1
|
Shin J, Bressler J, Grove ML, Brown M, Selvin E, Pankow JS, Fornage M, Morrison AC, Sarnowski C. DNA methylation markers of insulin resistance surrogate measures in the Atherosclerosis Risk in Communities (ARIC) study. Epigenetics 2025; 20:2498857. [PMID: 40327844 PMCID: PMC12064056 DOI: 10.1080/15592294.2025.2498857] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2024] [Revised: 04/03/2025] [Accepted: 04/21/2025] [Indexed: 05/08/2025] Open
Abstract
Insulin resistance (IR) is a risk factor for cardiovascular diseases and type 2 diabetes. Associations between DNA methylation (DNAm) and IR have been less studied in African ancestry (AA) populations than those of European ancestry (EA). We aimed to identify associations between whole blood DNAm and IR in up to 1,811 AA and 964 EA participants from the Atherosclerosis Risk in Communities (ARIC) study. We quantified IR using three surrogate measures: the homeostasis model assessment of insulin resistance (HOMA-IR), the triglyceride-glucose index (TyG), and the triglyceride glucose-body mass index (TyG-BMI). We used ancestry-stratified linear regression models to conduct epigenome-wide association studies of IR, adjusting for batch effects and relevant covariates. Among 484,436 tested CpG sites, 39 were significantly associated with IR, of which 31% (10 in AA and two in EA) were associated with TyG-BMI and not previously reported for IR or related traits. These include a positive association at cg18335991-SEMA7A in AA. SEMA7A inhibits adipogenesis of preadipocytes and lipogenesis of mature adipocytes. DNAm levels at cg18335991 have been reported to be negatively associated with SEMA7A expression in blood. After additionally adjusting for smoking and drinking status, 15 of the 39 significant CpG sites remained significant or suggestive. Our study identified novel IR-associated CpG sites, contributing to a broader understanding of the epigenetic mechanisms underlying IR in diverse populations.
Collapse
Affiliation(s)
- Jeewoen Shin
- Human Genetics Center, Department of Epidemiology, University of Texas Health Science Center at Houston, Houston, School of Public Health, TX, USA
| | - Jan Bressler
- Human Genetics Center, Department of Epidemiology, University of Texas Health Science Center at Houston, Houston, School of Public Health, TX, USA
| | - Megan L. Grove
- Human Genetics Center, Department of Epidemiology, University of Texas Health Science Center at Houston, Houston, School of Public Health, TX, USA
| | - Michael Brown
- Human Genetics Center, Department of Epidemiology, University of Texas Health Science Center at Houston, Houston, School of Public Health, TX, USA
| | - Elizabeth Selvin
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - James S. Pankow
- Division of Epidemiology and Community Health, University of Minnesota School of Public Health, Minneapolis, MN, USA
| | - Myriam Fornage
- Human Genetics Center, Department of Epidemiology, University of Texas Health Science Center at Houston, Houston, School of Public Health, TX, USA
- Brown Foundation Institute of Molecular Medicine, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Alanna C. Morrison
- Human Genetics Center, Department of Epidemiology, University of Texas Health Science Center at Houston, Houston, School of Public Health, TX, USA
| | - Chloé Sarnowski
- Human Genetics Center, Department of Epidemiology, University of Texas Health Science Center at Houston, Houston, School of Public Health, TX, USA
| |
Collapse
|
2
|
He F, Zhang H, Zheng G, Zhou B, Fang Z, Zhu H, Dong Y, Hao G. Adverse childhood experiences and cardiometabolic biomarkers: A causal analysis. J Affect Disord 2025; 381:418-426. [PMID: 40194629 DOI: 10.1016/j.jad.2025.04.039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/08/2024] [Revised: 04/03/2025] [Accepted: 04/04/2025] [Indexed: 04/09/2025]
Abstract
BACKGROUND The associations between adverse childhood experiences (ACEs) and cardiometabolic biomarkers need to be further studied. Our objective was to investigate whether ACEs are causally associated with cardiometabolic biomarkers using observational study and two-sample Mendelian randomization (MR) analysis. METHODS The China Health and Retirement Longitudinal Study (CHARLS) data from 2014 to 2015 was used in the observational study. ACEs were divided into 4 groups (0, 1, 2, and 3 or more) according to whether they had experienced 12 items of negative experiences in childhood. A multilevel model was used to estimate the association between ACEs and each cardiometabolic biomarker. Further, we used two-sample MR to identify their potential causality. RESULTS A total of 11,422 participants (age:45-96) were eligible for the analyses in the observational study. Participants who experienced more ACEs were significantly higher in high-sensitivity C-reactive protein (hs-CRP) and high-density lipoprotein cholesterol (HDL-C) (all P < 0.05). Compared with those without ACE exposure, participants who experienced 3 or more ACEs had significantly lower total cholesterol (P < 0.05). In addition, a stronger association between ACEs and hs-CRP in males, as well as a stronger association between ACEs and HDL-C (Pinteraction = 0.036) in participants with higher education levels were observed. Consistently, in two-sample MR, we observed causal associations between DNA methylation loci and those cardiometabolic biomarkers. CONCLUSION Our results indicate that ACEs were causally associated with several cardiometabolic biomarkers. Further, adversity-associated DNA methylation loci might reflect buffering mechanisms against childhood adversity, which provides novel insight to the cardiovascular risk interventions.
Collapse
Affiliation(s)
- Fudong He
- School of Public Health, Guangdong Pharmaceutical University, Guangzhou, China
| | - Haofeng Zhang
- School of Public Health, Guangdong Pharmaceutical University, Guangzhou, China
| | - Guangjun Zheng
- Department of Public Health and Preventive Medicine, School of Medicine, Jinan University, Guangzhou, China
| | - Biying Zhou
- Department of Public Health and Preventive Medicine, School of Medicine, Jinan University, Guangzhou, China
| | - Zhenger Fang
- Department of Public Health and Preventive Medicine, School of Medicine, Jinan University, Guangzhou, China
| | - Haidong Zhu
- Georgia Prevention Institute, Department of Medicine, Medical College of Georgia, Augusta University, Augusta, GA, USA
| | - Yanbin Dong
- Georgia Prevention Institute, Department of Medicine, Medical College of Georgia, Augusta University, Augusta, GA, USA
| | - Guang Hao
- School of Public Health, Guangdong Pharmaceutical University, Guangzhou, China.
| |
Collapse
|
3
|
Zhang Y, Du X, Yang Y, Ren Y, Zhou L, Hua J, Wang H. A multi-omics and mediation-based genetic screening approach identifies STX4 as a key link between epigenetic regulation, immune cells, and childhood asthma. Clin Epigenetics 2025; 17:101. [PMID: 40514687 DOI: 10.1186/s13148-025-01908-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2025] [Accepted: 05/23/2025] [Indexed: 06/16/2025] Open
Abstract
BACKGROUND Childhood asthma presents a multifaceted immune-driven pathology shaped by genetic, epigenetic, and immune regulatory interactions. Despite extensive genome-wide analyses pinpointing multiple susceptibility loci, the precise functional contributors to asthma pathogenesis remain elusive. This study employs a comprehensive multi-omics framework and Mendelian randomization (MR) analysis to systematically identify and validate key genetic determinants implicated in childhood asthma. METHODS A genome-wide screening of over 19,000 human genes was performed to identify cis-eQTL-regulated genes associated with childhood asthma. Two-sample MR was conducted to assess causality, followed by Summary-based Mendelian Randomization (SMR) to validate findings in independent datasets. Colocalization analysis determined whether gene expression and asthma GWAS signals share a common causal variant. Protein quantitative trait loci (pQTL) analysis further validated gene associations at the protein level. DNA methylation quantitative trait loci (mQTL) MR and mediation analysis explored epigenetic regulatory mechanisms, while linkage disequilibrium score regression (LDSC) quantified genome-wide genetic correlations. Immune cell mediation analysis examined potential immune-driven effects, and Phenome-Wide Association Study (PheWAS) evaluated pleiotropy and therapeutic safety. RESULTS Following systematic screening, STX4 emerged as a strong candidate gene for childhood asthma. MR and SMR analyses confirmed its causal role, while colocalization analysis provided robust genetic evidence supporting STX4's regulatory influence on childhood asthma susceptibility. pQTL validation confirmed that STX4's effects extend to the protein level, strengthening its biological relevance. DNA methylation analysis revealed key CpG (Cytosine-phosphate-Guanine) sites regulating STX4 expression, with higher methylation levels reducing childhood asthma risk. Immune cell mediation analysis demonstrated that STX4 influences childhood asthma risk via CD4+ and CD8+ T cell subsets. LDSC analysis reinforced a significant genetic correlation between STX4 and childhood asthma, while PheWAS detected no major pleiotropy, suggesting that STX4 is a specific and promising therapeutic target. CONCLUSIONS This study systematically identifies and validates STX4 as a key genetic regulator in childhood asthma by integrating large-scale genetic, epigenetic, and immune regulatory data. These findings provide strong evidence for STX4's role in childhood asthma pathogenesis, highlighting STX4 as a potential target for future precision therapies in childhood asthma.
Collapse
Affiliation(s)
- Yuan Zhang
- Laboratory of Pediatric Research, Children's Hospital of Wujiang District, Suzhou, 215234, Jiangsu, China
- Department of Clinical Laboratory, Children's Hospital of Wujiang District, Suzhou, 215234, Jiangsu, China
| | - Xiaochen Du
- Department of Emergency and Intensive Care Unit, Children's Hospital of Soochow University, Suzhou, 215025, Jiangsu, China
- Department of Pediatric, Children's Hospital of Wujiang District, Suzhou, 215234, Jiangsu, China
| | - Yujuan Yang
- Department of Clinical Laboratory, Children's Hospital of Wujiang District, Suzhou, 215234, Jiangsu, China
| | - Yaqiong Ren
- Laboratory of Pediatric Research, Children's Hospital of Wujiang District, Suzhou, 215234, Jiangsu, China
- Department of Clinical Laboratory, Children's Hospital of Wujiang District, Suzhou, 215234, Jiangsu, China
| | - Lijun Zhou
- Laboratory of Pediatric Research, Children's Hospital of Wujiang District, Suzhou, 215234, Jiangsu, China
- Department of Clinical Laboratory, Children's Hospital of Wujiang District, Suzhou, 215234, Jiangsu, China
| | - Jun Hua
- Laboratory of Pediatric Research, Children's Hospital of Wujiang District, Suzhou, 215234, Jiangsu, China.
- Department of Respiratory Medicine, Children's Hospital of Soochow University, Suzhou, 215025, Jiangsu, China.
| | - Hongying Wang
- Laboratory of Pediatric Research, Children's Hospital of Wujiang District, Suzhou, 215234, Jiangsu, China.
- Department of Clinical Laboratory, Children's Hospital of Wujiang District, Suzhou, 215234, Jiangsu, China.
- Institute of Pediatric Research, Children's Hospital of Soochow University, Suzhou, 215025, Jiangsu, China.
| |
Collapse
|
4
|
Jeon Y, Kwon Y, Kim YJ, Jeon S, Ryu H, An K, Kim BC, Kim W, Lee SY, Bae JW, Hwang JY, Kang MG, Kang Y, Bhak J, Shin ES. Epigenetic Modulation of Vascular Smooth Muscle Cell Phenotype Switching in Early-Onset Acute Myocardial Infarction. Arterioscler Thromb Vasc Biol 2025; 45:e217-e230. [PMID: 40242870 DOI: 10.1161/atvbaha.125.322503] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2025] [Accepted: 03/27/2025] [Indexed: 04/18/2025]
Abstract
BACKGROUND The epigenetic mechanisms underlying early-onset acute myocardial infarction (AMI) remain insufficiently characterized. The present study aims to elucidate the pathophysiology of early-onset AMI by investigating its epigenetic features as molecular indicators. METHODS A comparative differential methylation analysis was performed on whole blood samples from 298 patients with early-onset AMI with clinical follow-up and 247 controls using targeted bisulfite sequencing. Clusters of differentially methylated sites (CDMSs) were defined to highlight regions of concentrated methylation changes in patients with early-onset AMI. Cox proportional hazards regression was conducted to evaluate the prognostic significance of the methylation biomarkers. RESULTS A total of 692 differentially methylated sites (DMSs) were identified as biomarkers associated with early-onset AMI. Among these, 396 DMSs were grouped into 147 CDMSs. Notably, the UHRF1 and STIMATE genes, which regulate synthetic and osteoblast-like vascular smooth muscle cell phenotypes, respectively, contained CDMSs with the highest number of significant DMSs. UHRF1 demonstrated a CDMS with 10 significant DMSs within a 117-bp region, while STIMATE included a 264-bp CDMS with 10 significant DMSs. Both regions also exhibited consistent methylation patterns in coronary tissues, comparing human coronary plaque to normal coronary artery samples. Additionally, the HIPK3 gene, which modulates STAT3 (signal transducer and activator of transcription 3) expression, thereby promoting osteoblast-like transformation in vascular smooth muscle cells, showed a CDMS with 5 significant DMSs within a 123-bp region, with further validation in the corresponding tissues. Furthermore, over 66% biomarkers demonstrated significant associations with mortality in patients with early-onset AMI, providing evidence of the impact of these biomarkers on the pathophysiology of the disease. CONCLUSIONS This innovative epigenomic study into early-onset AMI not only identifies biomarkers associated with the disease and its mortality but also highlights the critical role of vascular smooth muscle cell phenotype regulation in early-onset AMI pathogenesis. Our findings suggest that changes in vascular smooth muscle cell phenotypes toward synthetic and osteoblast-like states play a crucial role in early-onset AMI.
Collapse
Affiliation(s)
- Yeonsu Jeon
- Clinomics Inc., Osong, Republic of Korea (Y.J., Y.J.K., S.J., H.R., B.-C.K., Y. Kang, J.B.)
| | - Yoonsung Kwon
- Korean Genomics Center (KOGIC), Ulsan National Institute of Science and Technology (UNIST), Ulsan, Republic of Korea (Y. Kwon, K.A., J.B.)
- Department of Biomedical Engineering, College of Information-Bio Convergence Engineering, Ulsan National Institute of Science and Technology (UNIST), Ulsan, Republic of Korea (Y. Kwon, K.A., J.B.)
| | - Yeo Jin Kim
- Clinomics Inc., Osong, Republic of Korea (Y.J., Y.J.K., S.J., H.R., B.-C.K., Y. Kang, J.B.)
| | - Sungwon Jeon
- Clinomics Inc., Osong, Republic of Korea (Y.J., Y.J.K., S.J., H.R., B.-C.K., Y. Kang, J.B.)
| | - Hyojung Ryu
- Clinomics Inc., Osong, Republic of Korea (Y.J., Y.J.K., S.J., H.R., B.-C.K., Y. Kang, J.B.)
| | - Kyungwhan An
- Korean Genomics Center (KOGIC), Ulsan National Institute of Science and Technology (UNIST), Ulsan, Republic of Korea (Y. Kwon, K.A., J.B.)
