Skip to main content
Communications Biology logoLink to Communications Biology
. 2025 May 24;8:796. doi: 10.1038/s42003-025-08121-9

Father’s adolescent body silhouette is associated with offspring asthma, lung function and BMI through DNA methylation

Negusse Tadesse Kitaba 1, Toril Mørkve Østergaard 2, Marianne Lønnebotn 3, Simone Accordini 4, Francisco Gómez Real 5, Andrei Malinovschi 6, Anna Oudin 7, Bryndis Benediktsdottir 8, Francisco Javier Callejas González 9, Leopoldo Palacios Gómez 10, Mathias Holm 11, Nils Oskar Jõgi 6, Shyamali C Dharmage 12, Svein Magne Skulstad 2, Vivi Schlünssen 13, Cecilie Svanes 2,14,✉,#, John W Holloway 1,15,#
PMCID: PMC12102279  PMID: 40410506

Abstract

Boys’ pubertal overweight associates with future offspring’s asthma and low lung function. To identify how paternal overweight is associated with offspring’s DNA methylation (DNAm), we conducted an epigenome-wide association study of father’s body silhouette (FBS) at three timepoints (age 8, voice break and 30) and change in FBS between these times, with offspring DNAm, in the RHINESSA cohort (N = 339). We identified 2005 differentially methylated cytosine-phosphate-guanine (dmCpG) sites (FDR < 0.05), including dmCpGs associated with offspring asthma (119), lung function (178) and BMI (291). Voice break FBS associated with dmCpGs in loci including KCNJ10, FERMT1, NCK2 and WWP1. Change in FBS across sexual maturation associated with DNAm at loci including NOP10, TRRAP, EFHD1, MRPL17 and NORD59A;ATP5B and showed strong correlation in reduced gene expression in loci NAP1L5, ATP5B, ZNF695, ZNF600, VTRNA2-1, SOAT2 and AGPAT2. We identified 24 imprinted genes including: VTRNA2-1, BLCAP, WT1, NAP1L5 and PTPRN2. Identified pathways relate to lipid and glucose metabolism and adipogenesis. Father’s overweight at puberty and during reproductive maturation was strongly associated with offspring DNA, suggesting a key role for epigenetic mechanisms in intergenerational transfer from father to offspring in humans. The results support an important vulnerability window in male puberty for future offspring health.

graphic file with name 42003_2025_8121_Figa_HTML.jpg

Subject terms: Risk factors, Asthma, Predictive markers, Germline development, DNA methylation


Epigenome association study of father’s preconception body silhouette with offspring DNA methylation, providing mechanistic insight into how epigenetic inheritance impacts offspring obesity, asthma and lung function.

Introduction

Even though the impact of obesity on the individual is clearly understood, the prevalence of childhood obesity is increasing across the globe1. Recent epidemiological studies from the RHINESSA/RHINE/ECRHS cohorts and the Tasmanian Longitudinal Health Study (TAHS) have shown that overweight in boys at the age of voice break (puberty) may impair not only their own health but also the health of their future offspring, in terms of higher asthma risk, lower attained stature and lower lung function24.

An exposure-sensitive period during prepubertal years has also been highlighted by observations from the Överkalix and ALSPAC cohorts, where excess food supply and smoking in mid-childhood have been linked to metabolic and cardiovascular health, and risk of obesity in subsequent generation(s)57. Father’s preconception of overweight/obesity has further been associated with offspring’s obesity and altered metabolism8. Supporting a role for intergenerational epigenetic mechanisms in humans, Kitaba et al.9 found that fathers’ exposure to tobacco smoke, particularly during adolescent years, was associated with altered epigenetic patterns in their future offspring.

Efforts in identifying molecular mechanisms underlying these findings have suggested a pre-conceptional influence on epigenetic (re)programming during germ cell development, where the sperm epigenome is increasingly plastic and responsive to environmental exposures which can possibly affect epigenetic states and give rise to pleiotropic effects in future offspring if transmitted to the next generation at fertilisation1014.

Male obesity has been found to alter spermatocyte DNA methylation patterns1517 and non-coding tRNA content18, as well as seminal plasma composition19,20. Offspring of obese fathers have also been shown to display altered DNA methylation levels at several regulatory regions of imprinted genes21,22. Given that fathers’ BMI around conception has also been demonstrated to be an independent determinant of offspring metabolic health18 as well as to be associated with offspring birthweight and epigenome-wide methylation patterns up to the age of 723, this clearly supports the hypothesis that BMI-related differences in sperm content can be transmitted to subsequent offspring and impact on their health and development.

The present study aimed to investigate whether fathers’ body silhouettes (FBS) (as a surrogate measure of body composition) and their trajectory across adolescence and young adulthood were associated with offspring DNA methylation patterns and whether identified signals were associated with offspring phenotypic outcomes in terms of asthma, lung function and BMI. We hypothesised that differential DNA methylation patterns in offspring might reflect the molecular mechanisms underlying the effects of fathers’ obesity in adolescence on offspring health observed in epidemiological studies24. We have previously shown that the use of self-reported figural body silhouettes provides a valid tool for assessing overweight and obesity retrospectively24. In a two-generation cohort, we sought to identify the DNA methylation pattern in whole blood of offspring (aged 7–51 years) associated with (1) the father’s preconception body silhouette at ages 8, voice break and 30 years and (2) trajectory of father’s body silhouette across adolescence and young adulthood measured as change from age 8 years to voice break and from voice break to age 30. As previous epidemiological studies have reported sex-specific health outcomes of paternal obesity on offspring2, we also wanted to explore whether patterns of association between fathers’ preconception body silhouette and offspring DNA methylation were different between sons and daughters.

Methods

Study design and data

Data and samples from offspring were available from the RHINESSA study (www.rhinessa.net)25. Parental data was retrieved from the population-based European Community Respiratory Health Survey (ECRHS, www.ecrhs.org) and/or the Respiratory Health in Northern Europe (RHINE, www.rhine.nu) studies. In this study, 339 offspring-father pairs with complete data on fathers’ body silhouettes and offspring DNA methylation were included (Table 1). These participants, all of the white European ethnicity, were from six study centres (Aarhus, Denmark; Albacete/Huelva, Spain; Bergen, Norway; Melbourne, Australia; Tartu, Estonia). Medical research committees in each study centre approved the study and all participants gave written consent Ethical permissions were obtained for each study wave from the local ethics committee in each of the participating centres. Details of the ethics committees and approval reference numbers for each study centre are listed on www.rhinessa.net. All ethical regulations relevant to human research participants were followed.

Table 1.

Characteristics of study participants according to father’s body silhouette (FBS) at age 8 years, at voice break and at age 30 years

A: FBS 8b B: FBS Voice breakb C: FBS 30c D: FBS Voice break by sexd E: Offspring aged
Variable Normal N = 304a Overweight N = 34a pb Normal N = 307a Overweight N = 32a pb Normal N = 288a Overweight N = 57a pb Normal Overweight pc Normal Overweight pd
Offspring sex 0.4 0.4 0.4 0.4 0.12
F 147 (48%) 19 (56%) 147 (48%) 18 (56%) 138 (48%) 31 (54%) 147 (89%) 18 (11%) 25 (21, 32) 27 (21, 35)
M 157 (52%) 15 (44%) 160 (52%) 14 (44%) 150 (52%) 26 (46%) 160 (92%) 14 (8.0%)
F: Social classe 0.2 0.9 0.039
High 78 (26%) 5 (15%) 78 (25%) 7 (22%) 65 (23%) 22 (39%)
Low 122 (40%) 13 (38%) 122 (40%) 14 (44%) 119 (41%) 19 (33%)
Unknown 104 (34%) 16 (47%) 107 (35%) 11 (34%) 104 (36%) 16 (28%)
G: Offspring ever had asthmae >0.9 H: Offspring BMIf <0.001 I: Offspring current body weightg J: Offspring lung functionh
Normal Overweight <0.001 FVC_pre* FEV1_pre* FEV1_FVC_pre* FVC_post* FEV1_post* FEV1_FVC_post*
F 33 (20%) 22.4 (20.6, 24.6) 317 (92%) 28 (8.1%) 3.87 (3.50, 4.24) 3.24 (2.97, 3.57) 0.85 (0.80, 0.88) 3.85 (3.54, 4.24) 3.34 (3.04, 3.68) 0.87 (0.84, 0.90)
M 34 (20%) 25.0 (23.1, 27.9) 234 (62%) 145 (38%) 5.43 (4.83, 5.97) 4.43 (4.01, 4.82) 0.82 (0.78, 0.85) 5.30 (4.88, 5.90) 4.59 (4.12, 4.93) 0.84 (0.82, 0.88)

FEV1_pre pre-bronchodilator forced expiratory volume in n 1 s, FVC_post forced vital capacity post-bronchodilator, FEV1_post FEV1 post-bronchodilator, FEV1_FVC_post FEV1/FVC post-bronchodilator.

*p < 0.001.

an (%).

bPearson’s Chi-squared test.

cn (%), Median (Q1, Q3).

dPearson’s Chi-squared test; Wilcoxon rank sum test.

eMedian (Q1, Q3), n (%).

fWilcoxon rank sum test, Pearson’s Chi-squared test.

gMedian (Q1, Q3).

hWilcoxon rank sum test.

Definitions of father’s body silhouettes (FBS) before conception

Father’s body silhouette (FBS) was measured using a validated figural drawing scale of 9 sex-specific body silhouettes at each time point (age 8, voice break, 30 years)24. The figural drawing scale has been validated against self-reported height and weight both for current and past body silhouettes24. We applied a cut-off of Fig. 5 or greater to classify fathers as being overweight. This is the same cut-off that a previous validation study identified as optimal for identifying overweight people (BMI, 25–30 kg/m2)26. Change in the father’s body silhouette between age 8 and voice break, and voice break and age 30 were defined according to changes in body silhouette figure between the two time points. Aspects of change were also investigated according to whether they switched from being normal weight to overweight or vice versa across adolescence and young adulthood, and according to how many body figures gain or loss in body silhouettes spanned (see Supplementary Fig. 1).

Fig. 5. Alluvial plot showing the top 5 lipid-related traits gene ontology terms and gene set for dmCpGs associated with father’s body silhouette between voice break and age 30 years (FBS-V30c).

Fig. 5

Additional GO terms and related genes are provided in Supplementary Data 6.

Offspring outcomes

Adult offspring ever having asthma was based on answers to the question: “Have you ever had asthma diagnosed by a doctor?”. Lung function (forced expiratory volume in 1 s (FEV1) and forced vital capacity (FVC)) was measured at RHINESSA clinical examinations as previously described27.

Methylation profiling and processing

DNA methylation in offspring was measured in DNA extracted from peripheral blood, using a simple salting out procedure28. Bisulfite-conversion was undertaken using EZ 96-DNA methylation kits (Zymo Research, Irvine, CA, USA) at the Oxford Genomics Centre (Oxford, UK), and methylation was assessed using Illumina Infinium MethylationEPIC Beadchip arrays (Illumina, Inc., CA, USA) with samples randomly distributed on microarrays to control against batch effects.

DNA methylation quality control and normalisation

Data analysis was undertaken using R v 4.129 and methylation quality was assessed using minfi30 and Mefil31. To remove technical variation detected by SVD plot32 using champ33, combat from SVA was applied for both batch and slide variables34. Cell-type proportions were estimated using Epigenetics Dissection of Intra-Sample Heterogeneity (EpiDISH)35. Normalisation was carried out using BMIQ36. Probes were excluded from analysis using the following criteria: Detection p above 0.01, probes with a beadcount <3 in at least 5% of samples (N = 1357), non-cg probes (N = 2762), SNPs as identified by Zhou37 (N = 93,900), aligned to multiple locations as identified in Nordlund38 (N = 15), probes on the X or Y chromosomes (16,089), and cross-reactive probes on the EPIC array (2382)39. A total of 730,820 probes were used for downstream analysis.

