scispace - formally typeset
Search or ask a question

Showing papers by "Marc Jan Bonder published in 2017"


Journal ArticleDOI
Simone Wahl, Alexander W. Drong1, Benjamin Lehne2, Marie Loh3, Marie Loh2, Marie Loh4, William R. Scott2, William R. Scott5, Sonja Kunze, Pei-Chien Tsai6, Janina S. Ried, Weihua Zhang2, Weihua Zhang7, Youwen Yang2, Sili Tan8, Giovanni Fiorito9, Lude Franke10, Simonetta Guarrera9, Silva Kasela11, Jennifer Kriebel, Rebecca C Richmond12, Marco Adamo13, Uzma Afzal2, Uzma Afzal7, Mika Ala-Korpela12, Mika Ala-Korpela14, Mika Ala-Korpela3, Benedetta Albetti15, Ole Ammerpohl16, Jane F. Apperley2, Marian Beekman17, Pier Alberto Bertazzi15, S. Lucas Black2, Christine Blancher1, Marc Jan Bonder10, Mario Brosch18, Maren Carstensen-Kirberg19, Anton J. M. de Craen17, Simon de Lusignan20, Abbas Dehghan21, Mohamed Elkalaawy13, Krista Fischer11, Oscar H. Franco21, Tom R. Gaunt12, Jochen Hampe18, Majid Hashemi13, Aaron Isaacs21, Andrew Jenkinson13, Sujeet Jha22, Norihiro Kato, Vittorio Krogh, Michael Laffan2, Christa Meisinger, Thomas Meitinger23, Zuan Yu Mok8, Valeria Motta15, Hong Kiat Ng8, Zacharoula Nikolakopoulou5, Georgios Nteliopoulos2, Salvatore Panico24, Natalia Pervjakova11, Holger Prokisch23, Wolfgang Rathmann19, Michael Roden19, Federica Rota15, Michelle Ann Rozario8, Johanna K. Sandling25, Johanna K. Sandling26, Clemens Schafmayer, Katharina Schramm23, Reiner Siebert16, Reiner Siebert27, P. Eline Slagboom17, Pasi Soininen14, Pasi Soininen3, Lisette Stolk21, Konstantin Strauch28, E-Shyong Tai8, Letizia Tarantini15, Barbara Thorand, Ettje F. Tigchelaar10, Rosario Tumino, André G. Uitterlinden21, Cornelia M. van Duijn21, Joyce B. J. van Meurs21, Paolo Vineis, Ananda R. Wickremasinghe29, Cisca Wijmenga10, Tsun-Po Yang26, Wei Yuan6, Wei Yuan30, Alexandra Zhernakova10, Rachel L. Batterham13, George Davey Smith12, Panos Deloukas26, Panos Deloukas31, Panos Deloukas32, Bastiaan T. Heijmans17, Christian Herder19, Albert Hofman21, Cecilia M. Lindgren33, Cecilia M. Lindgren1, Lili Milani11, Pim van der Harst10, Annette Peters, Thomas Illig, Caroline L Relton12, Melanie Waldenberger, Marjo-Riitta Järvelin34, Valentina Bollati15, Richie Soong8, Tim D. Spector6, James Scott5, Mark I. McCarthy35, Mark I. McCarthy36, Mark I. McCarthy1, Paul Elliott2, Paul Elliott37, Jordana T. Bell6, Giuseppe Matullo9, Christian Gieger, Jaspal S. Kooner5, Harald Grallert, John C. Chambers 
05 Jan 2017-Nature
TL;DR: In this article, the authors used epigenome-wide association to show that body mass index (BMI), a key measure of adiposity, is associated with widespread changes in DNA methylation.
Abstract: Approximately 1.5 billion people worldwide are overweight or affected by obesity, and are at risk of developing type 2 diabetes, cardiovascular disease and related metabolic and inflammatory disturbances1,2. Although the mechanisms linking adiposity to associated clinical conditions are poorly understood, recent studies suggest that adiposity may influence DNA methylation3,4,5,6, a key regulator of gene expression and molecular phenotype7. Here we use epigenome-wide association to show that body mass index (BMI; a key measure of adiposity) is associated with widespread changes in DNA methylation (187 genetic loci with P < 1 × 10−7, range P = 9.2 × 10−8 to 6.0 × 10−46; n = 10,261 samples). Genetic association analyses demonstrate that the alterations in DNA methylation are predominantly the consequence of adiposity, rather than the cause. We find that methylation loci are enriched for functional genomic features in multiple tissues (P < 0.05), and show that sentinel methylation markers identify gene expression signatures at 38 loci (P < 9.0 × 10−6, range P = 5.5 × 10−6 to 6.1 × 10−35, n = 1,785 samples). The methylation loci identify genes involved in lipid and lipoprotein metabolism, substrate transport and inflammatory pathways. Finally, we show that the disturbances in DNA methylation predict future development of type 2 diabetes (relative risk per 1 standard deviation increase in methylation risk score: 2.3 (2.07–2.56); P = 1.1 × 10−54). Our results provide new insights into the biologic pathways influenced by adiposity, and may enable development of new strategies for prediction and prevention of type 2 diabetes and other adverse clinical consequences of obesity.