- Department of Biomedical Engineering, College of Information-Bio Convergence Engineering, Ulsan National Institute of Science and Technology (UNIST), Ulsan, Republic of Korea (Y. Kwon, K.A., J.B.)
| | - Byoung-Chul Kim
- Clinomics Inc., Osong, Republic of Korea (Y.J., Y.J.K., S.J., H.R., B.-C.K., Y. Kang, J.B.)
| | - Weon Kim
- Division of Cardiology, Department of Internal Medicine, Kyung Hee University Hospital, Kyung Hee University, Seoul, Republic of Korea (W.K.)
| | - Sang Yeub Lee
- Division of Cardiology, Department of Internal Medicine, Chung-Ang University College of Medicine, Chung-Ang University Gwangmyeong Hospital, Republic of Korea (S.Y.L.)
| | - Jang-Whan Bae
- Department of Internal Medicine, Chungbuk National University Hospital, College of Medicine, Chungbuk National University, Cheongju, Republic of Korea (J.-W.B.)
| | - Jin-Yong Hwang
- Department of Internal Medicine, Gyeongsang National University School of Medicine and Gyeongsang National University Hospital, Jinju, Republic of Korea (J.-Y.H., M.G.K.)
| | - Min Gyu Kang
- Department of Internal Medicine, Gyeongsang National University School of Medicine and Gyeongsang National University Hospital, Jinju, Republic of Korea (J.-Y.H., M.G.K.)
| | - Younghui Kang
- Clinomics Inc., Osong, Republic of Korea (Y.J., Y.J.K., S.J., H.R., B.-C.K., Y. Kang, J.B.)
| | - Jong Bhak
- Clinomics Inc., Osong, Republic of Korea (Y.J., Y.J.K., S.J., H.R., B.-C.K., Y. Kang, J.B.)
- Korean Genomics Center (KOGIC), Ulsan National Institute of Science and Technology (UNIST), Ulsan, Republic of Korea (Y. Kwon, K.A., J.B.)
- Department of Biomedical Engineering, College of Information-Bio Convergence Engineering, Ulsan National Institute of Science and Technology (UNIST), Ulsan, Republic of Korea (Y. Kwon, K.A., J.B.)
- Personal Genomics Institute, Genome Research Foundation, Osong, Republic of Korea (J.B.)
| | - Eun-Seok Shin
- Department of Cardiology, Ulsan University Hospital, University of Ulsan College of Medicine, Ulsan, Republic of Korea (E.-S.S.)
| |
Collapse
|
5
|
Aguilar-Lacasaña S, Cosin-Tomas M, Raimbault B, Gómez-Herrera L, Sánchez O, Zanini MJ, Capdevila RP, Foraster M, Gascon M, Rivas I, Llurba E, Gómez-Roig MD, Sunyer J, Bustamante M, Vrijheid M, Dadvand P. Epigenome-wide association study of pregnancy exposure to green space and placental DNA methylation. ENVIRONMENTAL RESEARCH 2025; 274:121286. [PMID: 40043929 DOI: 10.1016/j.envres.2025.121286] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/21/2024] [Revised: 02/28/2025] [Accepted: 03/02/2025] [Indexed: 05/04/2025]
Abstract
Green space exposure during pregnancy has been associated with lower risk of adverse birth outcomes, but the biological mechanisms remain unclear. Epigenetic changes, such as DNA methylation (DNAm), may contribute to this association. The placenta, crucial for foetal development, has been understudied in relation to prenatal green space exposure and DNAm on a genome-wide scale. Here, we aimed to investigate the association between green space exposure during pregnancy and epigenome-wide placental DNAm in 550 mother-child pairs from the Barcelona Life Study Cohort (BiSC) in Spain. Green space exposure was assessed as (i) residential surrounding greenness (satellite-based Normalized Difference Vegetation Index (NDVI) in buffers of 100 m, 300 m and 500 m), (ii) residential distance to the nearest major green space (meters), (iii) use of green space (hours/week), and (iv) visual access to greenery through the home window (≥half of the view). Placental DNAm was measured with the EPIC array. Differentially methylated positions (DMPs) were identified using robust linear regression models adjusted for covariates, while differentially methylated regions (DMRs) were identified using the dmrff method. After Bonferroni correction, cg14852540, annotated to SLC25A10 gene, showed an inverse association with residential greenness within 500 m buffer. Additionally, 101 DMPs were suggestively significant (p-values <1 × 10-5) and annotated to genes involved in glucocorticoid-related pathways, inflammatory response, oxidative stress response, and oocyte maturation. No DMRs were identified. Overall, we identified an association between residential greenness and DNAm levels at one CpG in the SLC25A10 gene. Larger studies are needed to validate these findings and understand the biological pathways.
Collapse
Affiliation(s)
- Sofía Aguilar-Lacasaña
- ISGlobal, Barcelona, Spain; Universitat de Barcelona (UB), Barcelona, Spain; Universitat Pompeu Fabra (UPF), Barcelona, Spain; CIBER Epidemiología y Salud Pública, Instituto de Salud Carlos III, Spain.
| | - Marta Cosin-Tomas
- ISGlobal, Barcelona, Spain; Universitat Pompeu Fabra (UPF), Barcelona, Spain; CIBER Epidemiología y Salud Pública, Instituto de Salud Carlos III, Spain
| | - Bruno Raimbault
- ISGlobal, Barcelona, Spain; Universitat Pompeu Fabra (UPF), Barcelona, Spain; CIBER Epidemiología y Salud Pública, Instituto de Salud Carlos III, Spain
| | - Laura Gómez-Herrera
- ISGlobal, Barcelona, Spain; Universitat de Barcelona (UB), Barcelona, Spain; Universitat Pompeu Fabra (UPF), Barcelona, Spain; CIBER Epidemiología y Salud Pública, Instituto de Salud Carlos III, Spain
| | - Olga Sánchez
- Primary Care Interventions to Prevent Maternal and Child Chronic Diseases of Perinatal and Developmental Origin Network (RICORS-SAMID) (RD21/0012/0001), Spain; Department of Obstetrics and Gynaecology. Hospital de la Santa Creu i Sant Pau, Institut de Recerca (IR SANT PAU), Barcelona, 08041, Spain
| | - Maria Julia Zanini
- Department of Obstetrics and Gynaecology. Hospital de la Santa Creu i Sant Pau, Institut de Recerca (IR SANT PAU), Barcelona, 08041, Spain
| | - Rosalia Pascal Capdevila
- Primary Care Interventions to Prevent Maternal and Child Chronic Diseases of Perinatal and Developmental Origin Network (RICORS-SAMID) (RD21/0012/0003), Spain; BCNatal. Barcelona Center for Maternal Foetal and Neonatal Medicine (Hospital Sant Joan de Déu and Hospital Clínic), University of Barcelona, Barcelona, Spain
| | - Maria Foraster
- PHAGEX Research Group, Blanquerna School of Health Science, Universitat Ramon Llull (URL), Barcelona, Spain
| | - Mireia Gascon
- ISGlobal, Barcelona, Spain; Universitat Pompeu Fabra (UPF), Barcelona, Spain; CIBER Epidemiología y Salud Pública, Instituto de Salud Carlos III, Spain; Unitat de Suport a la Recerca de la Catalunya Central, Fundació Institut Universitari per a la Recerca a l'Atenció Primària de Salut Jordi Gol i Gurina (IDIAPJGol), Manresa, Spain
| | - Ioar Rivas
- ISGlobal, Barcelona, Spain; Universitat Pompeu Fabra (UPF), Barcelona, Spain; CIBER Epidemiología y Salud Pública, Instituto de Salud Carlos III, Spain
| | - Elisa Llurba
- Primary Care Interventions to Prevent Maternal and Child Chronic Diseases of Perinatal and Developmental Origin Network (RICORS-SAMID) (RD21/0012/0001), Spain; Department of Obstetrics and Gynaecology. Hospital de la Santa Creu i Sant Pau, Institut de Recerca (IR SANT PAU), Barcelona, 08041, Spain
| | - Maria Dolores Gómez-Roig
- Primary Care Interventions to Prevent Maternal and Child Chronic Diseases of Perinatal and Developmental Origin Network (RICORS-SAMID) (RD21/0012/0003), Spain; BCNatal. Barcelona Center for Maternal Foetal and Neonatal Medicine (Hospital Sant Joan de Déu and Hospital Clínic), University of Barcelona, Barcelona, Spain; Institut de Recerca Sant Joan de Déu, Esplugues de Llobregat, Barcelona, Spain
| | - Jordi Sunyer
- ISGlobal, Barcelona, Spain; Universitat Pompeu Fabra (UPF), Barcelona, Spain; CIBER Epidemiología y Salud Pública, Instituto de Salud Carlos III, Spain
| | - Mariona Bustamante
- ISGlobal, Barcelona, Spain; Universitat Pompeu Fabra (UPF), Barcelona, Spain; CIBER Epidemiología y Salud Pública, Instituto de Salud Carlos III, Spain.
| | - Martine Vrijheid
- ISGlobal, Barcelona, Spain; Universitat Pompeu Fabra (UPF), Barcelona, Spain; CIBER Epidemiología y Salud Pública, Instituto de Salud Carlos III, Spain
| | - Payam Dadvand
- ISGlobal, Barcelona, Spain; Universitat Pompeu Fabra (UPF), Barcelona, Spain; CIBER Epidemiología y Salud Pública, Instituto de Salud Carlos III, Spain
| |
Collapse
|
6
|
Guo X, Sulaiman M, Neumann A, Zheng SC, Cecil CAM, Teschendorff AE, Heijmans BT. Unified high-resolution immune cell fraction estimation in blood tissue from birth to old age. Genome Med 2025; 17:63. [PMID: 40426256 PMCID: PMC12108007 DOI: 10.1186/s13073-025-01489-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2025] [Accepted: 05/16/2025] [Indexed: 05/29/2025] Open
Abstract
Variations in immune-cell fractions can confound or hamper interpretation of DNAm-based biomarkers in blood. Although cell-type deconvolution can address this challenge for cord and adult blood, currently there is no method applicable to blood from other age groups, including infants and children. Here we construct and extensively validate a DNAm reference panel, called UniLIFE, for 19 immune cell-types, applicable to blood tissue of any age. We use UniLIFE to delineate the dynamics of immune-cell fractions from birth to old age, and to infer disease associated immune cell fraction variations in newborns, infants, children and adults. In a prospective longitudinal study of type-1 diabetes in infants and children, UniLIFE identifies differentially methylated positions that precede type-1 diabetes diagnosis and that map to diabetes related signaling pathways. In summary, UniLIFE will improve the identification and interpretation of blood-based DNAm biomarkers for any age group, but specially for longitudinal studies that include infants and children. The UniLIFE panel and algorithms to estimate cell-type fractions are available from our EpiDISH Bioconductor R-package: https://bioconductor.org/packages/release/bioc/html/EpiDISH.html.
Collapse
Affiliation(s)
- Xiaolong Guo
- Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, 320 Yue Yang Road, Shanghai, 200031, China
| | - Mahnoor Sulaiman
- Department of Biomedical Data Sciences, Leiden University Medical Center, Einthovenweg 20, Leiden, 2333 ZC, The Netherlands
- Department of Child and Adolescent Psychiatry/Psychology, Sophia's Children Centre, Erasmus MC, Rotterdam, The Netherlands
| | - Alexander Neumann
- Department of Child and Adolescent Psychiatry/Psychology, Sophia's Children Centre, Erasmus MC, Rotterdam, The Netherlands
| | - Shijie C Zheng
- Pfizer Research & Development, Pfizer Inc, Groton, CT, USA
| | - Charlotte A M Cecil
- Department of Child and Adolescent Psychiatry/Psychology, Sophia's Children Centre, Erasmus MC, Rotterdam, The Netherlands.
- Department of Epidemiology, Erasmus MC, Rotterdam, The Netherlands.
| | - Andrew E Teschendorff
- Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, 320 Yue Yang Road, Shanghai, 200031, China.
| | - Bastiaan T Heijmans
- Department of Biomedical Data Sciences, Leiden University Medical Center, Einthovenweg 20, Leiden, 2333 ZC, The Netherlands.
| |
Collapse
|
7
|
Kitaba NT, Østergaard TM, Lønnebotn M, Accordini S, Real FG, Malinovschi A, Oudin A, Benediktsdottir B, González FJC, Gómez LP, Holm M, Jõgi NO, Dharmage SC, Skulstad SM, Schlünssen V, Svanes C, Holloway JW. Father's adolescent body silhouette is associated with offspring asthma, lung function and BMI through DNA methylation. Commun Biol 2025; 8:796. [PMID: 40410506 PMCID: PMC12102279 DOI: 10.1038/s42003-025-08121-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2024] [Accepted: 04/23/2025] [Indexed: 05/25/2025] Open
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.
Collapse
Grants
- 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 No. 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 No. 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 (PhD scholarship).
Collapse
Affiliation(s)
- Negusse Tadesse Kitaba
- Human Development and Health, Faculty of Medicine, University of Southampton, Southampton, UK
| | - Toril Mørkve Østergaard
- Department of Global Public Health and Primary Care, Centre for International Health, University of Bergen, Bergen, Norway
| | - Marianne Lønnebotn
- Department of Health and Caring Sciences, Western Norway University of Applied Sciences, Bergen, Norway
| | - Simone Accordini
- Unit of Epidemiology and Medical Statistics, Department of Diagnostics and Public Health, University of Verona, Verona, Italy
| | | | - Andrei Malinovschi
- Department of Medical Sciences: Clinical Physiology, Uppsala University, Uppsala, Sweden
| | - Anna Oudin
- Section of Sustainable Health, Department of Public Health and Clinical Medicine, Umeå University, Umeå, Sweden
| | - Bryndis Benediktsdottir
- Department of Allergy, Respiratory Medicine and Sleep, Landspitali University Hospital, Reykjavik, Iceland Faculty of Medicine, University of Iceland, Landspitali, Iceland
| | | | | | - Mathias Holm
- Occupational and Environmental Medicine, School of Public Health and Community Medicine, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Nils Oskar Jõgi
- Department of Medical Sciences: Clinical Physiology, Uppsala University, Uppsala, Sweden
| | - Shyamali C Dharmage
- Centre for Epidemiology and Biostatistics, Melbourne School of Population and Global Health, University of Melbourne, Melbourne, Australia
| | - Svein Magne Skulstad
- Department of Global Public Health and Primary Care, Centre for International Health, University of Bergen, Bergen, Norway
| | - Vivi Schlünssen
- Department of Public Health, Research Unit for Environment, Work and Health, Danish Ramazzini Centre, Aarhus University Denmark, Aarhus, Denmark
| | - Cecilie Svanes
- Department of Global Public Health and Primary Care, Centre for International Health, University of Bergen, Bergen, Norway.