Statistical analysis

To identify differentially methylated Cytosine-phosphate-Guanine sites (dmCpGs), the associations of offspring DNA methylation beta-value as outcome (continuous outcomes) with paternal body silhouette phenotypes (normal or overweight/obese, independent variables) were assessed using robust linear regression models using limma40. Covariates included offspring age, sex, estimated blood cell proportions (B-cells, Natural Killer cells, CD4 T-cells, CD8 T-cells, Monocyte, Neutrophils) and socio-economic class. Eosinophils were not included due to a very low estimate and to avoid potential multicollinearity41. Grandparents’ education was used as a proxy for paternal social class in childhood, either as low or high. Social class was assigned as high for university-level education or if both grandparents attended high school, otherwise it was assigned as low. Multiple test correction was applied using the Benjamini and Hochberg method42 where a false discovery rate (FDR) corrected p ≤ 0.05 was considered as statistically significant in the covariates corrected regression model.

The FBS-CpG associations were assessed by running

  • EWAS analyses at each of the paternal age time points: Father body silhouette at age 8 (FBS-8), Father body silhouette at voice break (FBS-V), and at Father body silhouette at age 30 (FBS-30).

  • A sex-stratified EWAS analysis of Father body silhouette at voice break in females (FBS-Vf) and Father body silhouette at voice break in males (FBS-Vm) to investigate whether epigenetic signals differ between male and female offspring.

  • EWAS analyses of change in paternal body silhouettes based on both ordinal (Father body silhouette change between age 8 and voice break (FBS-V8c), Father body silhouette change between voice break and age 30 (FBS-V30c) and categorical scale measures of gradations in body size (Father body silhouette gain or reduced between age 8 and voice break (FBS-V8gr)/Father body silhouette gain or reduced between voice break and age 30 (FBS-V30gr) and Father body silhouette retain or swap between age 8 and voice break (FBS-V8rs)/ Father body silhouette retain or swap between voice break and 30 (FBS-V30rs) to explore effects of change in FBS across adolescence/young adulthood on the offspring epigenome.

  • An EWAS analysis to assess the comparability of offspring's current BMI and body silhouette.

Downstream enrichment analysis and biological interpretation of dmCpGs

Manhattan plots were generated using qqman43. Inflation from systematic biases (measured by genomic factor lambda λ) was adjusted using BACON44. Differentially methylated regions were detected using DMRcate45 and dmrff46. Transcription factor binding site prediction was performed using eFORGE TF47. Gene–disease phenotype associations were identified using Open Targets48. The EWAS atlas49 was used to assess dmCpGs for association with known biological traits and the effect of methylation on gene expression. For dmCpGs mapped to genes, gene function ontology (GO) terms were identified using String50 and enrichr51. KEGG pathways were generated and visualised using Cytoscape52,53. The methylGSA R-package which accounts for the representation of probes per gene on the EPIC array was used to test for GO enrichment54. DMR regional enrichment was carried out using the goregion function from missMethyl R package55. Genes overlapping with the GWAS catalogue gene set for obesity traits were identified using FUMAGWAS GENEN2FUNC56 and dbGAP51. Lookup for the association of SNPs with methylation to identify methylation quantitative trait loci (mQTL) was carried out using the goDMC57 and the MeQTL EPIC58 databases. The look-up for overlap of known human imprinting genes used reference imprinting genes from https://geneimprint.com/site/genes-by-species.Homo+sapiens. Over-representation of metastable epialleles was compared with CpGs identified by Silver et al.59.

Association dmCpGs and offspring outcomes

The identified dmCpG sites across all EWAS were analysed for association with offspring clinical phenotypic traits, specifically BMI, asthma and lung function (FEV1 and FVC) using linear and logistic regression.

Reporting summary

Further information on research design is available in the Nature Portfolio Reporting Summary linked to this article.

Results

The present analyses included 339 RHINESSA offspring (Table 1), 174 males and 165 females, aged 7–50 years. Of these, 307 had a father who reported a normal body silhouette at voice break, and 32 of the offspring had a father who reported an overweight/obese body silhouette at voice break. There was a significant sex difference between males and females for BMI and lung function. Offspring current BMI, sex and age distribution by study centre are shown in Table 2.

Table 2.

Characteristics of offspring for Current BMI, age and sex by study centre (N = 724)

Offspring Albacete N = 48a Arhus N = 48a Bergen N = 395a Huelva N = 29a Melbourne N = 75a Tartu N = 129a p-valueb
Sex 0.7
Female 21 (44%) 26 (54%) 180 (46%) 15 (52%) 40 (53%) 63 (49%)
Male 27 (56%) 22 (46%) 215 (54%) 14 (48%) 35 (47%) 66 (51%)
Age 30 (25, 38) 29 (23, 37) 27 (23, 33) 36 (25, 39) 31 (25, 37) 30 (25, 34) <0.001
BMI 23.1 (21.0, 26.0) 22.9 (21.3, 25.8) 23.9 (21.9, 26.6) 23.1 (20.4, 27.5) 23.4 (21.1, 25.8) 23.8 (21.2, 27.0) 0.3

an (%); Median (Q1, Q3).

bPearson’s Chi-squared test; Kruskal–Wallis rank sum test.

Associations of father body silhouette (FBS) with offspring DNA methylation

The summary statistics and sample size for each EWAS are shown in Table 3. Of the three specific preconception age windows, father’s body silhouette at voice break was associated with a larger number of differentially methylated CpGs in offspring compared to father’s body silhouette in childhood or young adulthood. Sex-specific EWAS analyses identified more dmCpGs related to FBS at voice break in female than in male offspring. In particular, EWAS exploring changes in father’s body silhouette figures across adolescence, i.e. from a normal weight to an overweight body silhouette status or vice versa, were associated with altered DNA methylation patterns in the offspring. In total, we identified 2005 (1962 unique) dmCpGs that showed association with father’s normal versus overweight/obese body silhouette before conception (FDR corrected p ≤ 0.05). Key results for each EWAS are described below; the full list of dmCpGs and adjusted p for each EWAS are provided in Supplementary Data 1A.

Table 3.

Summary statistics of dmCpGs for all EWAS at FDR ≤ 0.05

EWAS on father body silhouette Label N dmCpGs Inflation N persons
Father body silhouette at age 8 years FBS-8 12 1.2 338
Father body silhouette at voice break FBS-V 41 1.3 339
Father body silhouette at voice break in male FBS-Vm 32 1.291 174
Father body silhouette at voice break in female FBS-Vf 370 1.4 165
Father body silhouette at age 30 years FBS-30 3 1.12 345
Father body silhouette change between voice break and age 8 FBS-V8c 277 1.17 334
Father body silhouette gain or reduced between voice break and age 30 FBS-V8gr 222 1.2 334
Father body silhouette retain or swap between age 8 and voice break FBS-V8rs 176 1.05 334
Father body silhouette change between voice break and age 30 FBS-V30c 791 1.176 334
Father body silhouette gain or reduced between age 8 and voice break FBS-V30gr 69 1.4 334
Father body silhouette retain or swap between voice break and age 30 FBS-V30rs 12 1.003 334
Total 2005

dmCpG differentially methylated cytosine-phosphate-guanine site, FDR < 0.05.

Father’s body silhouette at age 8 (FBS-8): We identified 12 dmCpG sites associated with father’s normal versus overweight/obese body silhouette at age 8 (FDR < 0.05). These sites were mapped to 7 protein-coding genes and 6 intergenic regions. Eight were hypermethylated and 4 hypomethylated with association coefficients ranging from −0.04 to 0.03. Eight of the sites were in the open sea (Supplementary Data 1A: FBS-8).

Father’s body silhouette at voice break (FBS-V, FBS-Vm, FBS-Vf): We identified 41 dmCpG sites associated with the father’s body silhouette at voice break; these mapped to 32 genes (29 protein-coding and 3 noncoding RNA) and 11 intergenic regions as shown in Table 4. The majority of dmCpGs were hypermethylated in the overweight group (71%) and, relative to CpG Islands, 66% were located in the open sea. For several of the top 10 dmCpGs (FDR corrected p ≤ 0.05), the methylation distribution showed a pattern of increasing or decreasing methylation according to increasing overweight in the father (increasing levels of FBS) (Fig. 1).

Table 4.

dmCpGs for father’s body silhouette at voice break (FBS-V) FDR ≤ 0.05

Name Effect size Average methylation Island Gene name Adj. p-value
cg20975419 0.09 0.18 OpenSea 0.000
cg11789449 0.20 0.60 OpenSea KCNJ10 0.000
cg06444433 −0.05 0.84 OpenSea FERMT1 0.015
cg23653826 0.03 0.93 OpenSea NCK2 0.016
cg09655253 −0.09 0.82 OpenSea MOCS1 0.016
cg12026976 −0.01 0.92 OpenSea 0.016
cg12587260 0.02 0.15 N_Shore PAPLN 0.016
cg19430728 0.01 0.94 OpenSea CD44 0.020
cg02157155 0.09 0.54 OpenSea EFCAB9 0.022
cg05144772 −0.02 0.90 OpenSea LOC389602 0.024
cg25020933 0.02 0.08 OpenSea B4GALNT4 0.024
cg21812470 0.06 0.20 N_Shelf CNGA1 0.026
cg25380281 0.03 0.91 OpenSea WWP1 0.026
cg15549838 0.01 0.90 OpenSea FNDC7 0.026
cg22341132 0.04 0.44 Ope nSea 0.026
cg13698153 0.02 0.12 Island VSIG10 0.031
cg05839509 −0.01 0.93 OpenSea KIAA0040 0.033
cg13242924 0.01 0.10 OpenSea 0.035
cg04165857 0.01 0.93 OpenSea 0.035
cg10053674 −0.02 0.89 OpenSea LOC102723544/SLC6A13 0.035
cg11737070 −0.03 0.79 OpenSea TBC1D4 0.035
cg18092219 0.01 0.10 OpenSea 0.035
cg11278727 0.02 0.07 OpenSea 0.036
cg05509659 0.00 0.02 Island ROBO3 0.037
cg06520845 −0.04 0.78 OpenSea EGFR-AS1 EGFR 0.040
cg05357152 0.03 0.23 Island GATA5 0.040
cg02212575 −0.01 0.92 S_Shore 0.044
cg04544017 0.02 0.86 N_Shelf SPATA2L 0.047
cg25361524 −0.02 0.83 OpenSea 0.047
cg24052851 0.00 0.98 Island 0.054
cg16612995 −0.01 0.94 Island ADAMTSL4 0.055
cg24420089 0.02 0.37 N_Shore PTDSS2 0.055
cg11949388 0.03 0.86 OpenSea FOXO3 0.055
cg27113059 0.00 0.02 Island LIPG 0.056
cg18109649 0.05 0.80 OpenSea MRPS28 0.056
cg15673187 0.01 0.04 Island DST 0.056
cg15009114 −0.01 0.94 N_Shore RASL11A 0.056
cg24906129 0.02 0.91 OpenSea 0.056
cg07405570 0.02 0.92 OpenSea LPCAT3 0.056
cg12110395 0.01 0.98 S_Shore ZNF570 0.056
cg11720773 0.04 0.24 OpenSea SYT1 0.056

Fig. 1. Box plots showing the distribution of methylation levels (beta-values) of top 10 dmCpGs (FDR p-value) for FBS at voice break.

