identified associations between rapid weight gain in infancy and small increases in childhood methylation at two CpG sites, one of which was replicated and was also associated with subsequent overweight/obese. It will be important to determine whether loci are markers of early rapid weight gain across different, larger populations. The mechanistic relevance of these differentially methylated sites requires further investigation.

AB - Background: High early postnatal weight gain has been associated with childhood adiposity; however, the mechanism remains unknown. DNA methylation is a hypothesised mechanism linking early life exposures and subsequent disease. However, epigenetic changes associated with high early weight gain have not previously been investigated. Our aim was to investigate the associations between early weight gain, peripheral blood DNA methylation, and subsequent overweight/obese. Data from the UK Avon Longitudinal study of Parents and Children (ALSPAC) cohort were used to estimate associations between early postnatal weight gain and epigenome-wide DNA CpG site methylation (Illumina 450 K Methylation Beadchip) in blood in childhood (n = 125) and late adolescence (n = 96). High weight gain in the first year (a change in weight z‐scores > 0.67), both unconditional (rapid weight gain) and conditional on birthweight (rapid thrive), was related to individual CpG site methylation and across regions using the meffil pipeline, with and without adjustment for cell type proportions, and with 5% false discovery rate correction. Variation in methylation at high weight gain-associated CpG sites was then examined with regard to body composition measures in childhood and adolescence. Replication of the differentially methylated CpG sites was sought using whole-blood DNA samples from 104 children from the UK Southampton Women’s Survey. Results: Rapid infant weight gain was associated with small (+ 1% change) increases in childhood methylation (age 7) for two distinct CpG sites (cg01379158 (NT5M) and cg11531579 (CHFR)). Childhood methylation at one of these CpGs (cg11531579) was also higher in those who experienced rapid weight gain and were subsequently overweight/obese in adolescence (age 17). Rapid weight gain was not associated with differential DNA methylation in adolescence. Childhood methylation at the cg11531579 site was also suggestively associated with rapid weight gain in the replication cohort. Conclusions: This study identified associations between rapid weight gain in infancy and small increases in childhood methylation at two CpG sites, one of which was replicated and was also associated with subsequent overweight/obese. It will be important to determine whether loci are markers of early rapid weight gain across different, larger populations. The mechanistic relevance of these differentially methylated sites requires further investigation.

KW - ALSPAC

KW - Conditional weight gain

KW - DNA methylation

KW - DOHAD

KW - Epigenetics

KW - EWAS

KW - Rapid weight gain

KW - SWS

U2 - 10.1186/s13148-020-00952-z

DO - 10.1186/s13148-020-00952-z

M3 - Journal article

C2 - 33436068

AN - SCOPUS:85099246961

VL - 13

JO - Clinical Epigenetics

JF - Clinical Epigenetics

SN - 1868-7075

IS - 1

M1 - 8

ER -