- Department of Occupational Medicine, Haukeland University Hospital, Bergen, Norway.
| | - John W Holloway
- Human Development and Health, Faculty of Medicine, University of Southampton, Southampton, UK
- NIHR Southampton Biomedical Research Centre, University Hospitals Southampton, Southampton, UK
| |
Collapse
|
8
|
Heikkinen A, Esser VFC, Lee SHT, Lundgren S, Hakkarainen A, Lundbom J, Kuula J, Groop PH, Heinonen S, Villicaña S, Bell JT, Maguolo A, Nilsson E, Ling C, Vaag A, Pajukanta P, Kaprio J, Pietiläinen KH, Li S, Ollikainen M. Twin pair analysis uncovers links between DNA methylation, mitochondrial DNA quantity and obesity. Nat Commun 2025; 16:4374. [PMID: 40355419 PMCID: PMC12069627 DOI: 10.1038/s41467-025-59576-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2024] [Accepted: 04/24/2025] [Indexed: 05/14/2025] Open
Abstract
Alterations in mitochondrial metabolism in obesity may indicate disrupted communication between mitochondria and nucleus, and DNA methylation may influence this interplay. Here, we leverage data from the Finnish Twin Cohort study subcohort (n = 173; 86 full twin pairs, 1 singleton), including comprehensive measurements of obesity-related outcomes, mitochondrial DNA quantity and nuclear DNA methylation levels in adipose and muscle tissue, to identify one CpG at SH3BP4 significantly associated with mitochondrial DNA quantity in adipose tissue (FDR < 0.05). We also show that SH3BP4 methylation correlates with its gene expression. Additionally, we find that 14 out of the 35 obesity-related traits display significant associations with both SH3BP4 methylation and mitochondrial DNA quantity in adipose tissue. We use data from TwinsUK and the Scandinavian T2D-discordant monozygotic twin cohort, to validate the observed associations. Further analysis using ICE FALCON suggests that mitochondrial DNA quantity, insulin sensitivity and certain body fat measures are causal to SH3BP4 methylation. Examining mitochondrial DNA quantity and obesity-related traits suggests causation from mitochondrial DNA quantity to obesity, but unmeasured within-individual confounding cannot be ruled out. Our findings underscore the impact of mitochondrial DNA quantity on DNA methylation and expression of the SH3BP4 gene within adipose tissue, with potential implications for obesity.
Collapse
Affiliation(s)
- Aino Heikkinen
- Institute for Molecular Medicine Finland (FIMM), HiLIFE, University of Helsinki, Helsinki, Finland.
- Minerva Foundation Institute for Medical Research, Helsinki, Finland.
| | - Vivienne F C Esser
- Centre for Epidemiology and Biostatistics, Melbourne School of Population and Global Health, University of Melbourne, Melbourne, VIC, Australia
| | - Seung Hyuk T Lee
- Department of Human Genetics, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Sara Lundgren
- Institute for Molecular Medicine Finland (FIMM), HiLIFE, University of Helsinki, Helsinki, Finland
| | - Antti Hakkarainen
- HUS Medical Imaging Center, Radiology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Jesper Lundbom
- HUS Medical Imaging Center, Radiology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research, Heinrich Heine University, Düsseldorf, Germany
| | - Juho Kuula
- HUS Medical Imaging Center, Radiology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
- Public Health Promotion Unit, National Institute for Health and Welfare, Helsinki, Finland
| | - Per-Henrik Groop
- Folkhälsan Institute of Genetics, Folkhälsan Research Center, Helsinki, Finland
- Research Program for Clinical and Molecular Metabolism, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- Abdominal Center, Nephrology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Sini Heinonen
- Obesity Research Unit, Research Program for Clinical and Molecular Metabolism, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- Department of Internal Medicine, Helsinki University Hospital, Helsinki, Finland
| | - Sergio Villicaña
- Department of Twin Research and Genetic Epidemiology, King's College London, London, UK
| | - Jordana T Bell
- Department of Twin Research and Genetic Epidemiology, King's College London, London, UK
| | - Alice Maguolo
- Epigenetics and Diabetes Unit, Department of Clinical Sciences in Malmö, Lund University Diabetes Centre, Scania University Hospital, Malmö, Sweden
| | - Emma Nilsson
- Epigenetics and Diabetes Unit, Department of Clinical Sciences in Malmö, Lund University Diabetes Centre, Scania University Hospital, Malmö, Sweden
| | - Charlotte Ling
- Epigenetics and Diabetes Unit, Department of Clinical Sciences in Malmö, Lund University Diabetes Centre, Scania University Hospital, Malmö, Sweden
| | - Allan Vaag
- Department of Clinical Sciences in Malmö, Lund University Diabetes Centre, Scania University Hospital, Malmö, Sweden
- Copenhagen University Hospital, Steno Diabetes Center Copenhagen, Herlev, Denmark
- Department of Endocrinology, Skåne University Hospital, Malmö, Sweden
| | - Päivi Pajukanta
- Department of Human Genetics, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
- Bioinformatics Interdepartmental Program, UCLA, Los Angeles, CA, USA
- Institute for Precision Health, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Jaakko Kaprio
- Institute for Molecular Medicine Finland (FIMM), HiLIFE, University of Helsinki, Helsinki, Finland
| | - Kirsi H Pietiläinen
- Obesity Research Unit, Research Program for Clinical and Molecular Metabolism, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- HealthyWeightHub, Endocrinology, Abdominal Center, Helsinki University Central Hospital and University of Helsinki, Helsinki, Finland
| | - Shuai Li
- Centre for Epidemiology and Biostatistics, Melbourne School of Population and Global Health, University of Melbourne, Melbourne, VIC, Australia
- Precision Medicine, School of Clinical Sciences at Monash Health, Monash University, Clayton, Victoria, Australia
| | - Miina Ollikainen
- Institute for Molecular Medicine Finland (FIMM), HiLIFE, University of Helsinki, Helsinki, Finland.
- Minerva Foundation Institute for Medical Research, Helsinki, Finland.
| |
Collapse
|
9
|
Cheng F, Shen RJ, Zheng Z, Chen ZJ, Huang PJ, Feng ZK, Li X, Lin N, Zheng M, Liang Y, Qu J, Lu F, Jin ZB, Yang J. Distinct methylomic signatures of high-altitude acclimatization and adaptation in the Tibetan Plateau. Cell Discov 2025; 11:45. [PMID: 40328746 PMCID: PMC12056056 DOI: 10.1038/s41421-025-00795-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2023] [Accepted: 03/17/2025] [Indexed: 05/08/2025] Open
Abstract
High altitude presents a challenging environment for human settlement. DNA methylation is an essential epigenetic mechanism that responds to environmental stimuli, but its roles in high-altitude short-term acclimatization (STA) and long-term adaptation (LTA) are poorly understood. Here, we conducted a methylome-wide association study involving 687 native highlanders and 299 acclimatized newcomers in the Tibetan Plateau and 462 native lowlanders to identify differentially methylated sites (DMSs) associated with STA or LTA. We identified 93 and 4070 DMSs for STA and LTA, respectively, which had no overlap, showed opposite asymmetric effect size patterns, and resided near genes enriched in distinct biological pathways/processes (e.g., cell cycle for STA and immune diseases and calcium signalling pathway for LTA). Epigenetic clock analysis revealed evidence of accelerated ageing in the acclimatized newcomers compared to the native lowlanders. Our research provides novel insights into epigenetic regulation in relation to high altitude and intervention strategies for altitude-related ageing or illnesses.
Collapse
Affiliation(s)
- Feifei Cheng
- School of Life Sciences, Westlake University, Hangzhou, Zhejiang, China
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland, Australia
| | - Ren-Juan Shen
- Beijing Institute of Ophthalmology, Beijing Tongren Hospital, Capital Medical University, Beijing, China
| | - Zhili Zheng
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland, Australia
| | - Zhen Ji Chen
- School of Ophthalmology & Optometry, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Peng-Juan Huang
- Beijing Institute of Ophthalmology, Beijing Tongren Hospital, Capital Medical University, Beijing, China
- School of Ophthalmology & Optometry, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Zhuo-Kun Feng
- School of Ophthalmology & Optometry, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Xiaoman Li
- School of Ophthalmology & Optometry, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Na Lin
- School of Ophthalmology & Optometry, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Meiqin Zheng
- School of Ophthalmology & Optometry, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Yuanbo Liang
- School of Ophthalmology & Optometry, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Jia Qu
- School of Ophthalmology & Optometry, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Fan Lu
- School of Ophthalmology & Optometry, Wenzhou Medical University, Wenzhou, Zhejiang, China.
| | - Zi-Bing Jin
- Beijing Institute of Ophthalmology, Beijing Tongren Hospital, Capital Medical University, Beijing, China.
- School of Ophthalmology & Optometry, Wenzhou Medical University, Wenzhou, Zhejiang, China.
| | - Jian Yang
- School of Life Sciences, Westlake University, Hangzhou, Zhejiang, China.
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang, China.
| |
Collapse
|
10
|
Kaczmarczyk B, de la Calle-Fabregat C, Conde A, Duarte AC, Mena-Vazquez N, Fernandez-Nebro A, Triguero-Martinez A, Castañeda S, Dos-Santos Sobrin R, Mera-Varela A, Lopez-Pedrera C, Escudero-Contreras A, Vela-Casasempere P, Molina M, Narvaez J, Retuerto-Guerrero M, Pablos JL, Sarmiento-Monroy JC, Sanmarti R, Gomez-Carrera L, Bonilla G, Remuzgo-Martinez S, Gonzalez-Gay MA, Leiro-Fernandez V, Perez-Gomez N, Vadillo-Font C, Abasolo L, Casafont-Sole I, Mateo-Soria L, Castillo-Gonzalez AC, Marras C, Perez-Pampin E, Ballestar E, Gonzalez A. DNA methylome biomarkers of rheumatoid arthritis-associated interstitial lung disease reflecting lung fibrosis pathways, an exploratory case-control study. Sci Rep 2025; 15:15123. [PMID: 40301499 PMCID: PMC12041357 DOI: 10.1038/s41598-025-99755-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2024] [Accepted: 04/22/2025] [Indexed: 05/01/2025] Open
Abstract
Rheumatoid Arthritis-associated Interstitial Lung Disease (RA-ILD) significantly reduces life quality and survival, necessitating improvements in its understanding and clinical management. We addressed these goals using DNA methylation analysis, which has not been done in RA-ILD samples, by comparing 32 RA patients with ILD diagnosed less than one year before (cases) and 32 matched RA patients without ILD (controls). This analysis identified 6679 differentially methylated positions (DMPs) with Δβ ≥ 2% and FDR < 0.05, and 576 differentially methylated regions in RA-ILD. Some DMPs were near mucin, collagen, and telomere maintenance genes. Also, the most notably enriched gene set (up to padj = 1.9 × 10-38) included genes overexpressed in fibrosis by monocytes and alveolar macrophages. Other significantly enriched gene sets, known to be dysregulated in fibrosis, included the mitotic spindle and the Rho GTPases. Additionally, analysis of transcription factor binding sites around DMPs showed unique enrichment near the liver X receptor element (LXRE), which is associated with fibrosis in multiple tissues. These results were consistent and unaffected by stricter significance thresholds. They indicated that differential DNA methylation may serve as blood biomarkers for RA-ILD including some related to lung fibrosis pathways.
Collapse
Affiliation(s)
- Bartosz Kaczmarczyk
- Experimental and Observational Rheumatology and Rheumatology Unit, Instituto Investigacion Sanitaria-Hospital Clinico Universitario de Santiago, Santiago de Compostela, Spain
| | | | - Adrian Conde
- Experimental and Observational Rheumatology and Rheumatology Unit, Instituto Investigacion Sanitaria-Hospital Clinico Universitario de Santiago, Santiago de Compostela, Spain
| | - Ana Catarina Duarte
- Rheumatology Department, Unidade Local de Saúde de Almada-Seixal - Hospital Garcia de Orta, Almada, Portugal
| | - Natalia Mena-Vazquez
- Department of Rheumatology, University Regional Hospital of Malaga (HRUM). Institute for Biomedical Research in Malaga (IBIMA), Malaga University, Málaga, Spain
| | - Antonio Fernandez-Nebro
- Department of Rheumatology, University Regional Hospital of Malaga (HRUM). Institute for Biomedical Research in Malaga (IBIMA), Malaga University, Málaga, Spain
| | - Ana Triguero-Martinez
- Rheumatology Department, Hospital Universitario de La Princesa, Instituto de Investigación Sanitaria la Princesa (IIS-Princesa), Madrid, Spain
| | - Santos Castañeda
- Rheumatology Department, Hospital Universitario de La Princesa, Instituto de Investigación Sanitaria la Princesa (IIS-Princesa), Madrid, Spain
| | - Raquel Dos-Santos Sobrin
- Experimental and Observational Rheumatology and Rheumatology Unit, Instituto Investigacion Sanitaria-Hospital Clinico Universitario de Santiago, Santiago de Compostela, Spain
| | - Antonio Mera-Varela
- Experimental and Observational Rheumatology and Rheumatology Unit, Instituto Investigacion Sanitaria-Hospital Clinico Universitario de Santiago, Santiago de Compostela, Spain
- Department of Medicine. Faculty of Medicine, University of Santiago de Compostela, Santiago de Compostela, Spain
| | - Chary Lopez-Pedrera
- Rheumatology Service, Maimonides Institute of Biomedical Research of Cordoba (IMIBIC), Reina Sofia University Hospital, University of Cordoba, Cordoba, Spain
| | - Alejandro Escudero-Contreras
- Rheumatology Service, Maimonides Institute of Biomedical Research of Cordoba (IMIBIC), Reina Sofia University Hospital, University of Cordoba, Cordoba, Spain
| | | | - Maria Molina
- Pneumology Department, Hospital Universitario Belvitge, Barcelona, Spain
| | - Javier Narvaez
- Rheumatology Department, Hospital Universitario Belvitge, Barcelona, Spain
| | - Miriam Retuerto-Guerrero
- Rheumatology Department, Hospital 12 de Octubre and Universidad Complutense de Madrid, Madrid, Spain
| | - Jose L Pablos
- Rheumatology Department, Hospital 12 de Octubre and Universidad Complutense de Madrid, Madrid, Spain
| | | | - Raimon Sanmarti
- Rheumatology Department, Hospital Clinic and IDIBAPS, Barcelona, Spain
| | - Luis Gomez-Carrera
- Pneumology Department, Instituto de Investigación Hospital Universitario La Paz (IDIPAZ), Madrid, Spain
| | - Gema Bonilla
- Rheumatology Department, Instituto de Investigación Hospital Universitario La Paz (IDIPAZ), Madrid, Spain
| | - Sara Remuzgo-Martinez
- Rheumatology Department, Hospital Universitario Marques de Valdecilla, Santander, Spain
| | - Miguel Angel Gonzalez-Gay
- Department of Medicine and Psychiatry, University of Cantabria, Santander, Spain
- Rheumatology Division, IIS-Fundación Jiménez Díaz, Madrid, Spain
| | - Virginia Leiro-Fernandez
- Pneumology Department, NeumoVigo I+i Research Group, Complejo Hospitalario Universitario de Vigo, Galicia Sur Health Research Institute (IIS Galicia Sur), SERGAS-UVIGO. CIBERES. ISCIII, Vigo, Spain
| | | | - Cristina Vadillo-Font
- Rheumatology Department, Hospital Clínico San Carlos - Instituto Investigación Sanitaria San Carlos (IdISSC), Madrid, Spain
| | - Lydia Abasolo
- Rheumatology Department, Hospital Clínico San Carlos - Instituto Investigación Sanitaria San Carlos (IdISSC), Madrid, Spain
| | - Ivette Casafont-Sole
- Rheumatology Department, Hospital Universitari Germans Trias I Pujol, Badalona, Spain
| | - Lourdes Mateo-Soria
- Rheumatology Department, Hospital Universitari Germans Trias I Pujol, Badalona, Spain
| | | | - Carlos Marras
- Rheumatology Unit, Hospital Universitario Virgen de la Arrixaca, Murcia, Spain
| | - Eva Perez-Pampin
- Experimental and Observational Rheumatology and Rheumatology Unit, Instituto Investigacion Sanitaria-Hospital Clinico Universitario de Santiago, Santiago de Compostela, Spain
- Department of Medicine. Faculty of Medicine, University of Santiago de Compostela, Santiago de Compostela, Spain
| | - Esteban Ballestar
- Epigenetics and Immune Disease Group, Josep Carreras Leukaemia Research Institute (IJC), Barcelona, Spain
| | - Antonio Gonzalez
- Experimental and Observational Rheumatology and Rheumatology Unit, Instituto Investigacion Sanitaria-Hospital Clinico Universitario de Santiago, Santiago de Compostela, Spain.