667 citations


Journal ArticleDOI
TL;DR: It is shown that disease-associated variants have widespread effects on DNA methylation in trans that likely reflect differential occupancy of trans binding sites by cis-regulated transcription factors.
Abstract: Most disease-associated genetic variants are noncoding, making it challenging to design experiments to understand their functional consequences. Identification of expression quantitative trait loci (eQTLs) has been a powerful approach to infer the downstream effects of disease-associated variants, but most of these variants remain unexplained. The analysis of DNA methylation, a key component of the epigenome, offers highly complementary data on the regulatory potential of genomic regions. Here we show that disease-associated variants have widespread effects on DNA methylation in trans that likely reflect differential occupancy of trans binding sites by cis-regulated transcription factors. Using multiple omics data sets from 3,841 Dutch individuals, we identified 1,907 established trait-associated SNPs that affect the methylation levels of 10,141 different CpG sites in trans (false discovery rate (FDR) < 0.05). These included SNPs that affect both the expression of a nearby transcription factor (such as NFKB1, CTCF and NKX2-3) and methylation of its respective binding site across the genome. Trans methylation QTLs effectively expose the downstream effects of disease-associated variants.

380 citations


Journal ArticleDOI
TL;DR: This work generated peripheral blood RNA–seq data from 2,116 unrelated individuals and systematically identified context-dependent eQTLs using a hypothesis-free strategy that does not require previous knowledge of the identity of the modifiers.
Abstract: Genetic risk factors often localize to noncoding regions of the genome with unknown effects on disease etiology. Expression quantitative trait loci (eQTLs) help to explain the regulatory mechanisms underlying these genetic associations. Knowledge of the context that determines the nature and strength of eQTLs may help identify cell types relevant to pathophysiology and the regulatory networks underlying disease. Here we generated peripheral blood RNA-seq data from 2,116 unrelated individuals and systematically identified context-dependent eQTLs using a hypothesis-free strategy that does not require previous knowledge of the identity of the modifiers. Of the 23,060 significant cis-regulated genes (false discovery rate (FDR) ≤ 0.05), 2,743 (12%) showed context-dependent eQTL effects. The majority of these effects were influenced by cell type composition. A set of 145 cis-eQTLs depended on type I interferon signaling. Others were modulated by specific transcription factors binding to the eQTL SNPs.

373 citations


Journal ArticleDOI
TL;DR: An epigenetic predictor of age in mice is identified and characterised, which will be instrumental for understanding the biology of ageing and will allow modulation of its ticking rate and resetting the clock in vivo to study the impact on biological age.
Abstract: DNA methylation changes at a discrete set of sites in the human genome are predictive of chronological and biological age. However, it is not known whether these changes are causative or a consequence of an underlying ageing process. It has also not been shown whether this epigenetic clock is unique to humans or conserved in the more experimentally tractable mouse. We have generated a comprehensive set of genome-scale base-resolution methylation maps from multiple mouse tissues spanning a wide range of ages. Many CpG sites show significant tissue-independent correlations with age which allowed us to develop a multi-tissue predictor of age in the mouse. Our model, which estimates age based on DNA methylation at 329 unique CpG sites, has a median absolute error of 3.33 weeks and has similar properties to the recently described human epigenetic clock. Using publicly available datasets, we find that the mouse clock is accurate enough to measure effects on biological age, including in the context of interventions. While females and males show no significant differences in predicted DNA methylation age, ovariectomy results in significant age acceleration in females. Furthermore, we identify significant differences in age-acceleration dependent on the lipid content of the diet. Here we identify and characterise an epigenetic predictor of age in mice, the mouse epigenetic clock. This clock will be instrumental for understanding the biology of ageing and will allow modulation of its ticking rate and resetting the clock in vivo to study the impact on biological age.