Fig. 1

The distribution of each dmCpG for the father’s “normal” vs. “overweight” body silhouette, and across the father’s body silhouette numbers 1–7 is shown. The p-value comparing normal vs. overweight is shown above the box plot for each dmCpG.

In male offspring, we identified 32 dmCpGs mapped to 26 genes and 6 intergenic regions. 16 were located in open sea (see Supplementary Data 1A: FBS-Vm). In female offspring, we identified 370 dmCpG sites. Of these, 248 dmCpGs were mapped to coding genes and 122 to intergenic regions (Supplementary Data 1A: FBS-Vf).

We identified 6 genomic regions that were differentially methylated in offspring whose fathers had a normal versus overweight/obese FBS status at voice break. Using DMRcate differentially methylated regional enrichment analysis at voice break identified a DMR in SLC44A4 (8 sites) which was enriched for choline metabolism in cancer, glycolysis/gluconeogenesis, citrate cycle (TCA cycle) and fatty acid metabolism (see Supplementary Data 2: DMR:FBS-V DMRcate). In the analysis stratified by offspring sex, we detected 1 DMR in males and 15 DMRs in females. The female DMR sites included ADAMTS16, RNASE1 and AGAP2 at dmr.p.adjust < 0.05 (Supplementary Data 2).

To identify any overlap between the methylation of dmCpGs and known transcription factor (TF) binding sites, we interrogated eForgeTF by selecting CD34_T0 cell line, at https://eforge-tf.altiusinstitute.org (accessed on 23 August 2023). In the FBS analyses at voice break, we found 17 dmCpGs overlapping with 28 TF binding sites (q-value < 0.05). The methylation of cg25020933 (B4GALNT4) overlapped with 6 TFs, cg24420089 (PTDSS2) overlapped with 4 TFs and both cg05509659 (ROBO3) and cg09655253 (MOCS1) overlapped with 2 TFs (Supplementary Data 3). In sex-stratified analyses of FBS at voice break, we found overlapping with 30 TFs (q-value < 0.05) in male offspring and with 6 TFs (q-value < 0.05) in females (Supplementary Data 3).

In dmCpGs associated with FBS at voice break, sites located to ZNF570, ZNF569, B4GALNT4, LIPG, PTDSS2 and GATA5 showed correlation with gene expression (Supplementary Data 4: FBS-V). In the female-stratified analyses of FBS at voice break (n = 123), several of the identified dmCpGs showed correlation with gene expression and mapped to genes including MAGI2, GALNT9, SLCO3A1, OR2I1P, GPR153, TBC1D2B, TNFRSF1, NCAM1 and JAG1 (Supplementary Data 4: FBS-Vf).

The summary of the number of dmCpGs that showed correlation with gene expression and full details for both promoter and gene-body associated dmCpGs across the six tissues are provided in Supplementary Data 4.

Pathway analysis: Enrichr was used to assess the enrichment of curated signalling pathways among identified dmCpGs. For FBS at voice break-related genes, 31 GO terms (p < 0.05) were identified including gastrin signalling pathway (FOXO3; EGFR; CD44), molybdenum cofactor (Moco) biosynthesis (MOCS1), insulin-signalling in adipocytes (TBC1D4), NAD metabolism, sirtuns and aging (FOXO3), metabolic pathway of LDL, HDL and TG (LIPG), glycerolipids and glycerophospholipids (PTDSS2) and ferroptosis (LPCAT3). Using methylRR package GSEA we identified GO terms including cellular response to lipopolysaccharide (GO:0071222) and with AGSEA-Promotor1, cellular lipid catabolic process (GO:0044242) (Supplementary Data 5).

Father’s body silhouette at age 30 years (FBS-30): We identified three dmCpGs mapping to three genes, QSOX2, DLGAP2 and PCDHG (Supplementary Data 1A: FBS-30).

Change in father’s body silhouette between age 8 and voice break: A total of 277 dmCpGs showed association with change in FBS between age 8 and voice break (λ = 1.17) (Supplementary Data 1: FBS-V8c). The top hit, cg20668887, mapped to NOP10 (FDR = 1.68e−91); the association coefficients ranged from −0.34 to 0.42 (Figs. 23). Some genomic loci had multiple dmCpGs such as NFYA;LOC22144, PTPRN2 and NAP1L5;HERC3 with the majority (81%) of dmCpGs showing hypermethylation. In the EWAS model using categorical cut-offs of FBS change, we identified 222 dmCpGs at FDR ≤ 0.05 associated with father´s gain or loss in FBS (Supplementary Data 1: FBS-V8gr). We detected 176 dmCpGs (λ = 1.05) associated with father´s switch in normal or overweight/obese FBS status between age 8 and voice break. The top hit, cg10157663, mapped to CCDC178 (FDR = 3.91e−21) with 78% of dmCpGs showing hypermethylation. Many loci were represented by more than 1 dmCpG including PTCH1 (10 sites), GABRG1 (7 sites), BLCAP (6 sites), SORCS1 and HIST1H2BE (3 sites) (Supplementary Data 1A: FBS-V8rs).

Fig. 2. Volcano plot for regression coefficients of dmCpGs associated with a change in father’s body silhouettes.

Fig. 2

A Between voice break and age 8 years (FBS-V8c) and B between voice break and age 30 years (FBS-V30c). The X-axis shows regression coefficients, and the Y-axis represents −log10 of the p-values. Positive coefficients show hypermethylated dmCpGs, while the magnitude shows the strength of the association.

Fig. 3. Manhattan plots showing the genome-wide distribution of dmCpGs associated with a change in father’s body silhouette.

Fig. 3

A Between age of 8 years and voice break (FBS-V8c) and B between voice break and age 30 years (FBS-V30c). The x-axis shows the position across autosomal chromosomes. The y-axis represents −log10 of the p-value for each dmCpG (indicated by dots). The green dots show loci with more than 1 dmCpG at FDR-corrected p < 0.05. The top dmCpGs on each chromosome were annotated to the closest gene. For intergenic dmCpGs, CpG name was used.

Change in father’s body silhouette between voice break and age 30: We identified 791 dmCpGs associated with a change in FBS between voice break and age 30 years; 559 were mapped to coding genes (Supplementary Data 1: FBS-V30c). The coefficient of association ranged from −0.53 to 0.82, as shown by the volcano plot, where 68% showed hypermethylation (Fig. 2). The genome-wide distribution is shown in Fig. 3. The top hit, cg18950772, mapped to an intergenic region (FDR = 6.23e−261) followed by SLC25A10 (FDR = 3.21e−205). Some genes were represented by many dmCpGs including NUP210L and TRPM4, WT1, MIR886, NFYA;LOC221442, DIP2C and AGPAT2. In the EWAS models investigating categorial classifications of change in FBS from voice break to age 30, we identified 69 dmCpGs associated with the father´s gain or loss in body silhouette, and 12 dmCpGs related to father´s switch in normal or overweight FBS status between voice break and age 30 (Supplementary Data 1A: FBS-V30gr and FBS-V30rs).

The change in FBS DMR includes sites from NFYA, LARS2, NAP1L5 and CREBBP for FBS-V8c, BLCAP;NNAT, PTCH1 and GABRG1 for FBS-V8rs, PM20D, C22orf45, MBP, NUP210L, TRPM4, PACSIN1, ERICH1, MIR886, IQSEC3, BCL11B, AGPAT2 and NFYA for FBS-V30c, and NAPRT1, NUP210L and NAPRT for FBS-V30rs. Details of the DMRs are shown in Supplementary Data 2.

In DMR analysis with DMRCate at FBS-V8c, we identified 13 DMRs including gene loci FBXO47 (10 sites), CRISP2 (12), UPB1;ADORA2A-AS1 (14) which were enriched for many metabolic pathways (see Supplementary Data 2 DMR:FBS-V8c). For FBS-V30c, DMRcate analysis identified 81 DMRs including loci WT1 (5 sites), NNAT;BLCAB (37), AURKC (12) and NUP110L (10). They were enriched for glycerophospholipid metabolism, vasopressin-regulated water reabsorption, ubiquitin-mediated proteolysis, glycerolipid metabolism and phospholipase D signalling pathway (see Supplementary Data 2 DMR:FBS-V30C_DMRcate).

In the EWAS models investigating change in FBS across adolescence  from age 8 to voice break we found overlap with 29 TFs (q-value < 0.05) while gain/reduction in FBS from voice break to age 30 overlapped with 33 TFs (q-value < 0.05) including PPARG (cg22681255 and cg15965578) and HOXA3 (cg135272218). The list of TFs for each EWAS is provided in Supplementary Data 3.

For a change in FBS from voice break to age 30 (FBS-V30c), out of 791 dmCpGs, 329 showed significant correlation with gene expression (p < 0.05) including VTRNA2-1, TRPM4, GPRC5C, WDR97, ZNF695, NUP210L, GSE1, DIP2C, RP5-894D12.3, FAM26F, C5orf66, AC098614.2, SOCS1, SLC24A4, R3HDM4, ZNF600, RP11-715J22.6, BCL11B and MBP. Of these, 218 dmCpGs were associated with promoter regions (Supplementary Data 4: FBS-V30c). Among the identified dmCpGs in this EWAS analysis, we also observed a strong correlation with gene expression in ZNF695 (11 sites) and ZNF600 (7 sites). There were 6 sites each for CEP85, SOAT2 and CD52, and 5 sites for ZNF334, ZBTB16 and AGAPT2, as shown in Fig. 4.

Fig. 4. Correlation of methylation level with gene expression for dmCpGs located on gene promoters across 6 tissues, as associated with change in father’s body silhouettes.

Fig. 4

A Between voice break and age 30 years (FBS-V30c) (4 sites for VTRNA2-1 and AGPAT2 and 2 sites for IFLTD1, ZNF695, ZNF600 and ZBTB16) and B between voice break and age 8 years (FBS-V8c) (3 sites for NAP1L5 and ATP5B). The dot size corresponds to the level of correlation.

Several of the dmCpGs identified in relation to changes in FBS from age 8 to voice break (n = 94) showed correlation with gene expression and multiple sites mapped to genes including NAP1L5, PTPRN2, CACNA1B, AJAP1, CREBBP, ERGIC1, KCNB1, TBX5, CFLAR, AMH, FBXO47, DIRC3, DLL1, APIP, TIMD4, RP1-34B21.6 and GALNT14 (Supplementary Data 4: FBS-V8c). The effect of methylation located in a promoter region on gene expression across all tissue types for FBS-V8c is shown in Fig. 4. It shows a strong correlation for genes NAP1L5 (3 sites), ATF6B (3 sites), RXRG, CDH22 and LEPROT.

Enrichment for gene ontology terms: To characterise the biological function of the dmCpGs, each dmCpG was mapped to the nearest gene for each EWAS and detailed functional gene ontology terms for each gene for each EWAS were retrieved from String. The detailed gene descriptions, related GO terms and lipid-related traits are provided in Supplementary Data 6. The largest GO terms were for genes related to dmCpGs identified in the EWAS on change in FBS from voice break to age 30 (FBS-V30c) n = 106,616. In the look-up for ‘lipid’ related terms, we identified 66 GO terms including lipid metabolic process with 29 associated genes (ABO, ACSF3, ACSM6, AGPAT2, AGPAT5, AKR7A2, CDIPT, CHKA, COQ2, CPT1A, DHRS11, FABP5, GPIHBP1, HSD11B2, KIT, LMF1, LPCAT4, MBTPS1, NFE2L1, PIGT, PLD3, PRKAG2, SERPINA12, SOAT2, SOCS1, ST3GAL6, SYNJ1, TPRA1 and VAC14). The top associated GO terms and shared genes are shown in Fig. 5.