| |
Collapse
|
11
|
Hung RKY, Costeira R, Chen J, Schlosser P, Grundner-Culemann F, Booth JW, Sharpe CC, Bramham K, Sun YV, Marconi VC, Teumer A, Winkler CA, Post FA, Bell JT. Epigenetic associations with kidney disease in individuals of African ancestry with APOL1 high-risk genotypes and HIV. Nephrol Dial Transplant 2025; 40:997-1006. [PMID: 39448372 PMCID: PMC12035534 DOI: 10.1093/ndt/gfae237] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Indexed: 10/26/2024] Open
Abstract
BACKGROUND Apolipoprotein L1 (APOL1) high-risk variants are major determinants of chronic kidney disease (CKD) in people of African ancestry. Previous studies have identified epigenetic changes in relation to kidney function and CKD, but not in individuals with APOL1 high-risk genotypes. We conducted an epigenome-wide analysis of CKD and estimated glomerular filtration rate (eGFR) in in people of African ancestry and APOL1 high-risk genotypes with HIV. METHODS DNA methylation profiles from peripheral blood mononuclear cells of 119 individuals with APOL1 high-risk genotypes (mean age 48 years, 49% female, median CD4 count 515 cells/mm3, 90% HIV-1 RNA <200 copies/mL, 23% with CKD) were obtained by Illumina MethylationEPIC BeadChip. Differential methylation analysis of CKD considered technical and biological covariates. We also assessed associations with eGFR. Replication was pursued in three independent multi-ancestry cohorts with and without HIV. RESULTS DNA methylation levels at 14 regions were associated with CKD. The strongest signals were located in SCARB1, DNAJC5B and C4orf50. Seven of the 14 signals also associated with eGFR, and most showed evidence for a genetic basis. Four signals (in SCARB1, FRMD4A, CSRNP1 and RAB38) replicated in other cohorts, and 11 previously reported epigenetic signals for kidney function or CKD replicated in our cohort. We found no significant DNA methylation signals in, or near, the APOL1 promoter region. CONCLUSIONS We report several novel as well as previously reported epigenetic associations with CKD and eGFR in individuals with HIV having APOL1 high-risk genotypes. Further investigation of pathways linking DNA methylation to APOL1 nephropathies is warranted.
Collapse
Affiliation(s)
- Rachel K Y Hung
- Department of HIV and Sexual Health, King's College Hospital, London, UK
- Department of Twin Research & Genetic Epidemiology, King's College London, London, UK
| | - Ricardo Costeira
- Department of Twin Research & Genetic Epidemiology, King's College London, London, UK
| | - Junyu Chen
- Department of Epidemiology, Rollins School of Public Health, Emory University, Atlanta, GA, USA
| | - Pascal Schlosser
- Institute of Genetic Epidemiology, Faculty of Medicine and Medical Center, University of Freiburg, Freiburg, Germany
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Franziska Grundner-Culemann
- Institute of Genetic Epidemiology, Faculty of Medicine and Medical Center, University of Freiburg, Freiburg, Germany
| | - John W Booth
- Department of Renal Medicine, Bart's Health NHS Foundation Trust, London, UK
| | - Claire C Sharpe
- Department of Renal Medicine, King's College Hospital NHS Foundation Trust, London SE5 9RS, UK
| | - Kate Bramham
- Department of Renal Medicine, King's College Hospital NHS Foundation Trust, London SE5 9RS, UK
| | - Yan V Sun
- Department of Epidemiology, Rollins School of Public Health, Emory University, Atlanta, GA, USA
- Atlanta Veterans Affairs Health Care System, Decatur, GA, USA
| | - Vincent C Marconi
- Atlanta Veterans Affairs Health Care System, Decatur, GA, USA
- Department of Global Health, Rollins School of Public Health, Emory University, Atlanta, GA, USA
- School of Medicine, Emory University, Atlanta, GA, USA
| | - Alexander Teumer
- DZHK (German Centre for Cardiovascular Research), Partner Site Greifswald, Greifswald, Germany
- Department of Psychiatry and Psychotherapy, University Medicine Greifswald, Greifswald, Germany
- Department of Population Medicine and Lifestyle Diseases Prevention, Medical University of Bialystok, Bialystok, Poland
| | - Cheryl A Winkler
- Basic Reseach Program, Frederick National Laboratory for Cancer Research and the Cancer Innovation Laboratory, Center for Cancer Research, National Cancer Institute, Frederick, MD, USA
| | - Frank A Post
- Department of HIV and Sexual Health, King's College Hospital, London, UK
- Department of Twin Research & Genetic Epidemiology, King's College London, London, UK
| | - Jordana T Bell
- Department of Twin Research & Genetic Epidemiology, King's College London, London, UK
| |
Collapse
|
12
|
Zhou J, Mychaleckyj JC, Onengut-Gumuscu S, Orchard TJ, Costacou T, Miller RG. DNA methylation and 28 year incidence of two neuropathy phenotypes in type 1 diabetes: the Pittsburgh Epidemiology of Diabetes Complications cohort study. Diabetologia 2025:10.1007/s00125-025-06427-1. [PMID: 40266295 DOI: 10.1007/s00125-025-06427-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/22/2024] [Accepted: 02/19/2025] [Indexed: 04/24/2025]
Abstract
AIMS/HYPOTHESIS Diabetic peripheral neuropathy (DPN) and neuropathic pain (NP) are common complications of type 1 diabetes that can greatly affect quality of life. Studying DNA methylation (DNAm) may help identify potential therapeutic targets; however, epigenome-wide association studies (EWAS) of DPN and NP are lacking. We thus performed prospective EWAS of 28 year DPN and NP incidence in the Pittsburgh Epidemiology of Diabetes Complications (EDC) study of childhood-onset (<17 years) type 1 diabetes. METHODS DPN was defined as two or more of the following criteria: symptoms consistent with DPN; decreased tendon reflexes; or abnormal sensory examination. NP was reported as burning, aching or stabbing pain in the feet during an EDC examination or on the Michigan Neuropathy Screening Instrument (MNSI). The time of the first available blood-derived DNA specimen collected between 1988-1998 was considered the analytic 'baseline' (mean age 27 years; diabetes duration 19 years). After quality control, DNAm (EPIC array) at 683,597 CpGs was analysed in Cox models for time-to-DPN in 282 individuals free of DPN at baseline and time to NP in 365 individuals free of NP at baseline. False discovery rate (FDR) <0.05 was considered statistically significant. We also identified differentially methylated regions (DMRs), functional interaction networks and genetic variants associated with DNAm (methylation quantitative trait loci [meQTLs]), and performed Mendelian randomisation (MR) to assess evidence of causality. RESULTS Over 28 years, 154 individuals (54.6%) developed DPN and 148 (40.5%) developed NP. Greater methylation at three CpGs was significantly associated (FDR≤0.05) with reduced hazard of DPN: cg06163904 (CHMP6); cg10835127 (CACNA1B); and cg18945945 (PKNOX1). CpG associations with DPN remained similar after adjustment for clinical risk factors. We identified 75 meQTLs for cg18945945 in the PKNOX1 region, 59 of which were validated in an external diabetes cohort. One-sample MR provided nominal evidence for a potentially causal association between cg18945945 and DPN (p=0.01). While no individual CpGs were significantly associated with NP, there were 49 NP-associated DMRs. CONCLUSIONS/INTERPRETATION Our study identified associations between DNAm and 28 year incidence of DPN and NP at several biologically plausible loci. Most notably, we identified a novel association between DNAm of PKNOX1 and future DPN, including evidence of a genetic influence on PKNOX1 methylation that was validated in an external diabetes cohort. PKNOX1 has previously been implicated in drug-induced neuropathy; our results provide strong evidence that epigenetic regulation of PKNOX1 may also play a functional role in the development of diabetic neuropathy. Our results suggest that epigenetic modification of the identified loci warrants further study to inform potential targets for prevention of DPN.
Collapse
Affiliation(s)
- Jiayi Zhou
- Department of Epidemiology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Josyf C Mychaleckyj
- Center for Public Health Genomics, University of Virginia, Charlottesville, VA, USA
| | - Suna Onengut-Gumuscu
- Center for Public Health Genomics, University of Virginia, Charlottesville, VA, USA
| | - Trevor J Orchard
- Department of Epidemiology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Tina Costacou
- Department of Epidemiology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Rachel G Miller
- Department of Epidemiology, University of Pittsburgh, Pittsburgh, PA, USA.
| |
Collapse
|
13
|
Zhang W, Lukacsovich D, Young JI, Gomez L, Schmidt MA, Martin ER, Kunkle BW, Chen XS, O'Shea DM, Galvin JE, Wang L. DNA methylation signature of a lifestyle-based resilience index for cognitive health. Alzheimers Res Ther 2025; 17:88. [PMID: 40264239 PMCID: PMC12016380 DOI: 10.1186/s13195-025-01733-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2024] [Accepted: 04/06/2025] [Indexed: 04/24/2025]
Abstract
Cognitive resilience (CR) contributes to the variability in risk for developing and progressing in Alzheimer's disease (AD) among individuals. Beyond genetics, recent studies highlight the critical role of lifestyle factors in enhancing CR and delaying cognitive decline. DNA methylation (DNAm), an epigenetic mechanism influenced by both genetic and environmental factors, including CR-related lifestyle factors, offers a promising pathway for understanding the biology of CR. We studied DNAm changes associated with the Resilience Index (RI), a composite measure of lifestyle factors, using blood samples from the Healthy Brain Initiative (HBI) cohort. After corrections for multiple comparisons, our analysis identified 19 CpGs and 24 differentially methylated regions significantly associated with the RI, adjusting for covariates age, sex, APOE ε4, and immune cell composition. The RI-associated methylation changes are significantly enriched in pathways related to lipid metabolism, synaptic plasticity, and neuroinflammation, and highlight the connection between cardiovascular health and cognitive function. By identifying RI-associated DNAm, our study provided an alternative approach to discovering future targets and treatment strategies for AD, complementary to the traditional approach of identifying disease-associated variants directly. Furthermore, we developed a Methylation-based Resilience Score (MRS) that successfully predicted future cognitive decline in an external dataset from the Alzheimer's Disease Neuroimaging Initiative (ADNI), even after accounting for age, sex, APOE ε4, years of education, baseline diagnosis, and baseline MMSE score. Our findings are particularly relevant for a better understanding of epigenetic architecture underlying cognitive resilience. Importantly, the significant association between baseline MRS and future cognitive decline demonstrated that DNAm could be a predictive marker for AD, laying the foundation for future studies on personalized AD prevention.
Collapse
Affiliation(s)
- Wei Zhang
- Division of Biostatistics, Department of Public Health Sciences, University of Miami, Miller School of Medicine, Miami, FL, 33136, USA
| | - David Lukacsovich
- Division of Biostatistics, Department of Public Health Sciences, University of Miami, Miller School of Medicine, Miami, FL, 33136, USA
| | - Juan I Young
- Dr. John T Macdonald Foundation Department of Human Genetics, University of Miami, Miller School of Medicine, Miami, FL, 33136, USA
- John P. Hussman Institute for Human Genomics, University of Miami Miller School of Medicine, Miami, FL, 33136, USA
| | - Lissette Gomez
- John P. Hussman Institute for Human Genomics, University of Miami Miller School of Medicine, Miami, FL, 33136, USA
| | - Michael A Schmidt
- Dr. John T Macdonald Foundation Department of Human Genetics, University of Miami, Miller School of Medicine, Miami, FL, 33136, USA
- John P. Hussman Institute for Human Genomics, University of Miami Miller School of Medicine, Miami, FL, 33136, USA
| | - Eden R Martin
- Dr. John T Macdonald Foundation Department of Human Genetics, University of Miami, Miller School of Medicine, Miami, FL, 33136, USA
- John P. Hussman Institute for Human Genomics, University of Miami Miller School of Medicine, Miami, FL, 33136, USA
| | - Brian W Kunkle
- Dr. John T Macdonald Foundation Department of Human Genetics, University of Miami, Miller School of Medicine, Miami, FL, 33136, USA
- John P. Hussman Institute for Human Genomics, University of Miami Miller School of Medicine, Miami, FL, 33136, USA
| | - X Steven Chen
- Division of Biostatistics, Department of Public Health Sciences, University of Miami, Miller School of Medicine, Miami, FL, 33136, USA
- Sylvester Comprehensive Cancer Center, University of Miami, Miller School of Medicine, Miami, FL, 33136, USA
| | - Deirdre M O'Shea
- Comprehensive Center for Brain Health, Department of Neurology, University of Miami Miller School of Medicine, Miami, FL, 33433, USA.
| | - James E Galvin
- Comprehensive Center for Brain Health, Department of Neurology, University of Miami Miller School of Medicine, Miami, FL, 33433, USA.
| | - Lily Wang
- Division of Biostatistics, Department of Public Health Sciences, University of Miami, Miller School of Medicine, Miami, FL, 33136, USA.
- Dr. John T Macdonald Foundation Department of Human Genetics, University of Miami, Miller School of Medicine, Miami, FL, 33136, USA.
- John P. Hussman Institute for Human Genomics, University of Miami Miller School of Medicine, Miami, FL, 33136, USA.