298 citations


Journal ArticleDOI
TL;DR: A Bayesian method to control bias and inflation in EWAS and TWAS based on estimation of the empirical null distribution is proposed and it is demonstrated that the method maximizes power while properly controlling the false positive rate.
Abstract: We show that epigenome- and transcriptome-wide association studies (EWAS and TWAS) are prone to significant inflation and bias of test statistics, an unrecognized phenomenon introducing spurious findings if left unaddressed. Neither GWAS-based methodology nor state-of-the-art confounder adjustment methods completely remove bias and inflation. We propose a Bayesian method to control bias and inflation in EWAS and TWAS based on estimation of the empirical null distribution. Using simulations and real data, we demonstrate that our method maximizes power while properly controlling the false positive rate. We illustrate the utility of our method in large-scale EWAS and TWAS meta-analyses of age and smoking.

228 citations


Journal ArticleDOI
Melissa A. Richard1, Tianxiao Huan, Symen Ligthart2, Rahul Gondalia3, Min A. Jhun4, Jennifer A. Brody5, Marguerite R. Irvin6, Riccardo E. Marioni7, Riccardo E. Marioni8, Jincheng Shen9, Pei-Chien Tsai10, May E. Montasser11, Yucheng Jia12, Catriona Syme13, Elias Salfati14, Eric Boerwinkle1, Eric Boerwinkle15, Weihua Guan16, Thomas H. Mosley17, Jan Bressler1, Alanna C. Morrison1, Chunyu Liu18, Michael M. Mendelson19, André G. Uitterlinden2, Joyce B. J. van Meurs2, Bastiaan T. Heijmans2, Peter A.C. ’t Hoen3, Joyce B. C van Meurs2, A Isaacs3, Rick Jansen3, Lude Franke5, Dorret I. Boomsma, René Pool, Jenny van Dongen4, Jouke J. Hottenga, Marleen M.J. van Greevenbroek, Coen D.A. Stehouwer, Carla J.H. van der Kallen, Casper G. Schalkwijk, Cisca Wijmenga8, Alexandra Zhernakova20, Ettje F. Tigchelaar21, P. Eline Slagboom22, Marian Beekman18, Joris Deelen10, Diana van Heemst11, J. H. Veldink11, Leonard H. van den Berg11, Cornelia M. van Duijn12, Albert Hofman23, P. Mila Jhamai24, Michael M. P. J. Verbiest13, H. Eka D. Suchiman25, Marijn Verkerk11, Ruud van der Breggen12, Jeroen van Rooij13, Nico Lakenberg14, Hailiang Mei5, Maarten van Iterson4, Michiel van Galen26, Jan Bot7, Peter Van ‘t Hof22, Patrick Deelen10, Irene Nooren3, Matthijs Moed2, Martijn Vermaat, Dasha V. Zhernakova1, René Luijk, Marc Jan Bonder, Freerk van Dijk, Wibowo Arindrarto, Szymon M. Kielbasa, Morris A. Swertz, Erik W. van Zwet, Oscar H. Franco2, Guosheng Zhang3, Yun Li3, James D. Stewart3, Joshua C. Bis5, Bruce M. Psaty5, Yii-Der Ida Chen12, Sharon L.R. Kardia4, Wei Zhao4, Stephen Turner27, Devin Absher, Stella Aslibekyan6, John M. Starr7, Allan F. McRae8, Lifang Hou20, Allan C. Just21, Joel Schwartz22, Pantel S. Vokonas18, Cristina Menni10, Tim D. Spector10, Alan R. Shuldiner11, Alan R. Shuldiner28, Coleen M. Damcott11, Jerome I. Rotter12, Walter Palmas23, Yongmei Liu24, Tomáš Paus13, Tomáš Paus29, Steve Horvath25, Jeffrey R. O'Connell11, Xiuqing Guo12, Zdenka Pausova13, Themistocles L. Assimes14, Nona Sotoodehnia5, Jennifer A. Smith4, Donna K. Arnett26, Ian J. Deary7, Andrea A. Baccarelli22, Jordana T. Bell10, Eric A. Whitsel3, Abbas Dehghan2, Abbas Dehghan30, Daniel Levy, Myriam Fornage1 
TL;DR: A two-stage meta-analysis of the cross-sectional associations of systolic and diastolic BP with blood-derived genome-wide DNA methylation measured on the Infinium HumanMethylation450 BeadChip suggests that heritableDNA methylation plays a role in regulating BP independently of previously known genetic variants.
Abstract: Genome-wide association studies have identified hundreds of genetic variants associated with blood pressure (BP), but sequence variation accounts for a small fraction of the phenotypic variance. Epigenetic changes may alter the expression of genes involved in BP regulation and explain part of the missing heritability. We therefore conducted a two-stage meta-analysis of the cross-sectional associations of systolic and diastolic BP with blood-derived genome-wide DNA methylation measured on the Infinium HumanMethylation450 BeadChip in 17,010 individuals of European, African American, and Hispanic ancestry. Of 31 discovery-stage cytosine-phosphate-guanine (CpG) dinucleotides, 13 replicated after Bonferroni correction (discovery: N = 9,828, p 30%) and independent of known BP genetic variants, explaining an additional 1.4% and 2.0% of the interindividual variation in systolic and diastolic BP, respectively. Bidirectional Mendelian randomization among up to 4,513 individuals of European ancestry from 4 cohorts suggested that methylation at cg08035323 (TAF1B-YWHAQ) influences BP, while BP influences methylation at cg00533891 (ZMIZ1), cg00574958 (CPT1A), and cg02711608 (SLC1A5). Gene expression analyses further identified six genes (TSPAN2, SLC7A11, UNC93B1, CPT1A, PTMS, and LPCAT3) with evidence of triangular associations between methylation, gene expression, and BP. Additional integrative Mendelian randomization analyses of gene expression and DNA methylation suggested that the expression of TSPAN2 is a putative mediator of association between DNA methylation at cg23999170 and BP. These findings suggest that heritable DNA methylation plays a role in regulating BP independently of previously known genetic variants.