For dmCpG-related genes in the EWAS on change in FBS from age 8 to voice break (FBS-V8c) there were 97 ‘lipid’ related GO terms, where lipid or lipoprotein measurement was represented by 36 genes (AGPAT3, ATP8A2, BTNL2, CAMK2B, CAMTA1, CCDC60, CDA, CDC42BPA, CDH13, COL4A2, CYP2B6, DCLK2, DGCR8, DSCAM, FRMD4A, HERC3, LAMC2, LEPR, LRP1, MACROD2, MGMT, MORN1, NAV2, NCOR2, PAPPA2, PHYHIP, PKNOX2, PPP1R13B, PTPRN2, RBMS3, RPA2, SHROOM3, SLC39A11, SLC44A4, SPTBN2, TIMD4).

For genes linked to dmCpGs associated with a change in FBS from age 8 to voice break (FBS-V8c), the KEGG pathway enrichment included: O-glycan biosynthesis (GALNT14, XXYLT1), Hippo signalling regulation pathways (CDH13, EPHA2, FGFR2, PRKACA, PRKCH), Adipocytokine signalling pathway (ACSL1, LEPR, RXRG) and Cholesterol metabolism (LRP1, LRPAP1) (see Fig. 6 and Supplementary Data 7 : FBS-V8c).

Fig. 6. Network plot showing KEGG for dmCpGs.

Fig. 6

A Between voice break and age 8 years (FBS-V8c). B Between voice break and age 30 years (FBS-V30c). Detailed pathways are included in Supplementary Data 7.

For dmCpG-related genes associated with a change in FBS from voice break to age 30 (FBS-V30c), the KEGG pathway showed lipid metabolic associated signalling pathways including NRP1-triggered (AKT3, BCAR1, CDH5, CHD2, COL1A1, EGFR, MAP2K1, RAC1), HIF-1 (AKT3, EGFR, IGF1R, IL6R, MAP2K1, MKNK2, NOS2, NOS3, STAT3), Adipocytokine (AKT3, CAMKK2, CPT1A, PRKAG2, RXRG, STAT3), Leptin (MAP2K1, NOS3, RAC1, STAT3), Sterol regulatory element-binding proteins (SREBP) (MBTPS1, MED15, NFYA, PRKAG2), PPAR (CPT1A, FABP5, RXRG), Sphingolipid (ABCC1, AKT3, GNA12, GNAI1, MAP2K1, NOS3, PPP2R2D, PPP2R5B, RAC1) and GABAergic synapse (ADCY5, CACNA1C, GABBR2, GABRP, GNAI1, GNAO1, KCNJ6). KEGG pathway network and shared genes are shown in Fig. 6. All identified pathways are shown in Supplementary Data 7.

GO terms: To identify the overlap of dmCpG mapped gene lists with known obesity-related traits in the GWAS catalogue, we used FUMAGWAS (p < 0.05) and Enrichr (dbGAP). For FBS at voice break in the female strata, we identified 11 traits and 20 genes related to obesity. These include 2 traits, and 18 obesity-related genes identified in the EWAS on change in FBS from age 8 to voice break (FBS-V8c) and 31 traits in the EWAS of change in FBS from voice break to age 30 (see Supplementary Data 8: FBS-V30c). The latter included waist circumference adjusted for body mass index (n = 36), adult body size (n = 28), apolipoprotein A1 levels (n = 23), waist-to-hip ratio adjusted for BMI (n = 34), offspring birth weight (n = 11), type-2 diabetes (n = 25), appendicular lean mass (n = 26) and hip circumference adjusted for BMI (n = 31). The top look-up from dbGAP for the EWAS model of difference in FBS from age 8 to voice break (FBS-V8c) was cholesterol and HDL (RNF157, CHCHD3, BORA, PTPRN2, ERBB4, DEFA3, ROR1, NPHP4, TACC2, CDC42BPA, RXRG, RBMS3). For the EWAS on change in FBS from voice break to age 30 (FBS-V30c) the top look-up was BMI (ATL2, XYLT1, AGAP3, ITGAE, LDLRAD4, ASB18, GNAI1, GPR176, ADAMTS2, IMMP2L, SCML4, MTHFD1L, SCFD2, HAS3, OPCML, NTNG1, KDM4C, SFMBT1, COG5, LMNTD1, PCGF3, KCNIP4, RASSF8, WNT9A, MRPL52, NCAPD3, WDFY4, STAM2, NFE2L1). The list of GWAS catalogue enriched obesity-related traits is provided in Supplementary Data 8.

Biological interpretation of dmCpGs

EWAS atlas lookup: We interrogated the EWAS atlas (7/02/2025) for known trait associations and identified 835 dmCpGs showing 2005 known associations for 217 traits (Supplementary Data 9). The top associated traits include smoking status (n = 193), aging (n = 188), Down’s syndrome (n = 123), ancestry (n = 89), asthma (n = 35) and obesity (n = 21).

Correlation between dmCpG methylation and gene expression: For each EWAS, using dmCpG as input, we assessed the known regulatory effect of methylation on gene expression across 6 tissue types (testis, stomach, colon, brain, liver and kidney) using the EWASAtlas (https://ngdc.cncb.ac.cn/ewas/atlas, accessed 7/02/2025). Out of 2005 dmCpG sites, 776 (38%) showed significant correlation with gene expression and we identified 2627 associations between dmCpGs and gene pairs (see Supplementary Data 4 for each EWAS).

The detailed results are provided for overlap with known transcription factor (TF) binding sites, association with other traits (Supplementary results and Supplementary Data 5), KEGG signalling pathway (Fig. 6 and Supplementary Data 7) and obesity-related traits in the GWAS catalogue (Supplementary results and Supplementary Data 8).

Lookup for the effect of genetics on methylation (meQTL): To identify methylation as a consequence of known genetic variants, we searched for dmCpGs using goDMC (designed for 450K n = 420,509 CpGs) and the mQTL EPIC database (n = 724,499 dmCpGs). We found 698 (35%) and 1209 (60%) dmCpGs that showed meQTL association in the goDMC and meQTL epic database (p < 0.05) respectively. Full details of the dmCpGs and associated SNPs are provided in Supplementary Data 10A and B.

Look-up for imprinting and metastable epialleles: To investigate whether the methylation sites associated with father’s preconception body silhouettes were related to imprinted genes, we used the Geneimprint database at geneimprint.com (accessed on 17 January 2025). We identified 47 dmCpGs mapped to 24 genes known to be imprinting genes. Some of these genes were represented by more than one dmCpG, including 6 dmCpGs for BLCAP and WT1, 4 for VTRNA2-1 (MIR886), 3 for NAP1L5 and PTPRN2 and 2 each for MAGI2, MEG3, HOXA2, RASGRF1, B4GALNT4 and ZFAT. Of these genes, 13, 10 and 1 were known to show maternal, paternal and isoform-dependent allelic expression, respectively (Supplementary Data 11).

In the comparison of our unique dmCpGs (n = 1962) with known human metastable epialleles (n = 2408)59 we identified an overlap of 37 dmCpGs (Supplementary Data 10: metastable epialleles). In the EWAS Atlas look-up, these dmCpGs showed known associations with paternal uniparental disomy (n = 10), aging (n = 8) and Down’s syndrome (n = 7) (Supplementary Data 11: EWASAtlas_metastableEpialelles).

Six dmCpGs were identified by both methods, 2 from NAPL1L5 and 4 from VTRNA2-1; both genes are paternal imprinting genes and metastable alleles (Supplementary Data 10: metastable imprint). In the EWASAtlas NAPL1L5 (cg01026744) was linked with paternal uniparental disomy. For VTRNA2-1, all four dmCpGs (cg00124993, cg06536614, cg25340688 and cg26896946) were linked with Down’s syndrome, Parkinson’s disease and breast cancer, while three were linked with gestational diabetes, Clopidogrel resistance, and glycaemic response to glucagon-like peptide-1 analogue therapy in type 2 diabetes mellitus (Supplementary Data 11: ImprintedGene_EWAStraits).

Association of identified dmCpGs with offspring health outcomes: Asthma, lung function and BMI

Asthma: In total, 119 dmCpGs associated with father´s body silhouette across adolescence showed an association with offspring asthma (p < 0.05) (Supplementary Data 12A). The paternal phenotypes showing the largest number of dmCpGs associated with offspring asthma were in the EWAS analysis of change in FBS from voice break to age 30 (FBS-V30c) n = 52 (2 sites from BCL11B). Other associations with offspring asthma included dmCpGs associated with FBS at voice break in female offspring (FBS-Vf) n = 19 and dmCpGs associated with change in FBS from age 8 to voice break (FBS-V8c) n = 14 as well as dmCpGs related to switch in normal or overweight FBS status from age 8 to voice break (FBS-V8rs) (2 sites from BLCAP and HIST1H2BE).

Lung function: Analysis of the 2005 dmCpGs in offspring associated with father’s body silhouette phenotypes showed that methylation at 982 dmCpGs were associated with different measures of lung function in offspring including pre-bronchodilator FEV1 (n = 151), FVC (n = 145), and FEV1/FVC (n = 178), FVC post (n = 73), FEV1 post (n = 154) and FEV1/FVC post (n = 281). Again, the FBS phenotype dmCpGs showing the greatest number of associations with offspring lung function were with measures of FBS change across adolescence (Supplementary Data 12B).

BMI: Across all the 2005 dmCpGs identified for all FBS phenotypes, 291 dmCpGs showed an association with offspring BMI (p < 0.05). The strongest associations with offspring BMI were seen for dmCpGs associated with a change in FBS between voice break and age 30 (FBS-V30c) where 110 dmCpGs showed nominal association including DIP2C (3 sites), NFYA;LOC221442 (2 sites) and IQSEC3 (2 sites). For dmCpGs associated with a change in FBS from age 8 to voice break (FBS-V8c), there were n = 82 dmCpGs including DIP2C (3 sites), NFYA;LOC221442 (2 sites) and IQSEC3 (2 sites) that were nominally associated with offspring BMI. In female offspring dmCpGs associated with FBS at voice break (FBS-Vf), there were n = 53/370 (2 sites from FERMT10) that were associated with offspring BMI.

When categorising offspring BMI (Normal, Overweight and Obese) 225/2005 dmCpGs showed a nominal association. Detailed information is provided in Supplementary Data 12C.

Offspring current BMI and current body silhouette

To establish whether offspring DNA methylation associations with fathers’ body silhouette phenotypes were confounded by shared environmental exposures, we conducted EWAS of offspring's current BMI and current offspring body silhouette to compare shared dmCpGs between paternal and child exposures. We found 182 dmCpGs associated with offspring BMI and 1 dmCpG associated with offspring body silhouette at FDR corrected p < 0.05. The top hit for both was cg20217160 which maps to LACTB. When we compared the offspring’s top dmCpGs related to current BMI with the top 100 dmCpGs associated with the current offspring's body silhouette, 31 dmCpGs were shared (Supplementary Fig. 2 and Supplementary Data 13). We further compared the regression coefficient between current body silhouette and current BMI using all dmCpGs at nominal p < 0.05 (n = 24217 dmCpGs). The two regression coefficients showed a correlation r = 0.97 with p < 2.2e−16 (Supplementary Data 13 and Supplementary Fig. 2), further confirming the validity of the body silhouette approach to measuring offspring adiposity. However, of the 182 dmCpGs identified in the EWAS of offspring BMI only cg19640090 (ETV6) of 1962 unique dmCpGs were also associated with FBS phenotypes, suggesting the relationship between offspring DNA methylation and FBS is not significantly confounded by shared familial environment leading to high BMI in both generations.