- Sylvester Comprehensive Cancer Center, University of Miami, Miller School of Medicine, Miami, FL, 33136, USA.
| |
Collapse
|
14
|
McKenna BG, Lussier AA, Suderman MJ, Walton E, Simpkin AJ, Hüls A, Dunn EC. Strengthening Rigor and Reproducibility in Epigenome-Wide Association Studies of Social Exposures and Brain-Based Health Outcomes. Curr Environ Health Rep 2025; 12:19. [PMID: 40254641 PMCID: PMC12009779 DOI: 10.1007/s40572-024-00469-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/16/2024] [Indexed: 04/22/2025]
Abstract
PURPOSE OF REVIEW Studies examining the effects of social factors on the epigenome have proliferated over the last two decades. Social epigenetics research to date has broadly demonstrated that social factors spanning childhood adversity, to neighborhood disadvantage, educational attainment, and economic instability are associated with alterations to DNA methylation that may have a functional impact on health. These relationships are particularly relevant to brain-based health outcomes such as psychiatric disorders, which are strongly influenced by social exposures and are also the leading cause of disability worldwide. However, social epigenetics studies are limited by the many challenges faced by both epigenome-wide association studies (EWAS) and the study of social factors. FINDINGS In this manuscript, we provide a framework to achieve greater rigor and reproducibility in social epigenetics research. We discuss current limitations of the social epigenetics field, as well as existing and new solutions to improve rigor and reproducibility. Readers will gain a better understanding of the current considerations and processes that could maximize rigor when conducting social epigenetics research, as well as the technologies and approaches that merit attention and investment to propel continued discovery into the future.
Collapse
Affiliation(s)
- Brooke G McKenna
- Center for Psychiatric and Neurodevelopmental Genetics Unit, Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA.
- Department of Psychiatry, Harvard Medical School, Boston, MA, USA.
- Department of Sociology, Purdue University, West Lafayette, IN, USA.
| | - Alexandre A Lussier
- Center for Psychiatric and Neurodevelopmental Genetics Unit, Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA
- Department of Psychiatry, Harvard Medical School, Boston, MA, USA
| | - Matthew J Suderman
- MRC Integrative Epidemiology Unit, Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
| | - Esther Walton
- Department of Psychology, University of Bath, Bath, UK
| | - Andrew J Simpkin
- School of Mathematical and Statistical Sciences, University of Galway, Galway, Ireland
| | - Anke Hüls
- Rollins School of Public Health, Emory University, Atlanta, GA, USA
| | - Erin C Dunn
- Center for Psychiatric and Neurodevelopmental Genetics Unit, Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA
- Department of Psychiatry, Harvard Medical School, Boston, MA, USA
- Department of Sociology, Purdue University, West Lafayette, IN, USA
| |
Collapse
|
15
|
Hong P, Waldenberger M, Pritsch M, Gilberg L, Brand I, Bruger J, Frese J, Castelletti N, Garí M, Geldmacher C, Hoelscher M, Peters A, Matías-García PR. Differential DNA methylation 7 months after SARS-CoV-2 infection. Clin Epigenetics 2025; 17:60. [PMID: 40251596 PMCID: PMC12008906 DOI: 10.1186/s13148-025-01866-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2025] [Accepted: 03/26/2025] [Indexed: 04/20/2025] Open
Abstract
BACKGROUND Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) causes coronavirus disease 2019 (COVID-19), and SARS-CoV-2 has been linked to changes in DNA methylation (DNAm) patterns. Studies focused on post-SARS-CoV-2 infection and DNAm have been mainly carried out among severe COVID-19 cases or without distinguishing the severity of cases. However, investigations into mild and asymptomatic cases after SARS-CoV-2 infection are limited. In this study, we analyzed DNAm patterns of mild and asymptomatic cases seven months after SARS-CoV-2 infection in a household setting by conducting epigenome-wide association studies (EWAS). RESULTS We identified DNAm changes at 42 CpG sites associated with anti-SARS-CoV-2 antibody levels. We additionally report EWAS between COVID-19 cases and controls, with the case status being confirmed by either an antibody test or a PCR test. The EWAS with an antibody test case definition identified 172 CpG sites to be differentially methylated, while the EWAS with a PCR test case definition identified 502 CpG sites. Two common sites, namely cg17126990 (annotated to AFAP1L2) and cg25483596 (annotated to PC), were identified to be hypermethylated across the three EWAS. Both CpG sites have been reported to be involved in molecular pathways after SARS-CoV-2 infection. While AFAP1L2 has been found to be upregulated after SARS-CoV-2 infection, the pyruvate carboxylase (PC) activity seems to be affected by SARS-CoV-2 infection resulting in changes to the host cell metabolism. Additionally, an EWAS to assess persistent health restrictions among PCR-confirmed cases showed 40 CpG sites to be differentially methylated. CONCLUSIONS We detected associations between DNAm in individuals who had asymptomatic and mild SARS-CoV-2 infections as compared to their household controls. These findings contribute to our understanding of the molecular consequences of SARS-CoV-2 infection observed months after infection.
Collapse
Grants
- 01KI20271 Bavarian State Ministry of Science and the Arts, University Hospital, LMU Munich, Helmholtz Centre Munich, University of Bonn, University of Bielefeld, German Ministry for Education and Research
- 01KI20271 Bavarian State Ministry of Science and the Arts, University Hospital, LMU Munich, Helmholtz Centre Munich, University of Bonn, University of Bielefeld, German Ministry for Education and Research
- 01KI20271 Bavarian State Ministry of Science and the Arts, University Hospital, LMU Munich, Helmholtz Centre Munich, University of Bonn, University of Bielefeld, German Ministry for Education and Research
- 01KI20271 Bavarian State Ministry of Science and the Arts, University Hospital, LMU Munich, Helmholtz Centre Munich, University of Bonn, University of Bielefeld, German Ministry for Education and Research
- 01KI20271 Bavarian State Ministry of Science and the Arts, University Hospital, LMU Munich, Helmholtz Centre Munich, University of Bonn, University of Bielefeld, German Ministry for Education and Research
- 01KI20271 Bavarian State Ministry of Science and the Arts, University Hospital, LMU Munich, Helmholtz Centre Munich, University of Bonn, University of Bielefeld, German Ministry for Education and Research
- 01KI20271 Bavarian State Ministry of Science and the Arts, University Hospital, LMU Munich, Helmholtz Centre Munich, University of Bonn, University of Bielefeld, German Ministry for Education and Research
- 01KI20271 Bavarian State Ministry of Science and the Arts, University Hospital, LMU Munich, Helmholtz Centre Munich, University of Bonn, University of Bielefeld, German Ministry for Education and Research
- 01KI20271 Bavarian State Ministry of Science and the Arts, University Hospital, LMU Munich, Helmholtz Centre Munich, University of Bonn, University of Bielefeld, German Ministry for Education and Research
- 01KI20271 Bavarian State Ministry of Science and the Arts, University Hospital, LMU Munich, Helmholtz Centre Munich, University of Bonn, University of Bielefeld, German Ministry for Education and Research
- 01KI20271 Bavarian State Ministry of Science and the Arts, University Hospital, LMU Munich, Helmholtz Centre Munich, University of Bonn, University of Bielefeld, German Ministry for Education and Research
- 01KI20271 Bavarian State Ministry of Science and the Arts, University Hospital, LMU Munich, Helmholtz Centre Munich, University of Bonn, University of Bielefeld, German Ministry for Education and Research
- 01KI20271 Bavarian State Ministry of Science and the Arts, University Hospital, LMU Munich, Helmholtz Centre Munich, University of Bonn, University of Bielefeld, German Ministry for Education and Research
- 101016167 European Union's Horizon 2020 research and innovation program, ORCHESTRA
- 101016167 European Union's Horizon 2020 research and innovation program, ORCHESTRA
- 101016167 European Union's Horizon 2020 research and innovation program, ORCHESTRA
- 101016167 European Union's Horizon 2020 research and innovation program, ORCHESTRA
- 101016167 European Union's Horizon 2020 research and innovation program, ORCHESTRA
- 101016167 European Union's Horizon 2020 research and innovation program, ORCHESTRA
- 101016167 European Union's Horizon 2020 research and innovation program, ORCHESTRA
- 101016167 European Union's Horizon 2020 research and innovation program, ORCHESTRA
- 101016167 European Union's Horizon 2020 research and innovation program, ORCHESTRA
- 101016167 European Union's Horizon 2020 research and innovation program, ORCHESTRA
- 101016167 European Union's Horizon 2020 research and innovation program, ORCHESTRA
- 101016167 European Union's Horizon 2020 research and innovation program, ORCHESTRA
- 101016167 European Union's Horizon 2020 research and innovation program, ORCHESTRA
- European Union’s Horizon 2020 research and innovation program, ORCHESTRA
- Helmholtz Zentrum München - Deutsches Forschungszentrum für Gesundheit und Umwelt (GmbH) (4209)
Collapse
Affiliation(s)
- Peizhen Hong
- Research Unit of Molecular Epidemiology, Institute of Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany.
- Institute of Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany.
- Institute for Medical Information Processing, Biometry, and Epidemiology (IBE), Faculty of Medicine, Ludwig-Maximilians-Universität (LMU) Munich, Munich, Germany.
- Pettenkofer School of Public Health, Munich, Germany.
| | - Melanie Waldenberger
- Research Unit of Molecular Epidemiology, Institute of Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
- Institute of Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
- German Centre for Cardiovascular Research (DZHK), Partner Site Munich Heart Alliance, Munich, Germany
| | - Michael Pritsch
- Institute of Infectious Diseases and Tropical Medicine, LMU University Hospital, LMU Munich, Munich, Germany
- German Center for Infection Research (DZIF), Partner Site Munich, Munich, Germany
| | - Leonard Gilberg
- Institute of Infectious Diseases and Tropical Medicine, LMU University Hospital, LMU Munich, Munich, Germany
- Department of Infectious Diseases, LMU University Hospital, LMU Munich, Munich, Germany
| | - Isabel Brand
- Institute of Infectious Diseases and Tropical Medicine, LMU University Hospital, LMU Munich, Munich, Germany
- Division of Clinical Pharmacology, Department of Medicine IV, LMU University Hospital, LMU, Munich, Germany
| | - Jan Bruger
- Institute of Infectious Diseases and Tropical Medicine, LMU University Hospital, LMU Munich, Munich, Germany
| | - Jonathan Frese
- Institute of Infectious Diseases and Tropical Medicine, LMU University Hospital, LMU Munich, Munich, Germany
| | - Noemi Castelletti
- Institute of Infectious Diseases and Tropical Medicine, LMU University Hospital, LMU Munich, Munich, Germany
| | - Mercè Garí
- Institute of Computational Biology, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
| | - Christof Geldmacher
- Institute of Infectious Diseases and Tropical Medicine, LMU University Hospital, LMU Munich, Munich, Germany
- German Center for Infection Research (DZIF), Partner Site Munich, Munich, Germany
- Immunology, Infection and Pandemic Research, Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, 80799, Munich, Germany
| | - Michael Hoelscher
- Institute of Infectious Diseases and Tropical Medicine, LMU University Hospital, LMU Munich, Munich, Germany
- German Center for Infection Research (DZIF), Partner Site Munich, Munich, Germany
- Center for International Health (CIH), University Hospital, LMU Munich, Munich, Germany
- Immunology, Infection and Pandemic Research, Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, 80799, Munich, Germany
| | - Annette Peters
- Research Unit of Molecular Epidemiology, Institute of Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
- Institute of Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
- Institute for Medical Information Processing, Biometry, and Epidemiology (IBE), Faculty of Medicine, Ludwig-Maximilians-Universität (LMU) Munich, Munich, Germany
- Pettenkofer School of Public Health, Munich, Germany
- German Centre for Cardiovascular Research (DZHK), Partner Site Munich Heart Alliance, Munich, Germany
| | - Pamela R Matías-García
- Research Unit of Molecular Epidemiology, Institute of Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany.
- Institute of Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany.
- German Centre for Cardiovascular Research (DZHK), Partner Site Munich Heart Alliance, Munich, Germany.
| |
Collapse
|
16
|
Chambers J, Roscoe CMP, Chidley C, Wisniewska A, Duggirala A. Molecular Effects of Physical Activity and Body Composition: A Systematic Review and Meta-Analysis. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2025; 22:637. [PMID: 40283858 PMCID: PMC12026539 DOI: 10.3390/ijerph22040637] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/18/2025] [Revised: 04/01/2025] [Accepted: 04/15/2025] [Indexed: 04/29/2025]
Abstract
Physical activity (PA) and body composition are important lifestyle factors that influence public health. Research suggests that DNA regions (CpG site locations) are differentially methylated in a physically active population. This meta-analysis aimed to identify CpG sites associated with various levels of PA and associated metabolic pathways. The meta-analysis followed PRISMA guidelines using PubMed, SportDISCUS, Embase, Scopus, Cochrane and Web of Science. Epigenomic analyses performed on DNA of participants with no underlying health conditions were included. Articles were screened using Rayyan AI and extracted CpG sites, and their location were confirmed using the EWAS catalogue. Six studies comprising 770 subjects were included in this meta-analysis. The meta-analysis was performed on clinical metrics extracted from the six studies and showed that BMI, blood pressure, insulin and glucose testing are significantly improved upon PA intervention. Amongst the included studies, a total of 257 CpG sites were differentially methylated in physically active participants, with 134 CpGs located in 92 genes associated with obesity-related pathways. The identified differentially methylated genes either belonged to the lipid metabolism or insulin signalling pathway. The genes which were differentially regulated in multiple tissue types and studies are JAZF1 (insulin signalling, and lipid and carbohydrate metabolism pathways) and NAV1 (mTOR signalling pathway). In conclusion, the current epigenomic meta-analysis showed that PA levels induce differential DNA methylation signatures on genes that affect metabolism. To understand the positive molecular effects of PA, further research on the above candidate genes needs to be conducted amongst various levels of a physically active population.
Collapse
Affiliation(s)
- Jenni Chambers
- Biomedical and Clinical Sciences, School of Science, University of Derby, Derby DE22 1GB, UK; (J.C.)