154 citations


Journal ArticleDOI
TL;DR: It was found that not only PPIs, but also antibiotics, antidepressants, statins and other commonly used medication were associated with distinct gut microbiota signatures, which could affect how the gut microbiota resist enteric infections, promote or ameliorate gut inflammation, or change the host's metabolism.
Abstract: Proton pump inhibitors (PPIs), used to treat gastro-esophageal reflux and prevent gastric ulcers, are among the most widely used drugs in the world. The use of PPIs is associated with an increased risk of enteric infections. Since the gut microbiota can, depending on composition, increase or decrease the risk of enteric infections, we investigated the effect of PPI-use on the gut microbiota. We discovered profound differences in the gut microbiota of PPI users: 20% of their bacterial taxa were statistically significantly altered compared with those of non-users. Moreover, we found that it is not only PPIs, but also antibiotics, antidepressants, statins and other commonly used medication were associated with distinct gut microbiota signatures. As a consequence, commonly used medications could affect how the gut microbiota resist enteric infections, promote or ameliorate gut inflammation, or change the host's metabolism. More studies are clearly needed to understand the role of commonly used medication in altering the gut microbiota as well as the subsequent health consequences.

123 citations


Journal ArticleDOI
R. Karlsson Linner1, R. Karlsson Linner2, Riccardo E. Marioni3, Cornelius A. Rietveld1, Andrew J Simpkin4, Neil M Davies5, Kyoko Watanabe2, Nicola J. Armstrong6, Kirsi Auro7, Kirsi Auro8, Clemens Baumbach, Marc Jan Bonder9, Jadwiga Buchwald8, Giovanni Fiorito10, Khadeeja Ismail8, Stella Iurato11, Anni Joensuu7, Anni Joensuu8, Pauliina Karell8, Silva Kasela12, Jari Lahti8, Allan F. McRae13, Pooja R. Mandaviya14, Ilkka Seppälä15, Yunzhang Wang16, Laura Baglietto17, Elisabeth B. Binder11, Elisabeth B. Binder18, Sarah E. Harris3, Allison M. Hodge19, Allison M. Hodge20, Steve Horvath21, Mikko Hurme15, Magnus Johannesson22, Antti Latvala8, Karen A. Mather23, S. E. Medland24, A. Metspalu12, Lili Milani12, Roger L. Milne20, Roger L. Milne19, Alison Pattie3, Nancy L. Pedersen16, Annette Peters, Silvia Polidoro, Katri Räikkönen8, Gianluca Severi25, Gianluca Severi19, John M. Starr3, Lisette Stolk14, M. Waldenberger, Johan G. Eriksson8, Johan G. Eriksson7, Tõnu Esko26, Tõnu Esko12, Lude Franke9, C Gieger, G.G. Giles20, G.G. Giles19, Sara Hägg16, Pekka Jousilahti7, Jaakko Kaprio8, Mika Kähönen15, Terho Lehtimäki15, Nicholas G. Martin24, J B C van Meurs14, Miina Ollikainen8, Markus Perola7, Markus Perola8, Danielle Posthuma2, Olli T. Raitakari27, Olli T. Raitakari28, Perminder S. Sachdev23, Erdogan Taskesen2, Erdogan Taskesen29, A.G. Uitterlinden14, A.G. Uitterlinden1, Paolo Vineis30, Cisca Wijmenga9, Margaret J. Wright13, Caroline L Relton5, G Davey Smith5, Ian J. Deary3, Philipp Koellinger2, Philipp Koellinger1, Daniel J. Benjamin31 
TL;DR: In this paper, the associations between epigenetic modifications and educational attainment (EA), a biologically distal environmental factor that is arguably among the most important life-shaping experiences for individuals, were investigated.