We conducted an overlap look-up for our 1261 genes (mapped from our 1962 dmCpGs) with obesity-related known genes in open targets (EFO_001073) (n = 5884) and DiseGeNET (N = 2820) (accessed on 16/07/2022). We identified overlap with 450 genes in either of the two databases while 171 genes were reported by both databases. Our 811 genes (64.3% of 1261) are new reports (see Supplementary Fig. 3 and Supplementary Data 14).

Discussion

In this study of DNA methylation in humans as related to father’s overweight body silhouettes during childhood, voice break, and young adulthood, we identified >2000 differentially methylated CpG sites and many differentially methylated regions that were associated with father’s preconception overweight body silhouettes. In particular, changes in the father’s body silhouette status across voice break, from normal weight to an overweight FBS or vice versa, were associated with altered DNA methylation patterns in the offspring. This adds to the emerging understanding of the pubertal transition phase to be a period of increased vulnerability for lifestyle influences to drive epigenetic modifications and impact early development and phenotypic outcomes in offspring. This also lends support to our previous study of epigenetic effects in offspring of father’s smoking which was by far most pronounced if the father started smoking before age 15 years9 and by epidemiological studies identifying male prepuberty as a critical exposure window for phenotypic outcomes in offspring27,9.

DmCpGs and DMRs were identified in genes related to insulin-regulation, glucose metabolism, obesity traits, adipogenesis, fat-metabolism, diabetes, asthma, lung function, telomere maintenance, body form and aging. In agreement with this, a number of identified dmCpGs and DMRs associated with the father’s adolescence overweight were also associated with phenotypic outcomes in the offspring such as asthma, lung function and BMI. Our findings thus suggest that epigenetic mechanisms may be important to explaining the associations of father’s overweight around voice break with offspring asthma3,4 height2 and lung function2 and, in general, in the transfer of paternal exposure effects to phenotypic changes in offspring.

We have previously established that overweight boys at the age of voice break may impair not only their own health but also the health of their future offspring24. Supporting a role for DNA methylation driving these phenotype associations, methylation at CpGs in offspring associated with paternal preconception body silhouettes was also associated with offspring BMI, asthma, and lung function. Although, this could potentially reflect a confounding effect of shared familial environment or shared genetic susceptibility between fathers and offspring for these health outcomes, only one site (cg19640090 in ETV6) of the 182 dmCpGs identified in the EWAS of offspring BMI was also among the 1962 unique dmCpGs associated with Father’s body silhouette phenotypes. This suggests that the relationship between offspring DNA methylation and the father’s preconception of overweight body silhouette is not significantly confounded by shared familial environment leading to high BMI in both generations. Similarly, evidence from animal models8,60 where these confounding factors can be controlled for, suggests a pre-conceptional influence on epigenetic (re)programming events during gametogenesis may, at least in part, be a biological mechanism underpinning these associations.

Examining the dmCpGs in offspring associated with paternal preconception body silhouettes may provide biological insight into the mechanisms linking paternal obesity across puberty with offspring phenotype.

The dmCpGs associated with father’s body silhouette at age 8 included cg01945624 located to SH3TC1 (a gene known to be associated with low lipoprotein), cg10366797 in HOXC4 (associated with waist-to-hip ratio adjusted for body mass index61 and body shape index62) and cg08975641 mapped to TH2LCRR (a gene known to give higher susceptibility to asthma and allergic disease by impact on Th2 cell activity63). Genomic loci in SH3TC115 and HOXC417 have previously been shown to be differentially methylated in the sperm of obese compared to normal-weight men. Although the sites identified in our study seem to be novel, we suggest this adds support to the conclusion that the methylation signals detected in our study are indeed related to overweight.

The top hit for the father’s body silhouette at voice break was cg11789449 in KCNJ10, which was hypermethylated in offspring whose fathers had an overweight FBS status at voice break. KCNJ10 is important in the permeability of pancreatic beta cells which release insulin, and is linked with diabetes64. Several other dmCpGs associated with father’s FBS status at voice break were also linked to genes with roles in adipogenesis: LIPG locus (cg27113059) is known to be associated with lipid traits65, HDL cholesterol level66, cardiovascular risk67 and reduced visceral adiposity68; LPCAT3 (cg07405570) is known to regulate triglyceride secretion69; PTDSS2 (cg24420089) is known to be correlated with fat mass and BMI70; TBC1D4 (cg11737070) regulates insulin-stimulated glucose uptake71 and is associated with severe obesity, insulin resistance72 and type 2 diabetes73,74; NCK2 (cg23653826) is involved in regulating adipogenesis75,76 and DNA methylation sites located within NCK2 have also been demonstrated to be associated with BMI and body composition in children77; FOXO3 (cg11949388) regulates lipid accumulation and adipocyte inflammation in adipocytes78. We also observed that the methylation pattern of cg25380281 in the WWP1 gene showed an increasing trend across body silhouette scales 1–5 (Fig. 1). WWP1 regulates adipogenesis and metabolism79, enhances glucose metabolism80 and protects against oxidative stress80. Interestingly, as for father’s body silhouette at age 8, we also observed that several of the annotated genes related to father’s body silhouette status at voice break, have previously been reported to be differentially methylated in mature spermatozoa of overweight men, such as those found by Donkin and colleagues15 (LIPG, PTDSS2, NCK2, FERMT, MOCS1, CD44, FNDC7, SLCGA13, ROBO3, GATA5, SPATA2L, B4GALNT4) and in a recent study by Keyhan et al.17 (B4GALNT4, CNGA1, RASL11A, NCK2, KIAA0040). Our results add support to the existing literature and to the growing evidence that BMI-related alterations in sperm DNA methylation indeed can be transmitted to the offspring11,16.

Many observational studies have shown sex-specific differences in disease risk and measurement of obesity traits and BMI81. From our sex-stratified EWAS analysis of the father’s body silhouette at voice break, we identified different CpG sites for male and female offspring without any overlap. In male offspring, associated dmCpGs were linked to known obesity-related genes including B4GALNT4 (cg25020933, cg15762098), ADCY9 (insulin secretion and thyroid hormone synthesis) (cg00610508), ADRB1 (cg13848598) and ATP10D (cg14009629). All these genes have previously been reported to harbour loci that are differentially methylated in obese compared to lean men15,17. In a recent study, the imprinted gene B4GALNT4 has also been associated with adiposity change, both during infancy and in childhood years82. In female offspring, out of 308 dmCpGs, 20 genes were known to be related to obesity traits in GWAS catalogues. FERMT1 (cg06444433, cg16271200 and cg16421850) and ESRRG (cg03224209) have putative roles in glucose and fat metabolism. These findings may suggest different underlying molecular mechanisms that can explain the differences between male and female obesity. The DMR analysis in female offspring showed striking signals in obesity-related genes including RNASE1 (which regulates feeding habit83 and childhood obesity84), CPT1A (known to be involved in fatty acid oxidation)85, SSTR1 (growth hormone synthesis)86, DGKZ (lipid metabolism)87 HIF3A (childhood obesity)88 and ADAMTS16 (related to anorexia nervosa)89. Aside from, SSTR1, also in the female strata, dmCpG annotated genes have been found in sperm samples of overweight men15. HIF3A has also been shown to be correlated with paternal BMI and DNA methylation levels in offspring cord blood, although in opposite directions in male and female offspring16.

Even more pronounced signals of association were identified in the EWAS models of change in father’s body silhouette status across sexual maturation. Among dmCpGs associated with a change in FBS between age 8 and voice break (FBS-V8c), 3 sites (cg25607226, cg25890575, cg22991232) mapped to PTPRN2 which is known to be required for normal accumulation of secretory vesicles in the hippocampus, pituitary, and pancreatic islets. It also plays a role in insulin secretion in response to glucose stimuli and is known to be associated with both childhood9093 and adult obesity15,17. Four sites were located in NFYA (cg09580153, cg04346459, cg02167203, cg06671660) which is related to pre-adipocyte maintenance and/or commitment to adipogenesis94, energy metabolism, lipid metabolism95,fatty acid synthesis95 and leptin gene expression96. The top hit, cg20668887 was located in NOP10 which is involved in ribosomal biosynthesis and telomere maintenance; shorter telomere length is known to be associated with childhood obesity97, age-related disease98100 and BMI101.

In the EWAS model investigating the switch in normal or overweight FBS status between age 8 and voice break (FBS-V8rs), we identified PTCH1 which is known to have a role in pulmonary function102, adult body height103 and regulating obesity104. We also detected 7 dmCpGs located to GABRG1, which is a gene known to be linked with childhood obesity, coronary artery disease105 and adipogenesis106. We have reported many dmCpGs that were associated with changes in the father’s body silhouette status from voice break to age 30 (FBS-V30c) including cg10460003 in SLC25A10, which has a role in fatty acid synthesis107 and adipocytes insulin sensitivity108. Donkin and colleagues also found that sperm DNA methylation patterns in these genes were significantly different in obese men15.

Six dmCpGs were located in NUP210L, which has been shown to be related to asthma and risk of obesity109 and diabetes110. We also identified 6 dmCpGs in TRPM4 which has a role in vascular formation111 and is a known target for diabetes112. Four sites mapped to AGPAT2 (cg15720535, cg13422804, cg02703247, cg03571320) which is linked with lipodystrophy113,114 and has a role in the synthesis of triglycerides and phospholipids115,116. The methylation pattern in the dmCpGs sites (cg04292615 and cg16848843) in ZNF600, a gene associated with phospholipid level117,118 is shown in Fig. 4. Some of the dmCpGs associated with father’s switch in normal or overweight FBS status between voice break and age 30 (FBS-V30rs) were also linked to genes known to be associated with regulating adipogenesis (SERPINA12 (cg01207931), ZNF423 (cg25096280) and ADAD2 (cg00225858)). Genomic loci in ZNF423 have also been shown to be associated with BMI and altered sperm DNA methylation in obese men15,17.

The DMR genes related to change in FBS are relevant to obesity. PM20D1 is an enzyme that regulates N-fatty acyl amino acid (NAAs) synthase/hydrolase by regulating the whole body's energy expenditure119,120. It converts free fatty acids and free amino acids into NAA. It is a potential anti-obesity target121 and is associated with cardiovascular risk119. NNAT regulates metabolic status122 and has been shown to be differentially methylated in sperm of obese men15,17 and to be correlated with paternal BMI in offspring cord blood16.

We found that many of our identified dmCpGs were associated with promoter regions that can influence gene expression, especially in primary organs associated with lipid processing such as the liver. Some of the genes that showed a strong correlation to a reduction in gene expression regulate metabolism in obesity122, growth restriction and obesity123, food intake124 and development of childhood obesity. These include the imprinted genes VTRNA2-1 (associated with childhood obesity)125 NAP1L5 (related to body fat percentage), and NNAT (linked to hyperglycaemia and obesity), and the non-imprinted genes LEPROT (fat mass), TIMD4 (total cholesterol measurement), ATF6B (phospholipid measurement), and SOAT2 (LDL, type 1 diabetes, cholesteryl ester measurement, familial lipoprotein lipase deficiency and chylomicron retention disease).

Dysregulation of imprinted genes has been associated with obesity126, which may also explain why several of the imprinted genes we have identified as related to father’s preconception body silhouettes have previously been shown to be differentially-methylated in sperm of overweight/obese compared to normal-weight men. These include NNAT, RASGRF1, PTPRN2, CDH13, ZFAT, PAOX, CDA, NTM, CHST8, TACC2, DSCAML, C10orf91, JPH3, KCNQ1DN15,PTPRN2, NAP1L5, WT1, HOXA2, B4GALNT, CELF4, DLGAP217,15), MEG3 and NNAT16,21,127). Altered methylation levels in Altered methylation levels in NNAT have also been observed in the cord blood of offspring with obese fathers21, which further supports the idea that epigenetic alterations at imprinted genes in the gametes might be passed onto offspring.