- Clinical Exercise Rehabilitation Research Centre, School of Sport and Exercise Science, University of Derby, Derby DE22 1GB, UK;
| | - Clare M. P. Roscoe
- Clinical Exercise Rehabilitation Research Centre, School of Sport and Exercise Science, University of Derby, Derby DE22 1GB, UK;
| | - Corinna Chidley
- Clinical Exercise Rehabilitation Research Centre, School of Sport and Exercise Science, University of Derby, Derby DE22 1GB, UK;
| | - Agnieszka Wisniewska
- Biomedical and Clinical Sciences, School of Science, University of Derby, Derby DE22 1GB, UK; (J.C.)
| | - Aparna Duggirala
- Biomedical and Clinical Sciences, School of Science, University of Derby, Derby DE22 1GB, UK; (J.C.)
| |
Collapse
|
17
|
Neumann A, Sammallahti S, Cosin-Tomas M, Reese SE, Suderman M, Alemany S, Almqvist C, Andrusaityte S, Arshad SH, Bakermans-Kranenburg MJ, Beilin L, Breton C, Bustamante M, Czamara D, Dabelea D, Eng C, Eskenazi B, Fuemmeler BF, Gilliland FD, Grazuleviciene R, Håberg SE, Herberth G, Holland N, Hough A, Hu D, Huen K, Hüls A, Jarvelin MR, Jin J, Julvez J, Koletzko BV, Koppelman GH, Kull I, Lu X, Maitre L, Mason D, Melén E, Merid SK, Molloy PL, Mori TA, Mulder RH, Page CM, Richmond RC, Röder S, Ross JP, Schellhas L, Sebert S, Sheppard D, Snieder H, Starling AP, Stein DJ, Tindula G, van IJzendoorn MH, Vonk J, Walton E, Witonsky J, Xu CJ, Yang IV, Yousefi PD, Zar HJ, Zenclussen AC, Zhang H, Tiemeier H, London SJ, Felix JF, Cecil C. Epigenetic timing effects on child developmental outcomes: a longitudinal meta-regression of findings from the Pregnancy And Childhood Epigenetics Consortium. Genome Med 2025; 17:39. [PMID: 40229801 PMCID: PMC11995515 DOI: 10.1186/s13073-025-01451-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2024] [Accepted: 03/10/2025] [Indexed: 04/16/2025] Open
Abstract
BACKGROUND DNA methylation (DNAm) is a developmentally dynamic epigenetic process; yet, most epigenome-wide association studies (EWAS) have examined DNAm at only one timepoint or without systematic comparisons between timepoints. Thus, it is unclear whether DNAm alterations during certain developmental periods are more informative than others for health outcomes, how persistent epigenetic signals are across time, and whether epigenetic timing effects differ by outcome. METHODS We applied longitudinal meta-regression models to published meta-analyses from the PACE consortium that examined DNAm at two timepoints-prospectively at birth and cross-sectionally in childhood-in relation to the same child outcome (ADHD symptoms, general psychopathology, sleep duration, BMI, asthma). These models allowed systematic comparisons of effect sizes and statistical significance between timepoints. Furthermore, we tested correlations between DNAm regression coefficients to assess the consistency of epigenetic signals across time and outcomes. Finally, we performed robustness checks, estimated between-study heterogeneity, and tested pathway enrichment. RESULTS Our findings reveal three new insights: (i) across outcomes, DNAm effect sizes are consistently larger in childhood cross-sectional analyses compared to prospective analyses at birth; (ii) higher effect sizes do not necessarily translate into more significant findings, as associations also become noisier in childhood for most outcomes (showing larger standard errors in cross-sectional vs prospective analyses); and (iii) DNAm signals are highly time-specific, while also showing evidence of shared associations across health outcomes (ADHD symptoms, general psychopathology, and asthma). Notably, these observations could not be explained by sample size differences and only partly to differential study-heterogeneity. DNAm sites changing associations were enriched for neural pathways. CONCLUSIONS Our results highlight developmentally-specific associations between DNAm and child health outcomes, when assessing DNAm at birth vs childhood. This implies that EWAS results from one timepoint are unlikely to generalize to another. Longitudinal studies with repeated epigenetic assessments are direly needed to shed light on the dynamic relationship between DNAm, development and health, as well as to enable the creation of more reliable and generalizable epigenetic biomarkers. More broadly, this study underscores the importance of considering the time-varying nature of DNAm in epigenetic research and supports the potential existence of epigenetic "timing effects" on child health.
Collapse
Affiliation(s)
- Alexander Neumann
- Child and Adolescent Psychiatry/Psychology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, Netherlands.
| | - Sara Sammallahti
- Department of Obstetrics and Gynecology, University of Helsinki, Helsinki, Finland
- Helsinki University Hospital, Helsinki, Finland
| | - Marta Cosin-Tomas
- ISGlobal, Barcelona, Spain
- Universitat Pompeu Fabra (UPF), Barcelona, Spain
- CIBER Epidemiología y Salud Pública, Madrid, Spain
| | - Sarah E Reese
- Clinical Research Practice, Westat, Rockville, MD, USA
| | - Matthew Suderman
- Bristol Medical School, Bristol Medical School, Population Health Sciences, University of Bristol, Bristol, UK
| | - Silvia Alemany
- Psychiatric Genetics Unit, Group of Psychiatry, Mental Health and Addictions, Vall d'Hebron Research Institute (VHIR), Barcelona, Spain
- Department of Mental Health, Hospital Universitari Vall d'Hebron, Barcelona, Spain
- Biomedical Network Research Centre on Mental Health (CIBERSAM), Biomedical Network Research Centre on Mental Health (CIBERSAM), Instituto de Salud Carlos III, Madrid, Spain
| | - Catarina Almqvist
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - Sandra Andrusaityte
- Department of Environmental Sciences, Vytautas Magnus University, Kaunas, Lithuania
| | - Syed H Arshad
- Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, UK
| | | | - Lawrence Beilin
- Medical School, Royal Perth Hospital Unit, the University of Western Australia, Perth, Australia
| | - Carrie Breton
- Population and Public Health Sciences, Environmental Health, University of Southern California, Los Angeles, USA
| | - Mariona Bustamante
- ISGlobal, Barcelona, Spain
- Universitat Pompeu Fabra (UPF), Barcelona, Spain
- CIBER Epidemiología y Salud Pública, Madrid, Spain
| | - Darina Czamara
- Department Genes and Environment, Max-Planck-Institute of Psychiatry, Munich, Germany
| | - Dana Dabelea
- Lifecourse Epidemiology of Adiposity and Diabetes (LEAD) Center, University of Colorado Anschutz Medical Campus, Aurora, USA
| | - Celeste Eng
- Department of Medicine, Pulmonary, Critical Care, Allergy and Sleep, University of California, San Francisco, USA
| | - Brenda Eskenazi
- Center for Environmental Research and Community Health (CERCH), School of Public Health, University of California, Berkeley, USA
| | - Bernard F Fuemmeler
- Family Medicine and Population Health, School of Medicine, Virginia Commonwealth University, Richmond, USA
| | - Frank D Gilliland
- Depatment of Population and Public Health Sciences, Keck Schools of Medicine, University of Southern California, Los Angeles, USA
| | | | - Siri E Håberg
- Centre for Fertility and Health, Norwegian Institute of Public Health, Oslo, Norway
| | - Gunda Herberth
- Department of Environmental Immunology, Helmholtz Centre for Environmental Research - UFZ, Leipzig, Germany
| | - Nina Holland
- Division of Environmental Health Sciences, School of Public Health, Children'S Environmental Health Laboratory, University of California, Berkeley, USA
| | - Amy Hough
- Born in Bradford, Bradford Institute for Health Research, Bradford Teaching Hospitals NHS Foundation Trust, Bradford, UK
| | - Donglei Hu
- Department of Medicine, Division of General Internal Medicine, University of California, San Francisco, USA
| | - Karen Huen
- Division of Environmental Health Sciences, School of Public Health, Children'S Environmental Health Laboratory, University of California, Berkeley, USA
| | - Anke Hüls
- Department of Epidemiology, Rollins School of Public Health, Emory University, Atlanta, USA
- Gangarosa Department of Environmental Health, Rollins School of Public Health, Emory University, Atlanta, USA
- Department of Biostatistics and Bioinformatics, Rollins School of Public Health, Emory University, Atlanta, USA
| | - Marjo-Riitta Jarvelin
- MRC Centre for Environment and Health, School of Public Health, Imperial College London, London, UK
- Research Unit of Population Health, Faculty of Medicine, University of Oulu, Oulu, Finland
- Unit of Primary Care, Oulu University Hospital, Oulu, Finland
- Department of Life Sciences, College of Health and Life Sciences, Brunel University London, London, UK
| | - Jianping Jin
- Public Health Practice, WESTAT, Research Triangle Park, Raleigh, NC, USA
| | - Jordi Julvez
- Clinical and Epidemiological Neuroscience (NeuroÈpia), Institut d'Investigació Sanitària Pere Virgili (IISPV), Tarragona, Spain
| | - Berthold V Koletzko
- Department of Paediatrics, Division of Metabolic and Nutritional Medicine, Hauner Children's Hospital, LMU - Ludwig Maximilians Universitaet Muenchen, Munich, Germany
| | - Gerard H Koppelman
- Department of Pediatric Pulmonology and Pediatric Allergology, Beatrix Children'S Hospital and GRIAC Research Institute, University of Groningen, University Medical Center Groningen, Groningen, Netherlands
| | - Inger Kull
- Department of Clinical Sciences and Education, Södersjukhuset, Karolinska Institutet, Stockholm, Sweden
| | - Xueling Lu
- Department of Epidemiology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Léa Maitre
- Environment and Health over the Lifecourse Program, Isglobal, Barcelona, Spain
- Universitat Pompeu Fabra (UPF), Barcelona, Spain
- CIBER Epidemiología y Salud Pública, Barcelona, Spain
| | - Dan Mason
- Born in Bradford, Bradford Institute for Health Research, Bradford Teaching Hospitals NHS Foundation Trust, Bradford, UK
| | - Erik Melén
- Department for Clinical Science and Education, Södersjukhuset, Karolinska Institutet, Stockholm, Sweden
| | - Simon K Merid
- Department for Clinical Science and Education, Södersjukhuset, Karolinska Institutet, Stockholm, Sweden
| | | | - Trevor A Mori
- Medical School, Royal Perth Hospital Unit, the University of Western Australia, Perth, Australia
| | - Rosa H Mulder
- Psychiatry/Psychology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Christian M Page
- Department of Physical Health and Ageing, Division for Physical and Mental Health, Norwegian Institute of Public Health, Oslo, Norway
| | - Rebecca C Richmond
- Bristol Medical School, Bristol Medical School, Population Health Sciences, University of Bristol, Bristol, UK
| | - Stefan Röder
- Department of Environmental Immunology, Helmholtz Centre for Environmental Research - UFZ, Leipzig, Germany
| | - Jason P Ross
- Human Health, Health and Biosecurity, CSIRO, Canberra, Australia
| | - Laura Schellhas
- School of Psychological Science, University of Bristol, Bristol, UK
| | - Sylvain Sebert
- Research Unit of Population Health, University of Oulu, Oulu, Finland
| | - Dean Sheppard
- Department of Medicine, Critical Care, Allergy and Sleep, University of California, PulmonarySan Francisco, San Francisco, CA, USA
| | - Harold Snieder
- Department of Epidemiology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Anne P Starling
- Department of Epidemiology, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, USA
| | - Dan J Stein
- SAMRC Unit on Risk & Resilience in Mental Disorders, Dept of Psychiatry & Neuroscience Institute, University of Cape Town, Cape Town, South Africa
| | - Gwen Tindula
- Epidemiology and Population Health, Stanford School of Medicine, Stanford University, Stanford, USA
| | - Marinus H van IJzendoorn
- Research Department of Clinical, Education and Health Psychology, Faculty of Brain Sciences, UCL, London, UK
- Faculty of Medicine, Nursing and Health, Psychiatry Monash Health, Monash University, Melbourne, Australia
- Faculty of Psychology and Humanities, Universidad San Sebastián, Valdivia, Chile
| | - Judith Vonk
- Department of Epidemiology, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
- GRIAC Research Institute, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Esther Walton
- Department of Psychology, University of Bath, Bath, UK
| | - Jonathan Witonsky
- Department of Pediatrics, Allergy, Immunology and BMT, University of California, San Francisco, San Francisco, CA, USA
| | - Cheng-Jian Xu
- Centre for Individualised Infection Medicine (Ciim), Helmholtz Centre for Infection Research (HZI), Hannover Medical School (MHH), Hanover, Germany
- Helmholtz Centre for Infection Research (HZI), TWINCORE, Hannover Medical School (MHH), Hanover, Germany
| | - Ivana V Yang
- Department of Biomedical Informatics, University of Colorado Anschutz Medical Campus, Aurora, USA
| | - Paul D Yousefi
- Bristol Medical School, Bristol Medical School, Population Health Sciences, University of Bristol, Bristol, UK
| | - Heather J Zar
- SAMRC Unit on Child Health, Dept of Paediatrics, University of Cape Town, Cape Town, South Africa
| | - Ana C Zenclussen
- Department of Environmental Immunology, Helmholtz Centre for Environmental Research - UFZ, Leipzig, Germany
| | - Hongmei Zhang
- Epidemiology, Biostatistics, School of Public Health, And Environmental Health, University of Memphis, Memphis, USA
| | - Henning Tiemeier
- Child and Adolescent Psychiatry/Psychology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, Netherlands
- Department of Social and Behavioral Science, Harvard T. H. Chan School of Public Health, Boston, USA
| | - Stephanie J London
- Immunity Inflammation and Disease Laboratory, National Institute of Environmental Health Sciences, Durham, USA
| | - Janine F Felix
- Generation R Study Group, Erasmus MC, University Medical Center Rotterdam, Rotterdam, Netherlands
- Department of Pediatrics, Erasmus MC, University Medical Center Rotterdam, Rotterdam, Netherlands
| | - Charlotte Cecil
- Child and Adolescent Psychiatry/Psychology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, Netherlands
- Department of Epidemiology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, Netherlands
- Department of Biomedical Data Sciences, Molecular Epidemiology, Leiden University Medical Center, Leiden, Netherlands
| |
Collapse
|
18
|
Montano C, Timp W. Evolution of genome-wide methylation profiling technologies. Genome Res 2025; 35:572-582. [PMID: 40228903 PMCID: PMC12047278 DOI: 10.1101/gr.278407.123] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/16/2025]
Abstract
In this mini-review, we explore the advancements in genome-wide DNA methylation profiling, tracing the evolution from traditional methods such as methylation arrays and whole-genome bisulfite sequencing to the cutting-edge single-molecule profiling enabled by long-read sequencing (LRS) technologies. We highlight how LRS is transforming clinical and translational research, particularly by its ability to simultaneously measure genetic and epigenetic information, providing a more comprehensive understanding of complex disease mechanisms. We discuss current challenges and future directions in the field, emphasizing the need for innovative computational tools and robust, reproducible approaches to fully harness the capabilities of LRS in molecular diagnostics.
Collapse
Affiliation(s)
- Carolina Montano
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, Maryland 21218, USA
- Division of Human Genetics, Department of Pediatrics, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania 19104, USA
| | - Winston Timp
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, Maryland 21218, USA;
| |
Collapse
|
19
|
Chybowska AD, Bernabeu E, Yousefi P, Suderman M, Hillary RF, Clark R, MacGillivray L, Murphy L, Harris SE, Corley J, Campbell A, Spires-Jones TL, McCartney DL, Cox SR, Price JF, Evans KL, Marioni RE. A blood- and brain-based EWAS of smoking. Nat Commun 2025; 16:3210. [PMID: 40180905 PMCID: PMC11968855 DOI: 10.1038/s41467-025-58357-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Accepted: 03/18/2025] [Indexed: 04/05/2025] Open
Abstract
DNA methylation offers an objective method to assess the impact of smoking. In this work, we conduct a Bayesian EWAS of smoking pack years (n = 17,865, ~850k sites, Illumina EPIC array) and extend it by analysing whole genome data of smokers and non-smokers from Generation Scotland (n = 46, ~4-21 million sites via TWIST and Oxford Nanopore sequencing). We develop mCigarette, an epigenetic biomarker of smoking, and test it in two British cohorts. Results of brain- and blood-based EWAS (nbrain=14, nblood = 882, >450k sites, Illumina arrays) reveal several loci with near-perfect discrimination of smoking status, but which do not overlap across tissues. Furthermore, we perform a GWAS of epigenetic smoking, identifying several smoking-related loci. Overall, we improve smoking-related biomarker accuracy and enhance the understanding of the effects of smoking by integrating DNA methylation data from multiple tissues and cohorts.