Abstract: The epigenome is associated with biological factors, such as disease status, and environmental factors, such as smoking, alcohol consumption and body mass index. Although there is a widespread perception that environmental influences on the epigenome are pervasive and profound, there has been little evidence to date in humans with respect to environmental factors that are biologically distal. Here we provide evidence on the associations between epigenetic modifications-in our case, CpG methylation-and educational attainment (EA), a biologically distal environmental factor that is arguably among the most important life-shaping experiences for individuals. Specifically, we report the results of an epigenome-wide association study meta-analysis of EA based on data from 27 cohort studies with a total of 10 767 individuals. We find nine CpG probes significantly associated with EA. However, robustness analyses show that all nine probes have previously been found to be associated with smoking. Only two associations remain when we perform a sensitivity analysis in the subset of never-smokers, and these two probes are known to be strongly associated with maternal smoking during pregnancy, and thus their association with EA could be due to correlation between EA and maternal smoking. Moreover, the effect sizes of the associations with EA are far smaller than the known associations with the biologically proximal environmental factors alcohol consumption, body mass index, smoking and maternal smoking during pregnancy. Follow-up analyses that combine the effects of many probes also point to small methylation associations with EA that are highly correlated with the combined effects of smoking. If our findings regarding EA can be generalized to other biologically distal environmental factors, then they cast doubt on the hypothesis that such factors have large effects on the epigenome.

69 citations


Journal ArticleDOI
TL;DR: This study provides new insights into the dynamic epigenetic landscape of the first 8 years of life by identifying 14,150 consistent age-differential methylation sites (a-DMSs) at epigenome-wide significance of p < 1.14 × 10−7.
Abstract: DNA methylation has been found to associate with disease, aging and environmental exposure, but it is unknown how genome, environment and disease influence DNA methylation dynamics in childhood. By analysing 538 paired DNA blood samples from children at birth and at 4–5 years old and 726 paired samples from children at 4 and 8 years old from four European birth cohorts using the Illumina Infinium Human Methylation 450 k chip, we have identified 14,150 consistent age-differential methylation sites (a-DMSs) at epigenome-wide significance of p < 1.14 × 10−7. Genes with an increase in age-differential methylation were enriched in pathways related to ‘development’, and were more often located in bivalent transcription start site (TSS) regions, which can silence or activate expression of developmental genes. Genes with a decrease in age-differential methylation were involved in cell signalling, and enriched on H3K27ac, which can predict developmental state. Maternal smoking tended to decrease methylation levels at the identified da-DMSs. We also found 101 a-DMSs (0.71%) that were regulated by genetic variants using cis-differential Methylation Quantitative Trait Locus (cis-dMeQTL) mapping. Moreover, a-DMS-associated genes during early development were significantly more likely to be linked with disease. Our study provides new insights into the dynamic epigenetic landscape of the first 8 years of life.