Epigenetic dysregulation of the imprinting genes identified is not only related to obesity but also to diseases including paternal uniparental disomy (BLCAP (cg01466133, cg07156273, cg10981598, cg14469070, cg18433380, cg23605670), NAP1L5128 (cg11300971, cg01026744), MEG3 (cg08698721), NNAT (cg11174847) and Down’s syndrome (BLCAP (cg07156273, cg10981598, cg14469070), VTRNA2-1 (cg00124993, cg06536614, cg25340688 and cg26896946)) in the EWASAtlas (Supplementary Data 11).

Intriguingly, evidence suggests that VTRNA2-1 (paternally expressed) is a metastable epiallele, with stable methylation levels shown to be preserved across populations129. Thus, the mechanism by which DNA methylation variability in VTRNA2-1 is inherited essentially appears to be non-genetic130. In this study, 4 dmCpGs (cg26896946, cg25340688, cg00124993, cg06536614) mapped to the promotor region of VTRNA2-1 showed a correlation with reduced gene expression across five tissue types (Fig. 4A). All four have been linked with Down’s syndrome, Parkinson’s disease, breast cancer and pre- and post-lenalidomide treatment in patients with myelodysplastic syndrome with isolated deletion (5q). Furthermore, gestational diabetes, Clopidogrel resistance131, and glycaemic response to glucagon-like peptide-1 analogue therapy in type 2 diabetes mellitus are each associated with three of the four dmCpGs in VTRNA2-1132. It is also known to be associated with BMI and insulin125 and sensitivity to periconceptional environmental exposure133 and glucose metabolism134.

For NAP1L5 (also paternally expressed) 3 dmCpGs (cg19151808, cg01026744, cg11300971) showed correlation with reduction in gene expression. Of these, cg01026744 is a metastable allele linked with paternal uniparental disomy (known to be associated with early-onset of obesity).

Our gene set analysis identified key pathways known to contribute to obesity pathogenesis including: Adipocytokine, AKT, PPAR, Wint, adipogenesis and lipid metabolism.

Of the dmCpGs (genes) associated with offspring health outcomes, HIST1H2BE (2 sites) and TBC1D14 (2 sites) are known to be linked with asthma severity135, PHF19 is linked with childhood asthma and SERPINB9P1 is a known asthma drug target. NOX3 is also an asthma gene136 and SH3TC1 is linked with COPD137. Our top hits KCNJ10 and FERMT1 also showed association with asthma. In the female EWAS, genes well known to be linked with asthma include MUC1, RASGRF1 and IL9138. The lung function-associated loci include dmCpGs from PTCH1 (cg22073802 and cg16581009), MUC1, IL9 known to be related to pulmonary function102 and SERPINA12139. For dmCpGs associated with offspring BMI, 2 or more sites from DIP2C, NFYA and QSEC3 were identified both at change FBS-V8 and FBS-V30. This suggests that they play a key role in body composition change during childhood and adolescence. Furthermore, voice break-related dmCpGs FERMT1, NCK2 (adipogenesis), MOCS1 and VSIG10 show a clear separation between normal and overweight FBS, suggesting a link with intergenerational BMI.

The main strength of this study is that we have been able to specify the timing of preconception overweight across childhood, voice break and early adulthood in many fathers and have related this to rich data from their offspring including DNA methylation measurements. The current and retrospective body silhouettes have been validated against measured height and weight at different time points in adulthood. Moreover, when we compared the regression coefficients between the offspring's current body silhouette and current BMI in the present study, they showed a correlation r = 0.97 with p < 2.2e−16, which further confirms the validity of the body silhouette approach to measure adiposity. Also, remarkably, many of the annotated genes have previously been linked to obesity and BMI in epigenetic studies on mature spermatozoa1517 which adds credibility that the differentially methylated sites identified are truly associated with the father’s preconception body composition and overweight, thus represent potential candidates for validation in other studies.

We also acknowledge that our study faces several limitations. The study results are yet to be confirmed in an independent cohort; thus, our findings need further validation. Furthermore, some of the EWAS models assessing timepoints and trajectories of FBS in our study have small sample sizes; despite this, however, significant associations were still identified. Our offspring study population had a large age range, and the subjects come from different study centres and therefore can be considered a heterogenous population. However, we did include age and study centre as covariates in the EWAS regression models to mitigate against these effects and in the RHINESSA cohort. Regarding mothers’ overweight status in different time periods and other preconception maternal factors, these were not considered true confounders in the analyses. Previous analyses found these were not associated with adult offspring’s asthma3 and there is minimal overlap in dmCpGs associated with maternal smoking and those associated with paternal BDS (Supplementary Data 9). Potential reverse confounding can however not be excluded.

This study reveals important associations between a father’s body silhouette across adolescence and offspring DNA methylation, which strongly supports the idea that a father’s preconception metabolic status can impact the epigenome of his future offspring. Further, our study supports the view that the period around puberty may be a particularly susceptible age window for such impact, a concept that may be a game-changer in public health intervention strategies. The identified DNA methylation patterns are related to key signalling pathways known to contribute to obesity pathogenesis and related functions such as insulin regulation, glucose metabolism, adipogenesis, body form, telomere maintenance, asthma and lung function. Our findings showed a 35.7% overlap with previously reported loci linked to obesity, which suggests that about 64% of our genes are novel associations. These sites have the potential to serve as predictive biomarkers for population studies screening for metabolic and respiratory disease, and as therapeutic targets for intervention.

Supplementary information

Supplementary Information (304.9KB, pdf)
Reporting summary (73.2KB, pdf)

Acknowledgements

We thank all the study participants, fieldworkers and scientists in RHINESSA, Co-ordination of the RHINESSA study has received funding from the Research Council of Norway (Grants Nos. 274767, 214123, 228174, 230827 and 273838), ERC StG project BRuSH #804199, the European Union’s Horizon 2020 research and innovation programme under grant agreement No. 633212 (the ALEC Study), the Bergen Medical Research Foundation, and the Western Norwegian Regional Health Authorities (Grants Nos. 912011, 911892 and 911631). Study centres have further received local funding from the following: Bergen: the above grants for study establishment and co-ordination, and, in addition, World University Network (REF and Sustainability grants), Norwegian Labour Inspection, and the Norwegian Asthma and Allergy Association. Albacete and Huelva: Sociedad Española de Patología Respiratoria (SEPAR) Fondo de Investigación Sanitaria (FIS PS09). Gøteborg, Umeå and Uppsala: the Swedish Heart and Lung Foundation, the Swedish Asthma and Allergy Association. Reykjavik: Iceland University. Melbourne: National Health and Medical Research Council (NHMRC) of Australia (research grants 299901 and 1021275). Tartu: The Estonian Research Council (Grant No. PUT562). Århus: The Danish Wood Foundation (Grant No. 444508795), the Danish Working Environment Authority (Grant No. 20150067134), Aarhus University (Ph.D. scholarship). The body silhouette image used in the graphic abstract and Supplementary Fig. 1 was generated using Clip Studio Paint and kindly provided by Alejandro Villén Real (alejandrovillen@gmail.com).

Author contributions

C.S., T.M.Ø., M.L., S.A., F.G.R., N.T.K. and J.W.H. contributed to conceptualisation. T.M.Ø., M.L., S.A., N.T.K. and J.W.H. performed data curation. N.T.K., C.S. and J.W.H. carried out formal analysis. N.T.K., S.A., C.S. and J.W.H. provided methodology. C.S. performed project administration. N.T.K., T.M.Ø., M.L., F.G.R., C.S. and J.W.H. performed writing—original draft. N.T.K., T.M.Ø., M.L., S.A., F.G.R., A.M., A.O., B.B., F.J.C.G., L.P.G., M.H., N.O.J., S.C.D., S.M.S., V.S., C.S. and J.W.H. contributed to writing—review, editing and final approval.

Peer review

Peer review information

Communications Biology thanks Nady El Hajj and the other, anonymous, reviewer for their contribution to the peer review of this work. Primary Handling Editors: Chien-Yu Chen and Kaliya Georgieva. A peer review file is available.

Funding

Open access funding provided by University of Bergen.

Data availability

Supplementary Data 114 and summary statistics for epigenome-wide association analyses are available from 10.5258/SOTON/D3067. The full data cannot be shared openly in order to protect study participants’ privacy, but an anonymised, de-identified version with limited data can be made available on request to allow all results to be reproduced. All requests should be directed to CS, the RHINESSA Study Principal Investigator.

Code availability

The custom code used to generate graphics are available at GitHub repository: https://github.com/negusse2025/EWAS-of-Father-s-adolescent-body-silhouette-.git.

Competing interests

The authors declare no competing interests.

Footnotes

Publisher’s note Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

These authors contributed equally: Cecilie Svanes, John W. Holloway.

Supplementary information

The online version contains supplementary material available at 10.1038/s42003-025-08121-9.