Collapse
Affiliation(s)
- Aleksandra D Chybowska
- Centre for Genomic and Experimental Medicine, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, EH4 2XU, UK
| | - Elena Bernabeu
- Centre for Genomic and Experimental Medicine, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, EH4 2XU, UK
| | - Paul Yousefi
- Medical Research Council Integrative Epidemiology Unit at the University of Bristol, University of Bristol, Bristol, UK
- NIHR Bristol Biomedical Research Centre, University Hospitals Bristol and Weston NHS Foundation Trust and University of Bristol, Bristol, BS8 2BN, UK
- Population Health Science, Bristol Medical School, University of Bristol, Bristol, UK
| | - Matthew Suderman
- Medical Research Council Integrative Epidemiology Unit at the University of Bristol, University of Bristol, Bristol, UK
- NIHR Bristol Biomedical Research Centre, University Hospitals Bristol and Weston NHS Foundation Trust and University of Bristol, Bristol, BS8 2BN, UK
- Population Health Science, Bristol Medical School, University of Bristol, Bristol, UK
| | - Robert F Hillary
- Centre for Genomic and Experimental Medicine, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, EH4 2XU, UK
| | - Richard Clark
- Edinburgh Clinical Research Facility, University of Edinburgh, Western General Hospital, Edinburgh, EH4 2XU, UK
| | - Louise MacGillivray
- Edinburgh Clinical Research Facility, University of Edinburgh, Western General Hospital, Edinburgh, EH4 2XU, UK
| | - Lee Murphy
- Edinburgh Clinical Research Facility, University of Edinburgh, Western General Hospital, Edinburgh, EH4 2XU, UK
| | - Sarah E Harris
- Lothian Birth Cohorts, Department of Psychology, The University of Edinburgh, Edinburgh, UK
| | - Janie Corley
- Lothian Birth Cohorts, Department of Psychology, The University of Edinburgh, Edinburgh, UK
| | - Archie Campbell
- Centre for Genomic and Experimental Medicine, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, EH4 2XU, UK
- Usher Institute, University of Edinburgh, 5-7 Little France Road, Edinburgh, EH16 4UX, UK
| | - Tara L Spires-Jones
- Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh, UK
- UK Dementia Research Institute, University of Edinburgh, Edinburgh, UK
| | - Daniel L McCartney
- Centre for Genomic and Experimental Medicine, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, EH4 2XU, UK
| | - Simon R Cox
- Lothian Birth Cohorts, Department of Psychology, The University of Edinburgh, Edinburgh, UK
- Scottish Imaging Network, A Platform for Scientific Excellence (SINAPSE) Collaboration, Edinburgh, UK
| | - Jackie F Price
- Usher Institute, University of Edinburgh, 5-7 Little France Road, Edinburgh, EH16 4UX, UK
| | - Kathryn L Evans
- Centre for Genomic and Experimental Medicine, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, EH4 2XU, UK
| | - Riccardo E Marioni
- Centre for Genomic and Experimental Medicine, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, EH4 2XU, UK.
| |
Collapse
|
20
|
Hagenbeek FA, Pool R, Van Asselt AJ, Ehli EA, Smit AB, Bartels M, Hottenga JJ, Dolan CV, van Dongen J, Boomsma DI. Intergenerational transmission of complex traits and the offspring methylome. Mol Psychiatry 2025:10.1038/s41380-025-02981-7. [PMID: 40181191 DOI: 10.1038/s41380-025-02981-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/17/2024] [Revised: 03/03/2025] [Accepted: 03/24/2025] [Indexed: 04/05/2025]
Abstract
The genetic makeup of parents can directly or indirectly affect their offspring phenome through genetic transmission or via the environment that is influenced by parental heritable traits. Our understanding of the mechanisms by which indirect genetic effects operate is limited. Here, we hypothesize that one mechanism is via the offspring methylome. To test this hypothesis, polygenic scores (PGSs) for schizophrenia, smoking initiation, educational attainment (EA), social deprivation, body mass index (BMI), and height were analyzed in a cohort of 1528 offspring and their parents (51.5% boys, mean [SD] age = 10 [2.8] years). We modelled parent and offspring PGSs on offspring buccal-DNA methylation, accounting for the own PGS of offspring, and found significant associations between parental PGSs for schizophrenia, EA, BMI, and height, and offspring buccal methylation sites, comprising 16, 2, 1, and 6 sites, respectively (alpha = 2.7 × 10-5). More DNA methylation sites were associated with maternal than paternal PGSs, possibly reflecting the maternal pre- and periconceptional environment or stronger maternal involvement in shaping the offspring's environment during early childhood.
Collapse
Affiliation(s)
- Fiona A Hagenbeek
- Department of Biological Psychology, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands.
- Amsterdam Public Health (APH) research institute, Amsterdam, The Netherlands.
- Institute for Molecular Medicine Finland (FIMM), HiLIFE, University of Helsinki, Helsinki, Finland.
| | - René Pool
- Department of Biological Psychology, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
- Amsterdam Public Health (APH) research institute, Amsterdam, The Netherlands
| | | | - Erik A Ehli
- Avera McKennan Hospital, University Health Center, Sioux Falls, SD, USA
| | - August B Smit
- Department of Molecular and Cellular Neurobiology, Center for Neurogenomics and Cognitive Research, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Meike Bartels
- Department of Biological Psychology, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
- Amsterdam Public Health (APH) research institute, Amsterdam, The Netherlands
| | - Jouke Jan Hottenga
- Department of Biological Psychology, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
- Amsterdam Public Health (APH) research institute, Amsterdam, The Netherlands
- Neurological Disorder Research Center, Qatar Biomedical Research Institute (QBRI), Hamad Bin Khalifa University (HBKU), Qatar Foundation, Doha, Qatar
| | - Conor V Dolan
- Department of Biological Psychology, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
- Amsterdam Public Health (APH) research institute, Amsterdam, The Netherlands
| | - Jenny van Dongen
- Department of Biological Psychology, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
- Amsterdam Public Health (APH) research institute, Amsterdam, The Netherlands
- Amsterdam Reproduction & Development (AR&D) research institute, Amsterdam, The Netherlands
| | - Dorret I Boomsma
- Department of Biological Psychology, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
- Amsterdam Public Health (APH) research institute, Amsterdam, The Netherlands
- Amsterdam Reproduction & Development (AR&D) research institute, Amsterdam, The Netherlands
- Department of Complex Trait Genetics, Center for Neurogenomics and Cognitive Research, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| |
Collapse
|
21
|
Bezerra OCL, Rodger M, Munsch G, Kovacs MJ, Le Gal G, Morange PE, Trégouët DA, Greenwood CMT, Gagnon F. Sex-specific DNA methylation marks associated with sex-biased risk of recurrence in unprovoked venous thromboembolism. J Thromb Haemost 2025; 23:1379-1392. [PMID: 39848545 DOI: 10.1016/j.jtha.2025.01.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2024] [Revised: 12/01/2024] [Accepted: 01/06/2025] [Indexed: 01/25/2025]
Abstract
BACKGROUND Whether to stop oral anticoagulants after a first unprovoked venous thromboembolism (VTE) is challenging, partially due to an intriguingly higher risk of VTE recurrence (rVTE) in men after therapy discontinuation. DNA methylation (DNAm) differences between men and women might underlie this sex-biased rVTE risk difference. OBJECTIVES To investigate sex-specific associations between DNAm at cytosine-phosphate-guanine (CpG) sites and rVTE. METHODS In 417 unprovoked VTE patients, including 101 experiencing recurrences over a 5-year follow-up (REcurrent VEnous thromboembolism Risk Stratification Evaluation [REVERSE] I), we analyzed blood DNAm using the Illumina EPIC array and performed a sex-stratified epigenome-wide association study. We further examined 181 major provoked VTE patients, including 36 recurrences over a 14-year follow-up (the MARseille THrombosis Association [MARTHA]), to investigate whether DNAm is a risk factor for rVTE after anticoagulation therapy. RESULTS Hypomethylated CpGs at genes TBC1D22B-cg01060850 and ZHX2-cg07808424 in men and DIP2B-ch.12.1038646R and DENND3-cg03401656 in women were associated with rVTE at genome-wide level (P < 7x10-8). Though not statistically significant, DENND3-cg03401656 had the same direction of effect in MARTHA women. Sensitivity analysis confirmed the robustness of the estimates, including potential confounders, adaptations of the Cox model, non-Europeans, and proximal methylation quantitative trait loci in the association. The associated CpGs were situated at genes for membrane trafficking, corroborating the participation of Rab regulatory proteins in rVTE and transcription factors. CONCLUSION We identified DNAm marks as potential risk factors for sex-biased recurrence in unprovoked VTE. Further replication and experimental validation could refine our understanding of the regulation of the identified DNAm sites and help optimize personalized decision-making for long-term anticoagulation after a first VTE.
Collapse
Affiliation(s)
- Ohanna C L Bezerra
- Dalla Lana School of Public Health, University of Toronto, Toronto, Ontario, Canada. https://twitter.com/ohannaclb
| | - Marc Rodger
- Department of Medicine, McGill University, Montreal, Quebec, Canada
| | - Gaëlle Munsch
- Bordeaux Population Health Research Center, Institut national de la santé et de la recherche médicale (INSERM), Unité Mixte de Recherche (UMR) 1219, F-33000, University of Bordeaux, Bordeaux, France
| | - Michael J Kovacs
- Department of Medicine, Western University, London, Ontario, Canada
| | - Grégoire Le Gal
- Department of Medicine, Ottawa Hospital Research Institute, University of Ottawa, Ottawa, Ontario, Canada
| | | | - David-Alexandre Trégouët
- Bordeaux Population Health Research Center, Institut national de la santé et de la recherche médicale (INSERM), Unité Mixte de Recherche (UMR) 1219, F-33000, University of Bordeaux, Bordeaux, France
| | - Celia M T Greenwood
- Lady Davis Institute for Medical Research, Jewish General Hospital, Montreal, Quebec, Canada; Department of Epidemiology, Biostatistics and Occupational Health, McGill University, Montreal, Quebec, Canada
| | - France Gagnon
- Dalla Lana School of Public Health, University of Toronto, Toronto, Ontario, Canada; Office of the Vice-Principal of Research and Innovation, University of Toronto Mississauga, Mississauga, Ontario, Canada.
| |
Collapse
|
22
|
Li X, Lin Q, Guan B, Yang M, Huang X, Li L, Chen C, Hong J, Zhang M. Multi-Omics Analysis Links Mitochondrial-Related Genes to Idiopathic Pulmonary Fibrosis and In Vivo Transcriptome Validation. Transl Res 2025; 278:10-21. [PMID: 39952317 DOI: 10.1016/j.trsl.2025.02.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/06/2025] [Revised: 02/07/2025] [Accepted: 02/10/2025] [Indexed: 02/17/2025]
Abstract
Mitochondrial dysfunction is closely associated with idiopathic pulmonary fibrosis (IPF). However, the causal association between mitochondria-related genes and IPF remains to be determined. We obtained gene expression, protein abundance, and methylation quantitative trait locus data for mitochondria-related genes from previous studies. Genome-wide association study data for patients with IPF were obtained from the FinnGen study. A two-sample Mendelian randomisation analysis was conducted to assess the association between mitochondria-related genes and IPF. Furthermore, we conducted validation of expression differences utilizing transcriptome data derived from the BLM-induced pulmonary fibrosis mouse model (n=15). Concurrently, multiphoton imaging was utilized to quantify collagen contents and structural assessment. The direction of causality was verified using the Steiger test, and colocalisation analysis was used to better validate causality. Single-cell data were used to explore the localisation and expression of positive genes across different cell types. The study identified significant associations between mitochondria-related genes and IPF, with POLG and NDUFB10 classified as Grade 1; LYRM4, NBR1, and ACSF3 as Grade 2; MCL1, GFER, MFN2, IVD, and SLC25A35 as Grade 3; and METAP1D and MTX1 as Grade 4. Single-cell analysis showed elevated expression of NBR1, MCL1, and MTX1 in pulmonary myofibroblasts of IPF. This study elucidated the causal effects of mitochondria-related genes on IPF, underscoring their significance in pathogenesis. These findings contribute to an improved understanding of the mechanisms underlying IPF, offering new potential therapeutic targets for interventions.
Collapse
Affiliation(s)
- Xiaoxia Li
- Department of Radiotherapy, Cancer Center, the First Affiliated Hospital of Fujian Medical University, Fuzhou, PR China; Department of Radiotherapy, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital of Fujian Medical University, Fuzhou, PR China
| | - Qiaojing Lin
- Department of Radiotherapy, Cancer Center, the First Affiliated Hospital of Fujian Medical University, Fuzhou, PR China; Department of Radiotherapy, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital of Fujian Medical University, Fuzhou, PR China
| | - Bingyue Guan
- Department of Radiotherapy, Cancer Center, the First Affiliated Hospital of Fujian Medical University, Fuzhou, PR China; Department of Radiotherapy, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital of Fujian Medical University, Fuzhou, PR China
| | - Minghuan Yang
- School of Public Health, Fujian Medical University, Fuzhou, PR China
| | - Xingxin Huang
- School of Arts and Sciences, Fujian Medical University, Fuzhou, PR China
| | - Lianhuang Li
- Key Laboratory of OptoElectronic Science and Technology for Medicine of Ministry of Education, Fujian Provincial Key Laboratory of Photonics Technology, Fujian Normal University, Fuzhou, PR China.
| | - Chun Chen
- Key Laboratory of Radiation Biology of Fujian Higher Education Institutions, the First Affiliated Hospital, Fujian Medical University, Fuzhou, PR China; School of Pharmacy, Fujian Medical University, Fuzhou, PR China.
| | - Jinsheng Hong
- Department of Radiotherapy, Cancer Center, the First Affiliated Hospital of Fujian Medical University, Fuzhou, PR China; Department of Radiotherapy, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital of Fujian Medical University, Fuzhou, PR China; Key Laboratory of Radiation Biology of Fujian Higher Education Institutions, the First Affiliated Hospital, Fujian Medical University, Fuzhou, PR China; School of Medical Imaging, Fujian Medical University, Fuzhou, PR China.
| | - Mingwei Zhang
- Department of Radiotherapy, Cancer Center, the First Affiliated Hospital of Fujian Medical University, Fuzhou, PR China; Department of Radiotherapy, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital of Fujian Medical University, Fuzhou, PR China; Key Laboratory of Radiation Biology of Fujian Higher Education Institutions, the First Affiliated Hospital, Fujian Medical University, Fuzhou, PR China; School of Medical Imaging, Fujian Medical University, Fuzhou, PR China.