55 citations


Journal ArticleDOI
01 Apr 2017-Gut
TL;DR: A genome-wide association study (GWAS) in two well-characterised population-based cohorts with genotype and defaecation data available and the average number of bowel movements per day did not differ between cohorts.
Abstract: Stool consistency and frequency patterns are complex traits that are often altered in GI disease, and recent studies published in Gut highlight the importance of stool frequency in relation to gut microbiota composition and the efficacy of pharmacological and dietary treatments in IBS.1–3 Despite reported heritability in invertebrates4 and similar evidence from open-field defaecation models in rats,5 the genetics of stool frequency has not been explored in humans. We undertook a genome-wide association study (GWAS) in two well-characterised population-based cohorts with genotype and defaecation data available: LifeLines-Deep (LLD) from the Netherlands (N=1546; 58% females; mean age 44 years (range 18–86)) and PopCol (PC) from Sweden (N=284; 60% females; mean age 54 years (range 22–71)).6 ,7 The average number of bowel movements per day (BM/d) was extracted from daily records kept by both populations and did not differ between cohorts (LLD=1.39±0.64SD; PC=1.42±0.74SD). Available CytoChip+Immunochip (LLD) and HumanOmniExpressExome (PC) Illumina single-nucleotide polymorphism (SNP) genotype data were imputed using IMPUTE2 (https://mathgen.stats.ox.ac.uk/impute/impute_v2.html) with the Genome of the Netherlands (http://www.nlgenome.nl/) as reference. SNPs were filtered on minor allele frequency >0.05 and Hardy–Weinberg equilibrium p>1E-04, samples were filtered on infoscore ≥0.8 and population outliers …

14 citations


Journal ArticleDOI
30 Oct 2017-PLOS ONE
TL;DR: The results do not show widespread changes in DNA-methylation across the genome, and therefore do not support the hypothesis that mildly elevated homocysteine is associated with widespread methylation changes in leukocytes.
Abstract: Background: DNA methylation is affected by the activities of the key enzymes and intermediate metabolites of the one-carbon pathway, one of which involves homocysteine. We investigated the effect of the well-known genetic variant associated with mildly elevated homocysteine: MTHFR 677C>T independently and in combination with other homocysteine-associated variants, on genome-wide leukocyte DNA-methylation. Methods: Methylation levels were assessed using Illumina 450k arrays on 9,894 individuals of European ancestry from 12 cohort studies. Linear-mixed-models were used to study the association of additive MTHFR 677C>T and genetic-risk score (GRS) based on 18 homocysteine-associated SNPs, with genome-wide methylation. Results: Meta-analysis revealed that the MTHFR 677C>T variant was associated with 35 CpG sites in cis, and the GRS showed association with 113 CpG sites near the homocysteine-associated variants. Genome-wide analysis revealed that the MTHFR 677C>T variant was associated with 1 trans-CpG (nearest gene ZNF184), while the GRS model showed association with 5 significant trans-CpGs annotated to nearest genes PTF1A, MRPL55, CTDSP2, CRYM and FKBP5. Conclusions: Our results do not show widespread changes in DNA-methylation across the genome, and therefore do not support the hypothesis that mildly elevated homocysteine is associated with widespread methylation changes in leukocytes.

Posted ContentDOI
22 Mar 2017-bioRxiv
TL;DR: This model, which estimates age based on DNA methylation at 329 unique CpG sites, has a median absolute error of 3.33 weeks, and has similar properties to the recently described human epigenetic clock, and is accurate enough to measure effects on biological age, including in the context of interventions.
Abstract: Background: DNA-methylation changes at a discrete set of sites in the human genome are predictive of chronological and biological age. However, it is not known whether these changes are causative or a consequence of an underlying ageing process. It has also not been shown whether this epigenetic clock is unique to humans or conserved in the more experimentally tractable mouse. Results: We have generated a comprehensive set of genome-scale base-resolution methylation maps from multiple mouse tissues spanning a wide range of ages. Many CpG sites show significant tissue-independent correlations with age and allowed us to develop a multi-tissue predictor of age in the mouse. Our model, which estimates age based on DNA methylation at 329 unique CpG sites, has a median absolute error of 3.33 weeks, and has similar properties to the recently described human epigenetic clock. Using publicly available datasets, we find that the mouse clock is accurate enough to measure effects on biological age, including in the context of interventions. While females and males show no significant differences in predicted DNA methylation age, ovariectomy results in significant age acceleration in females. Furthermore, we identify significant differences in age-acceleration dependent on the lipid content of the offspring diet. Conclusions: Here we identify and characterize an epigenetic predictor of age in mice, the mouse epigenetic clock. This clock will be instrumental for understanding the biology of ageing and will allow modulation of its ticking rate and resetting the clock in vivo to study the impact on biological age.