References

  • 1.Bentham, J. et al. Worldwide trends in body-mass index, underweight, overweight, and obesity from 1975 to 2016: a pooled analysis of 2416 population-based measurement studies in 128·9 million children, adolescents, and adults. Lancet390, 2627–2642 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Lønnebotn, M. et al. Parental prepuberty overweight and offspring lung function. Nutrients14, 1506 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Johannessen, A. et al. Being overweight in childhood, puberty, or early adulthood: changing asthma risk in the next generation?. J. Allergy Clin. Immunol.145, 791–799.e4 (2020). [DOI] [PubMed] [Google Scholar]
  • 4.Bowatte, G. et al. Parental preconception BMI trajectories from childhood to adolescence and asthma in the future offspring. J. Allergy Clin. Immunol.150, 67–74.e30 (2022). [DOI] [PubMed] [Google Scholar]
  • 5.Kaati, G. et al. Cardiovascular and diabetes mortality determined by nutrition during parents’ and grandparents’ slow growth period. Eur. J. Hum. Genet.10, 682–688 (2002). [DOI] [PubMed] [Google Scholar]
  • 6.Pembrey, M. E. et al. Sex-specific, male-line transgenerational responses in humans. Eur. J. Hum. Genet.14, 159–166 (2006). [DOI] [PubMed] [Google Scholar]
  • 7.Northstone, K. et al. Prepubertal start of father’s smoking and increased body fat in his sons: further characterisation of paternal transgenerational responses. Eur. J. Hum. Genet.22, 1382–1386 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Ornellas, F. et al. Obese fathers lead to an altered metabolism and obesity in their children in adulthood: review of experimental and human studies. J. Pediatr. (Rio J.)93, 551–559 (2017). [DOI] [PubMed] [Google Scholar]
  • 9.Kitaba, N. T. et al. Fathers’ preconception smoking and offspring DNA methylation. Clin. Epigenet.15, 131 (2023). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Wu, H. et al. Hauser, R. Environmental Susceptibility of the Sperm Epigenome During Windows of Male Germ Cell Development. Curr. Environ. Health Rep.2, 356–366 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Soubry, A. et al. A paternal environmental legacy: Evidence for epigenetic inheritance through the male germ line. BioEssays36, 359–371 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Klibaner-Schiff, E. et al. Environmental exposures influence multigenerational epigenetic transmission. Clin. Epigenet.16, 145 (2024). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Soubry, A. POHaD: why we should study future fathers. Environ. Epigenet.4, dvy007 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Panzeri, I. & Pospisilik, J. A. Epigenetic control of variation and stochasticity in metabolic disease. Mol. Metab.14, 26–38 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Donkin, I. et al. Obesity and bariatric surgery drive epigenetic variation of spermatozoa in humans. Cell Metab.23, 369–378 (2016). [DOI] [PubMed] [Google Scholar]
  • 16.Potabattula, R. et al. Male obesity effects on sperm and next-generation cord blood DNA methylation. PLoS ONE14, 1–13 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Keyhan, S. et al. Male obesity impacts DNA methylation reprogramming in sperm. Clin. Epigenet.13, 17 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Tomar, A. et al. Epigenetic inheritance of diet-induced and sperm-borne mitochondrial RNAs. Nature630, 720–727 (2024). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Simmons, L. W. et al. Condition-dependent seminal fluid gene expression and intergenerational paternal effects on ejaculate quality. Funct. Ecol.36, 798–811 (2022). [Google Scholar]
  • 20.Lassi, M. et al. Disruption of paternal circadian rhythm affects metabolic health in male offspring via nongerm cell factors. Sci. Adv.7, eabg6424 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Soubry, A. et al. Newborns of obese parents have altered DNA methylation patterns at imprinted genes. Int. J. Obes.39, 650–657 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Soubry, A. et al. Paternal obesity is associated with IGF2hypomethylation in newborns: results from a Newborn Epigenetics Study (NEST) cohort. BMC Med.11, 29 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Noor, N. et al. Association of periconception paternal body mass index with persistent changes in DNA methylation of offspring in childhood. JAMA Netw. Open2, e1916777 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Lønnebotn, M. et al. Body silhouettes as a tool to reflect obesity in the past. PLoS ONE13, 1–13 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Svanes, C. et al. Cohort profile: the multigeneration Respiratory Health in Northern Europe, Spain and Australia (RHINESSA) cohort. BMJ Open12, e059434 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Dratva, J. et al. Validation of self-reported figural drawing scales against anthropometric measurements in adults. Public Health Nutr.19, 1944–1951 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Miller, M. R. Standardisation of spirometry. Eur. Respir. J.26, 319–338 (2005). [DOI] [PubMed] [Google Scholar]
  • 28.Miller, S. A., Dykes, D. D. & Polesky, H. F. A simple salting out procedure for extracting DNA from human nucleated cells. Nucleic Acids Res.16, 1215 (1988). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.R. Core Team. R: A Language and Environment For Statistical Computing (R Foundation for Statical Computing, Vienna, Austria, 2020).
  • 30.Aryee, M. J. et al. Minfi: a flexible and comprehensive Bioconductor package for the analysis of Infinium DNA methylation microarrays. Bioinformatics30, 1363–1369 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Min, J. L. et al. Meffil: efficient normalization and analysis of very large DNA methylation datasets. Bioinformatics44, 1–7 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Teschendorff, A. E. et al. An epigenetic signature in peripheral blood predicts active ovarian cancer. PLoS ONE4, e8274 (2009). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Morris, T. J. et al. ChAMP: 450k chip analysis methylation pipeline. Bioinformatics30, 428–430 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Leek, J. T. et al. The SVA package for removing batch effects and other unwanted variation in high-throughput experiments. Bioinformatics28, 882–883 (2012). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Zheng, S. C. et al. EpiDISH web server: epigenetic dissection of intra-sample-heterogeneity with online GUI. Bioinformatics36, 1950–1951 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Teschendorff, A. E. et al. A beta-mixture quantile normalization method for correcting probe design bias in Illumina Infinium 450 k DNA methylation data. Bioinformatics29, 189–196 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Zhou, W., Laird, P. W. & Shen, H. Comprehensive characterization, annotation and innovative use of Infinium DNA methylation BeadChip probes. Nucleic Acids Res.45, e22 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Nordlund, J. et al. Genome-wide signatures of differential DNA methylation in pediatric acute lymphoblastic leukemia. Genome Biol.14, r105 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Pidsley, R. et al. Critical evaluation of the Illumina MethylationEPIC BeadChip microarray for whole-genome DNA methylation profiling. Genome Biol.17, 208 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Ritchie, M. E. et al. limma powers differential expression analyses for RNA-sequencing and microarray studies. Nucleic Acids Res.43, e47 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Barton, S. J. et al. In Epigenomic studies, including cell-type adjustments in regression models can introduce multicollinearity, resulting in apparent reversal of direction of association. Front. Genet.10, 816 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Benjamini, Y. & Hochberg, Y. Controlling the false discovery rate: a practical and powerful approach to multiple testing. J. R. Stat. Soc. Ser. B Methodol.57, 289–300 (1995). [Google Scholar]
  • 43.D. Turner, S. qqman: an R package for visualizing GWAS results using Q–Q and Manhattan plots. J. Open Source Softw. 3, 731 (2018).
  • 44.van Iterson, M., van Zwet, E. W. & Heijmans, B. T. Controlling bias and inflation in epigenome- and transcriptome-wide association studies using the empirical null distribution. Genome Biol. 18, 19 (2017). [DOI] [PMC free article] [PubMed]
  • 45.Peters, T. J. et al. De novo identification of differentially methylated regions in the human genome. Epigenet. Chromatin8, 6 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Suderman, M. et al. Dmrff: Identifying differentially methylated regions efficiently with power and control. bioRxiv 508556 10.1101/508556 (2018).
  • 47.Breeze, C. E. et al. eFORGE v2.0: updated analysis of cell type-specific signal in epigenomic data. Bioinformatics35, 1–3 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Carvalho-Silva, D. et al. Open Targets Platform: new developments and updates two years on. Nucleic Acids Res.47, D1056–D1065 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Li, M. et al. EWAS Atlas: a curated knowledgebase of epigenome-wide association studies. Nucleic Acids Res.47, D983–D988 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Szklarczyk, D. et al. STRING v11: protein–protein association networks with increased coverage, supporting functional discovery in genome-wide experimental datasets. Nucleic Acids Res.47, D607–D613 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Xie, Z. et al. Gene set knowledge discovery with enrichr. Curr. Protoc.1, e90 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Bindea, G. et al. ClueGO: a cytoscape plug-in to decipher functionally grouped gene ontology and pathway annotation networks. Bioinformatics25, 1091–1093 (2009). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Shannon, P. et al. Cytoscape: a software environment for integrated models of bimolecular interaction networks. Genome Res.13, 2498–2504 (2003). [DOI] [PMC free article] [PubMed]
  • 54.Ren, X. & Kuan, P. F. methylGSA: a Bioconductor package and Shiny app for DNA methylation data length bias adjustment in gene set testing. Bioinformatics35, 1958–1959 (2019). [DOI] [PubMed] [Google Scholar]
  • 55.Phipson, B., Maksimovic, J. & Oshlack, A. MissMethyl: an R package for analyzing data from Illumina’s HumanMethylation450 platform. Bioinformatics32, 286–288 (2016). [DOI] [PubMed] [Google Scholar]
  • 56.Watanabe, K. et al. Functional mapping and annotation of genetic associations with FUMA. Nat. Commun.8, 1826 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Min, J. L. et al. Genomic and phenotypic insights from an atlas of genetic effects on DNA methylation. Nat. Genet.53, 1311–1321 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Villicaña, S. et al. Genetic impacts on DNA methylation help elucidate regulatory genomic processes. Genome Biol.24, 176 (2023). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Silver, M. J. et al. Environmentally sensitive hotspots in the methylome of the early human embryo. eLife11, e72031 (2022). [DOI] [PMC free article] [PubMed]
  • 60.King, S. E. & Skinner, M. K. Epigenetic transgenerational inheritance of obesity susceptibility. Trends Endocrinol. Metab.31, 478–494 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Shungin, D. et al. New genetic loci link adipose and insulin biology to body fat distribution. Nature518, 187–196 (2015). [DOI] [PMC free article] [PubMed]
  • 62.Christakoudi, S. et al. GWAS of allometric body‑shape indices in UK Biobank identifies loci suggesting associations with morphogenesis, organogenesis, adrenal cell renewal and cancer. Sci. Rep.11, 10688 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Li, Y.-K. et al. Convergent evidence supports TH2LCRR as a novel asthma susceptibility gene. Am. J. Respir. Cell Mol. Biol.66, 283–292 (2022). [DOI] [PubMed] [Google Scholar]
  • 64.Farook, V. S. et al. Molecular analysis of KCNJ10 on 1q as a candidate gene for type 2 diabetes in Pima Indians. Diabetes51, 3342–3346 (2002). [DOI] [PubMed] [Google Scholar]
  • 65.Yin, R.-X. et al. Several genetic polymorphisms interact with overweight/obesity to influence serum lipid levels. Cardiovasc. Diabetol.11, 123 (2012). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Khetarpal, S. A. et al. Mining the LIPG allelic spectrum reveals the contribution of rare and common regulatory variants to HDL cholesterol. PLoS Genet.7, e1002393 (2011). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.Yin, R. X. et al. Several lipid-related gene polymorphisms interact with overweight/obesity to modulate blood pressure levels. Int. J. Mol. Sci.13, 12062–12081 (2012). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Paradis, M.-E. et al. Visceral adiposity and endothelial lipase. J. Clin. Endocrinol. Metab.91, 3538–3543 (2006). [DOI] [PubMed] [Google Scholar]
  • 69.Rong, X. et al. Lpcat3-dependent production of arachidonoyl phospholipids is a key determinant of triglyceride secretion. eLife2015, 1–23 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Sharma, N. K. et al. Phospholipid biosynthesis genes and susceptibility to obesity: analysis of expression and polymorphisms. PLoS ONE8, e65303 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.Stöckli, J. et al. Regulation of glucose transporter 4 translocation by the rab guanosine triphosphatase-activating protein AS160/TBC1D4: role of phosphorylation and membrane association. Mol. Endocrinol.22, 2703–2715 (2008). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Dash, S. et al. Analysis of TBC1D4 in patients with severe insulin resistance. Diabetologia53, 1239–1242 (2010). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73.Overvad, M. et al. The effect of diabetes and the common diabetogenic TBC1D4 p.Arg684Ter variant on cardiovascular risk in Inuit in Greenland. Sci. Rep.10, 22081 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74.Larsen, J. K. et al. Illumination of the endogenous insulin-regulated TBC1D4 interactome in human skeletal muscle. Diabetes71, 906–920 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.Haider, N. et al. Nck2, an unexpected regulator of adipogenesis. Adipocyte6, 154–160 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76.Dusseault, J. et al. Nck2 deficiency in mice results in increased adiposity associated with adipocyte hypertrophy and enhanced adipogenesis. Diabetes65, 2652–2666 (2016). [DOI] [PubMed] [Google Scholar]
  • 77.Rzehak, P. et al. DNA-methylation and body composition in preschool children: epigenome-wide-analysis in the European Childhood Obesity Project (CHOP)-Study. Sci. Rep.7, 14349 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78.Zhang, X. et al. FOXO3a regulates lipid accumulation and adipocyte inflammation in adipocytes through autophagy: role of FOXO3a in obesity. Inflamm. Res.70, 591–603 (2021). [DOI] [PubMed] [Google Scholar]
  • 79.Kobayashi, M. et al. Identification of WWP1 as an obesity-associated E3 ubiquitin ligase with a protective role against oxidative stress in adipocytes. Biochem. Biophys. Res. Commun.508, 117–122 (2019). [DOI] [PubMed] [Google Scholar]
  • 80.Hoshino, S. et al. WWP1 knockout in mice exacerbates obesity-related phenotypes in white adipose tissue but improves whole-body glucose metabolism. FEBS Open Bio10, 306–315 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81.Rask-Andersen, M., Karlsson, T., Ek, W. E. & Johansson, Å. Genome-wide association study of body fat distribution identifies adiposity loci and sex-specific genetic effects. Nat. Commun.10, 339 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82.Waldrop, S. W. et al. Differentially methylated regions interrogated for metastable epialleles associate with offspring adiposity. Epigenomics16, 1215–1230 (2024). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83.Wang, Z. et al. Evolution of digestive enzymes and RNASE1 provides insights into dietary switch of Cetaceans. Mol. Biol. Evol.33, 3144–3157 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84.Comuzzie, A. G. et al. Novel genetic loci identified for the pathophysiology of childhood obesity in the hispanic population. PLoS ONE7, e51954 (2012). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85.Kettunen, J. et al. Genome-wide association study identifies multiple loci influencing human serum metabolite levels. Nat. Genet.44, 269–276 (2012). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86.Luykx, J. J. et al. Genome-wide association study of monoamine metabolite levels in human cerebrospinal fluid. Mol. Psychiatry19, 228–234 (2014). [DOI] [PubMed] [Google Scholar]
  • 87.Lung, M. et al. Diacylglycerol kinase ϵ is selective for both acyl chains of phosphatidic acid or diacylglycerol. J. Biol. Chem.284, 31062–31073 (2009). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88.Wang, S. et al. HIF3A DNA methylation is associated with childhood obesity and ALT. PLoS ONE10, 1–9 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89.Zapata, R. C., Nasamran, C. A., Chilin-Fuentes, D. R., Dulawa, S. C. & Osborn, O. Identification of adipose tissue transcriptomic memory of anorexia nervosa. Mol. Med.29, 109 (2023). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90.Patel, P., Selvaraju, V., Babu, J. R., Wang, X. & Geetha, T. Novel differentially methylated regions identified by genome-wide DNA methylation analyses contribute to racial disparities in childhood obesity. Genes14, 1098 (2023). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91.Scudiero, O. et al. Childhood obesity: an overview of laboratory medicine, exercise and microbiome. Clin. Chem. Lab. Med.58, 1385–1406 (2020). [DOI] [PubMed] [Google Scholar]
  • 92.Selvanayagam, T. et al. Genome-wide copy number variation analysis identifies novel candidate loci associated with pediatric obesity. Eur. J. Hum. Genet.26, 1588–1596 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93.Lee, S. The association of genetically controlled CpG methylation (cg158269415) of protein tyrosine phosphatase, receptor type N2 (PTPRN2) with childhood obesity. Sci. Rep.9, 1–7 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94.Liu, Y. et al. Obesity-induced endoplasmic reticulum stress suppresses nuclear factor-Y expression. Mol. Cell. Biochem.426, 47–54 (2017). [DOI] [PubMed] [Google Scholar]
  • 95.Nishi-Tatsumi, M. et al. A key role of nuclear factor Y in the refeeding response of fatty acid synthase in adipocytes. FEBS Lett.591, 965–978 (2017). [DOI] [PubMed] [Google Scholar]
  • 96.Lu, Y.-H. et al. Nuclear Factor-Y is an adipogenic factor that regulates leptin gene expression. Mol. Metab.4, 392–405 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 97.Buxton, J. L. et al. Childhood obesity is associated with shorter leukocyte telomere length. J. Clin. Endocrinol. Metab.96, 1500–1505 (2011). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 98.Buxton, J. L. et al. Human leukocyte telomere length is associated with DNA methylation levels in multiple subtelomeric and imprinted loci. Sci. Rep.4, 1–8 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99.Tzanetakou, I. P. et al. Is obesity linked to aging?’. Adipose tissue and the role of telomeres. Ageing Res. Rev.11, 220–229 (2012). [DOI] [PubMed] [Google Scholar]
  • 100.Zgheib, N. K. et al. Short telomere length is associated with aging, central obesity, poor sleep and hypertension in Lebanese individuals. Aging Dis.9, 77–89 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 101.Gielen, M. et al. Body mass index is negatively associated with telomere length: a collaborative cross-sectional meta-analysis of 87 observational studies. Am. J. Clin. Nutr.108, 453–475 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 102.Hancock, D. B. et al. Meta-analyses of genome-wide association studies identify multiple novel loci associated with pulmonary function. Nat. Genet.42, 45–52 (2010). [DOI] [PMC free article] [PubMed]
  • 103.Weedon, N. M. et al. Genome-wide association analysis identifies 20 loci that influence adult height. Nat. Genet.40, 575–583 (2008). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 104.Shi, Y. & Long, F. Hedgehog signaling via Gli2 prevents obesity induced by high-fat diet in adult mice. eLife6, e31649 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 105.Wang, L. et al. Identification of potentially common loci between childhood obesity and coronary artery disease using pleiotropic approaches. Sci. Rep.12, 19513 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 106.Wong, H. S.-C. et al. Genome-wide association study identifies genetic risk loci for adiposity in a Taiwanese population. PLoS Genet.18, e1009952 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 107.Mizuarai, S. et al. Identification of dicarboxylate carrier Slc25a10 as malate transporter in de novo fatty acid synthesis. J. Biol. Chem.280, 32434–32441 (2005). [DOI] [PubMed] [Google Scholar]
  • 108.Kulyté, A. et al. Global transcriptome profiling identifies KLF15 and SLC25A10 as modifiers of adipocytes insulin sensitivity in obese women. PLoS ONE12, e0178485 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 109.Bindra, D. & Mishra, R. K. In pursuit of distinctiveness: transmembrane nucleoporins and their disease associations. Front. Oncol.11, 784319 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 110.Errafii, K., Boujraf, S. & Chikri, M. Transcriptomic analysis from normal glucose tolerance to T2D of obese individuals using bioinformatic tools. Int. J. Mol. Sci.24, 6337 (2023). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 111.Moraes, R. D. A., Webb, R. C. & Silva, D. F. Vascular dysfunction in diabetes and obesity: focus on TRP channels. Front. Physiol.12, 645109 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 112.Zsombok, A. & Derbenev, A. TRP channels as therapeutic targets in diabetes and obesity. Pharmaceuticals9, 50 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 113.Agarwal, A. K. et al. Regulated adipose tissue-specific expression of human AGPAT2 in lipodystrophic Agpat2-null mice results in regeneration of adipose tissue. iScience26, 107806 (2023). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 114.Subauste, A. R. et al. Alterations in lipid signaling underlie lipodystrophy secondary to AGPAT2 mutations. Diabetes61, 2922–2931 (2012). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 115.Agarwal, A. K. Lysophospholipid acyltransferases: 1-acylglycerol-3-phosphate O-acyltransferases. From discovery to disease. Curr. Opin. Lipidol.23, 290–302 (2012). [DOI] [PubMed] [Google Scholar]
  • 116.Triantafyllou, E.-A. et al. Expression of AGPAT2, an enzyme involved in the glycerophospholipid/triacylglycerol biosynthesis pathway, is directly regulated by HIF-1 and promotes survival and etoposide resistance of cancer cells under hypoxia. Biochim. Biophys. Acta BBA - Mol. Cell Biol. Lipids1863, 1142–1152 (2018). [DOI] [PubMed] [Google Scholar]
  • 117.Lemaitre, R. N. et al. Genetic loci associated with plasma phospholipid n-3 fatty acids: a meta-analysis of genome-wide association studies from the CHARGE Consortium. PLoS Genet.7, e1002193 (2011). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 118.de sá, P. M., Richard, A. J., Hang, H. & Stephens, J. M. Transcriptional regulation of adipogenesis. Compr. Physiol.7, 635–674 (2017). [DOI] [PubMed] [Google Scholar]
  • 119.Timóteo, A. T. Can PM20D1 be a new kid on the block in cardiovascular risk stratification? Do not run before you can walk. Arq. Bras. Cardiol.119, 380–381 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 120.Long, J. Z. et al. The secreted enzyme PM20D1 regulates lipidated amino acid uncouplers of mitochondria. Cell166, 424–435 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 121.Paul, L. Wasting energy to treat obesity. N. Engl. J. Med.375, 2298–2300 (2016). [DOI] [PubMed]
  • 122.Vrang, N. et al. The imprinted gene Neuronatin is regulated by metabolic status and associated with obesity. Obesity18, 1289–1296 (2010). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 123.Millership, S. J. et al. Neuronatin deletion causes postnatal growth restriction and adult obesity in 129S2/Sv mice. Mol. Metab.18, 97–106 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 124.Cimino, I. et al. Murine neuronatin deficiency is associated with a hypervariable food intake and bimodal obesity. Sci. Rep.11, 17571 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 125.Van Dijk, S. J. et al. DNA methylation in blood from neonatal screening cards and the association with BMI and insulin sensitivity in early childhood. Int. J. Obes.42, 28–35 (2018). [DOI] [PubMed] [Google Scholar]
  • 126.Butler, M. G. Genomic imprinting disorders in humans: a mini-review. J. Assist. Reprod. Genet.26, 477–486 (2009). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 127.Jima, D. D. et al. Genomic map of candidate human imprint control regions: the imprintome. Epigenetics00, 1–24 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 128.Joshi, R. S. et al. DNA methylation profiling of uniparental disomy subjects provides a map of parental epigenetic bias in the human genome. Am. J. Hum. Genet.99, 555–566 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 129.Marttila, S. et al. Methylation status of VTRNA2-1 / nc886 is stable across populations, monozygotic twin pairs and in majority of tissues. Epigenomics14, 1105–1124 (2022). [DOI] [PubMed] [Google Scholar]
  • 130.Dugué, P.-A. et al. VTRNA2-1: genetic variation, heritable methylation and disease association. Int. J. Mol. Sci.22, 2535 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 131.Yang, J. et al. Clopidogrel resistance is associated with DNA methylation of genes from whole blood of humans. Front. Genet.11, 583215 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 132.Lin, C.-H. et al. Methylation status of vault RNA 2-1 promoter is a predictor of glycemic response to glucagon-like peptide-1 analog therapy in type 2 diabetes mellitus. BMJ Open Diabetes Res. Care9, e001416 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 133.Silver, M. J. et al. Independent genomewide screens identify the tumor suppressor VTRNA2-1 as a human epiallele responsive to periconceptional environment. Genome Biol.16, 118 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 134.Marttila, S. et al. Methylation status of nc886 epiallele reflects periconceptional conditions and is associated with glucose metabolism through nc886 RNAs. Clin. Epigenet.13, 143 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 135.Modena, B. D. et al. Gene expression correlated with severe asthma characteristics reveals heterogeneous mechanisms of severe disease. Am. J. Respir. Crit. Care Med.195, 1449–1463 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 136.Tremblay, K. et al. Genes to diseases (G2D) computational method to identify asthma candidate genes. PLoS ONE3, e2907 (2008). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 137.Busch, R. et al. Differential DNA methylation marks and gene comethylation of COPD in African-Americans with COPD exacerbations. Respir. Res.17, 1–15 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 138.Li, B. et al. The transcriptome characteristics of severe asthma from the prospect of co-expressed gene modules. Front. Genet.12, 1–11 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 139.Himes, B. E. et al. Association of SERPINE2 with asthma. Chest140, 667–674 (2011). [DOI] [PMC free article] [PubMed]

Associated Data

This section collects any data citations, data availability statements, or supplementary materials included in this article.

Supplementary Materials

Supplementary Information (304.9KB, pdf)
Reporting summary (73.2KB, pdf)

Data Availability Statement

Supplementary Data 114 and summary statistics for epigenome-wide association analyses are available from 10.5258/SOTON/D3067. The full data cannot be shared openly in order to protect study participants’ privacy, but an anonymised, de-identified version with limited data can be made available on request to allow all results to be reproduced. All requests should be directed to CS, the RHINESSA Study Principal Investigator.

The custom code used to generate graphics are available at GitHub repository: https://github.com/negusse2025/EWAS-of-Father-s-adolescent-body-silhouette-.git.


Articles from Communications Biology are provided here courtesy of Nature Publishing Group

RESOURCES