01 Jan 2017
TL;DR: Two biological omics are focused on, the gut microbiome composition and the DNA-methylome, which are the collection of micro-organisms that live together in the human gut and the occurrence of a methyl group bound to the DNA and this mainly occurs at cysteine-guanine pairs.
Abstract: There are many factors involved in the development of human diseases and traits. In recent years the field of human genetics has been very successful in linking genetic variation to diseases and traits. By conducting large-scale studies comparing the genetic make-up of affected versus non-affected participants, we have identified thousands of variants in the human genome that are more or less commonly found in cases compared to controls. These genome-wide association studies (GWAS) have been instrumental in the identification of genes linked to a multitude of diseases and traits. Variants in the functional parts of a gene can be relatively straightforward to interpret. However, not all the variants linked to disease can be directly interpreted. By using intermediate molecular data layers, such as gene expression, DNA-methylation or protein levels, we can gain more insight into the genetic variants identified by GWAS. However, we only get a limited picture of disease by focusing on genetic variation. Another important factor related to disease is the environment. But it is much harder to quantify environmental factors than to determine the genetic differences between two individuals. Using the intermediate molecular data, or biological omics, we can gain insights into the environment of individuals. The environment surrounding individuals can, for instance, influence the composition of their microbiome, but also their gene expression, DNA-methylation and protein levels. By studying the differences in these biological omics in relation to phenotypes and disease, we can learn more about the environmental factors that lead to disease. However, as with GWAS studies, we do not always know what the differences in these biological data layers mean. In this thesis we have focused on two biological omics, the gut microbiome composition and the DNA-methylome. The gut microbiome is the collection of micro-organisms that live together in the human gut; DNA-methylation is the occurrence of a methyl group bound to the DNA and this mainly occurs at cysteine-guanine pairs. In the first part of the thesis we have focused on inter-individual differences influencing, or influenced by, differences in the microbiome composition, while in the second part, we have focused on changes in DNA-methylation associated to tissue differences and on the influence of genetic variation on DNA-methylation.

Posted ContentDOI
07 Mar 2017-bioRxiv
TL;DR: Evidence is provided on the associations between epigenetic modifications—in this case, CpG methylation—and educational attainment (EA), a biologically distal environmental factor that is arguably among of the most important life-shaping experiences for individuals.
Abstract: The epigenome has been shown to be influenced by biological factors, such as disease status, and environmental factors, such as smoking, alcohol consumption, and body mass index. Although there is a widespread perception that environmental influences on the epigenome are pervasive and profound, there has been little evidence to date in humans with respect to environmental factors that are biologically distal. Here, we provide evidence on the associations between epigenetic modifications, in our case, CpG methylation and educational attainment (EA), a biologically distal environmental factor that is arguably among of the most important life-shaping experiences for individuals. Specifically, we report the results of an epigenome-wide association study meta-analysis of EA based on data from 27 cohort studies with a total of 10,767 individuals. While we find that 9 CpG probes are significantly associated with EA, only two remain associated when we restrict the sample to never-smokers. These two are known to be strongly associated with maternal smoking during pregnancy, and thus their association with EA could be due to correlation between EA and maternal smoking. Moreover, their effect sizes on EA are far smaller than the known associations between CpG probes and biologically proximal environmental factors. Two analyses that combine the effects of many probes, polygenic methylation score and epigenetic-clock analyses, both suggest small associations with EA. If our findings regarding EA can be generalized to other biologically distal environmental factors, then they cast doubt on the hypothesis that such factors have large effects on the epigenome.