| |
Collapse
|
23
|
Golub Y, Wulff A, Plösch T. From haze to horizon: epigenetic research and artificial intelligence in child and adolescent psychiatry. Eur Child Adolesc Psychiatry 2025; 34:1245-1248. [PMID: 40111558 PMCID: PMC12000212 DOI: 10.1007/s00787-025-02686-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/17/2025] [Accepted: 02/18/2025] [Indexed: 03/22/2025]
Affiliation(s)
- Yulia Golub
- Department of Child and Adolescent Psychiatry, Psychosomatic and Psychotherapy, School of Medicine and Health Sciences, Carl von Ossietzky Universität Oldenburg, Oldenburg, Germany.
| | - Antje Wulff
- Big Data in Medicine, Department of Health Services Research, Carl von Ossietzky Universität Oldenburg, Oldenburg, Germany
| | - Torsten Plösch
- Department of Human Medicine, Division of Perinatal Neurobiology, School of Medicine and Health Science, Carl von Ossietzky Universität Oldenburg, Oldenburg, Germany
- Department of Obstetrics and Gynaecology, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| |
Collapse
|
24
|
Schmitz LL, Opsasnick LA, Ratliff SM, Faul JD, Zhao W, Hughes TM, Ding J, Liu Y, Smith JA. Epigenetic biomarkers of socioeconomic status are associated with age-related chronic diseases and mortality in older adults. PNAS NEXUS 2025; 4:pgaf121. [PMID: 40309465 PMCID: PMC12041747 DOI: 10.1093/pnasnexus/pgaf121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/21/2024] [Accepted: 03/26/2025] [Indexed: 05/02/2025]
Abstract
Later-life health is patterned by socioeconomic influences across the lifecourse. However, the pathways underlying the biological embedding of socioeconomic status (SES) and its consequences on downstream morbidity and mortality are not fully understood. Epigenetic markers like DNA methylation (DNAm) may be promising surrogates of underlying biological processes that can enhance our understanding of how SES shapes population health. Studies have shown that SES is associated with epigenetic aging measures, but few have examined relationships between early and later-life SES and DNAm sites across the epigenome. In this study, we trained and tested DNAm-based surrogates, or "biomarkers," of childhood and adult SES in two large, multiracial/ethnic samples of older adults-the Health and Retirement Study (n = 3,527) and the Multi-Ethnic Study of Atherosclerosis (n = 1,182). Both biomarkers were associated with downstream morbidity and mortality, and these associations persisted after controlling for measured SES, and in some cases, epigenetic aging clocks. Both childhood and adult SES biomarker CpG sites were enriched for genomic features that regulate gene expression (e.g. DNAse hypersensitivity sites and enhancers) and were implicated in prior epigenome-wide studies of inflammation, aging, and chronic disease. Distinct patterns also emerged between childhood CpGs and immune system dysregulation and adult CpGs and metabolic functioning, health behaviors, and cancer. Results suggest DNAm-based surrogate biomarkers of SES may be useful proxies for unmeasured social exposures that can augment our understanding of the biological mechanisms between social disadvantage and downstream health.
Collapse
Affiliation(s)
- Lauren L Schmitz
- Robert M. La Follette School of Public Affairs, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Lauren A Opsasnick
- Department of Epidemiology, School of Public Health, University of Michigan, Ann Arbor, MI 48109, USA
| | - Scott M Ratliff
- Department of Epidemiology, School of Public Health, University of Michigan, Ann Arbor, MI 48109, USA
| | - Jessica D Faul
- Survey Research Center, Institute for Social Research, University of Michigan, Ann Arbor, MI 48104, USA
| | - Wei Zhao
- Department of Epidemiology, School of Public Health, University of Michigan, Ann Arbor, MI 48109, USA
- Survey Research Center, Institute for Social Research, University of Michigan, Ann Arbor, MI 48104, USA
| | - Timothy M Hughes
- Department of Gerontology and Geriatric Medicine, School of Medicine, Wake Forest University, Winston-Salem, NC 27157, USA
| | - Jingzhong Ding
- Department of Gerontology and Geriatric Medicine, School of Medicine, Wake Forest University, Winston-Salem, NC 27157, USA
| | - Yongmei Liu
- Department of Medicine, Divisions of Cardiology and Neurology, Duke University Medical Center, Durham, NC 27710, USA
| | - Jennifer A Smith
- Department of Epidemiology, School of Public Health, University of Michigan, Ann Arbor, MI 48109, USA
- Survey Research Center, Institute for Social Research, University of Michigan, Ann Arbor, MI 48104, USA
| |
Collapse
|
25
|
Wang D, Li D, Dang X, Mu C, Liu C, Zeng Y, Yuan Y, Teng Z, Li Y, Luo XJ. Mendelian Randomization Reveals Causalities Between DNA Methylation and Schizophrenia. Biol Psychiatry 2025:S0006-3223(25)01100-X. [PMID: 40157589 DOI: 10.1016/j.biopsych.2025.03.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/07/2024] [Revised: 03/06/2025] [Accepted: 03/22/2025] [Indexed: 04/01/2025]
Abstract
BACKGROUND Epigenetic factors (such as DNA methylation) have been widely reported to be associated with schizophrenia (SCZ). However, the causal relationships between epigenetic factors and SCZ remain largely unknown. METHODS Here, we conducted a Mendelian randomization (MR) study to investigate the causal relationships between DNA methylation and SCZ. Brain methylation quantitative trait loci (mQTL) (N = 1160) and blood mQTL (N = 27,750) data were used as exposures, and genome-wide association data of SCZ (53,386 cases and 77,258 controls) were used as the outcome. RESULTS We identified 172 (mapped to 160 genes) and 157 (mapped to 155 genes) methylation sites whose methylation levels in brain and blood are causally associated with SCZ, respectively. Among the mapped genes, 36 overlapping genes were identified. Interestingly, 3 methylation sites (near BRD2, CNNM2, and RERE) showed significant associations in both brain and blood, with the same direction of effect. We also performed MR analysis using brain expression quantitative trait loci (eQTLs) as exposures and identified 123 genes whose expression levels were causally associated with SCZ. Comparing the significant genes from eQTLs and brain mQTLs prioritized 15 overlapping genes, suggesting that both epigenetic modification and expression of these genes confer risk of SCZ. Finally, we validated our findings with genome editing and animal model experiments. CONCLUSIONS Our study identified methylation sites whose methylation levels are causally associated with SCZ and demonstrated the important roles of epigenetic factors in SCZ. Our findings also reveal pivotal risk genes whose expression and epigenetic regulation are causally associated with SCZ.
Collapse
Affiliation(s)
- Danni Wang
- State Key Laboratory of Digital Medical Engineering, Department of Psychiatry and Psychosomatics, Zhongda Hospital, School of Medicine, Advanced Institute for Life and Health, Jiangsu Provincial Key Laboratory of Brain Science and Medicine, School of Life Science and Technology, Southeast University, Nanjing, Jiangsu, China
| | - Danyang Li
- The Second Affiliated Hospital of Kunming Medical University, Key Laboratory of Neurological and Psychiatric Disease Research of Yunnan Province, Yunnan Provincial Department of Education Gut Microbiota Transplantation Engineering Research Center, Kunming, China
| | - Xinglun Dang
- State Key Laboratory of Digital Medical Engineering, Department of Psychiatry and Psychosomatics, Zhongda Hospital, School of Medicine, Advanced Institute for Life and Health, Jiangsu Provincial Key Laboratory of Brain Science and Medicine, School of Life Science and Technology, Southeast University, Nanjing, Jiangsu, China
| | - Changgai Mu
- State Key Laboratory of Digital Medical Engineering, Department of Psychiatry and Psychosomatics, Zhongda Hospital, School of Medicine, Advanced Institute for Life and Health, Jiangsu Provincial Key Laboratory of Brain Science and Medicine, School of Life Science and Technology, Southeast University, Nanjing, Jiangsu, China
| | - Chang Liu
- State Key Laboratory of Digital Medical Engineering, Department of Psychiatry and Psychosomatics, Zhongda Hospital, School of Medicine, Advanced Institute for Life and Health, Jiangsu Provincial Key Laboratory of Brain Science and Medicine, School of Life Science and Technology, Southeast University, Nanjing, Jiangsu, China
| | - Yong Zeng
- The Second Affiliated Hospital of Kunming Medical University, Key Laboratory of Neurological and Psychiatric Disease Research of Yunnan Province, Yunnan Provincial Department of Education Gut Microbiota Transplantation Engineering Research Center, Kunming, China
| | - Yonggui Yuan
- State Key Laboratory of Digital Medical Engineering, Department of Psychiatry and Psychosomatics, Zhongda Hospital, School of Medicine, Advanced Institute for Life and Health, Jiangsu Provincial Key Laboratory of Brain Science and Medicine, School of Life Science and Technology, Southeast University, Nanjing, Jiangsu, China
| | - Zhaowei Teng
- The Second Affiliated Hospital of Kunming Medical University, Key Laboratory of Neurological and Psychiatric Disease Research of Yunnan Province, Yunnan Provincial Department of Education Gut Microbiota Transplantation Engineering Research Center, Kunming, China.
| | - Yifan Li
- State Key Laboratory of Digital Medical Engineering, Department of Psychiatry and Psychosomatics, Zhongda Hospital, School of Medicine, Advanced Institute for Life and Health, Jiangsu Provincial Key Laboratory of Brain Science and Medicine, School of Life Science and Technology, Southeast University, Nanjing, Jiangsu, China.
| | - Xiong-Jian Luo
- State Key Laboratory of Digital Medical Engineering, Department of Psychiatry and Psychosomatics, Zhongda Hospital, School of Medicine, Advanced Institute for Life and Health, Jiangsu Provincial Key Laboratory of Brain Science and Medicine, School of Life Science and Technology, Southeast University, Nanjing, Jiangsu, China.
| |
Collapse
|
26
|
Cilleros-Portet A, Lesseur C, Marí S, Cosin-Tomas M, Lozano M, Irizar A, Burt A, García-Santisteban I, Garrido-Martín D, Escaramís G, Hernangomez-Laderas A, Soler-Blasco R, Breeze CE, Gonzalez-Garcia BP, Santa-Marina L, Chen J, Llop S, Fernández MF, Vrijheid M, Ibarluzea J, Guxens M, Marsit C, Bustamante M, Bilbao JR, Fernandez-Jimenez N. Potentially causal associations between placental DNA methylation and schizophrenia and other neuropsychiatric disorders. Nat Commun 2025; 16:2431. [PMID: 40087310 PMCID: PMC11909199 DOI: 10.1038/s41467-025-57760-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Accepted: 02/26/2025] [Indexed: 03/17/2025] Open
Abstract
Increasing evidence supports the role of the placenta in neurodevelopment and in the onset of neuropsychiatric disorders. Recently, mQTL and iQTL maps have proven useful in understanding relationships between SNPs and GWAS that are not captured by eQTL. In this context, we propose that part of the genetic predisposition to complex neuropsychiatric disorders acts through placental DNA methylation. We construct a public placental cis-mQTL database including 214,830 CpG sites calculated in 368 fetal placenta DNA samples from the INMA project, and run cell type-, gestational age- and sex-imQTL models. We combine these data with summary statistics of GWAS on ten neuropsychiatric disorders using summary-based Mendelian randomization and colocalization. We also evaluate the influence of identified DNA methylation sites on placental gene expression in the RICHS cohort. We find that placental cis-mQTLs are enriched in placenta-specific active chromatin regions, and establish that part of the genetic burden for schizophrenia, bipolar disorder, and major depressive disorder confers risk through placental DNA methylation. The potential causality of several of the observed associations is reinforced by secondary association signals identified in conditional analyses, the involvement of cell type-imQTLs, and the correlation of identified DNA methylation sites with the expression levels of relevant genes in the placenta.
Collapse
Affiliation(s)
- Ariadna Cilleros-Portet
- Department of Genetics, Physical Anthropology and Animal Physiology, Biobizkaia Health Research Institute and University of the Basque Country (UPV/EHU), Leioa, Spain
| | - Corina Lesseur
- Department of Environmental Medicine and Public Health, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Sergi Marí
- Department of Genetics, Physical Anthropology and Animal Physiology, Biobizkaia Health Research Institute and University of the Basque Country (UPV/EHU), Leioa, Spain
| | - Marta Cosin-Tomas
- ISGlobal, Barcelona, Spain
- Spanish Consortium for Research on Epidemiology and Public Health (CIBERESP), Instituto de Salud Carlos III, Madrid, Spain
- Universitat Pompeu Fabra, Barcelona, Spain
| | - Manuel Lozano
- Epidemiology and Environmental Health Joint Research Unit, FISABIO-Universitat Jaume I-Universitat de València, Valencia, Spain
- Preventive Medicine and Public Health, Food Sciences, Toxicology and Forensic Medicine Department, Universitat de València, Valencia, Spain
| | - Amaia Irizar
- Spanish Consortium for Research on Epidemiology and Public Health (CIBERESP), Instituto de Salud Carlos III, Madrid, Spain
- Department of Preventive Medicine and Public Health, University of the Basque Country (UPV/EHU), Leioa, Spain
- Biogipuzkoa Health Research Institute, San Sebastian, Spain
| | - Amber Burt
- Gangarosa Department of Environmental Health, Rollins School of Public Health, Emory University, Atlanta, GA, USA
| | - Iraia García-Santisteban
- Department of Genetics, Physical Anthropology and Animal Physiology, Biobizkaia Health Research Institute and University of the Basque Country (UPV/EHU), Leioa, Spain
| | - Diego Garrido-Martín
- Department of Genetics, Microbiology and Statistics, Faculty of Biology, Universitat de Barcelona (UB), Barcelona, Spain
| | - Geòrgia Escaramís
- Spanish Consortium for Research on Epidemiology and Public Health (CIBERESP), Instituto de Salud Carlos III, Madrid, Spain
- Departament de Biomedicina, Facultat de Medicina i Ciències de la Salut, Institut de Neurociències, Universitat de Barcelona, Barcelona, Spain
| | - Alba Hernangomez-Laderas
- Department of Genetics, Physical Anthropology and Animal Physiology, Biobizkaia Health Research Institute and University of the Basque Country (UPV/EHU), Leioa, Spain
| | - Raquel Soler-Blasco
- Spanish Consortium for Research on Epidemiology and Public Health (CIBERESP), Instituto de Salud Carlos III, Madrid, Spain
- Epidemiology and Environmental Health Joint Research Unit, FISABIO-Universitat Jaume I-Universitat de València, Valencia, Spain
- Department of Nursing, Universitat de València, Valencia, Spain
| | | | - Bárbara P Gonzalez-Garcia
- Department of Genetics, Physical Anthropology and Animal Physiology, Biobizkaia Health Research Institute and University of the Basque Country (UPV/EHU), Leioa, Spain
| | - Loreto Santa-Marina
- Spanish Consortium for Research on Epidemiology and Public Health (CIBERESP), Instituto de Salud Carlos III, Madrid, Spain
- Biogipuzkoa Health Research Institute, San Sebastian, Spain
- Department of Health of the Basque Government, Subdirectorate of Public Health of Gipuzkoa, San Sebastian, Spain
| | - Jia Chen
- Department of Environmental Medicine and Public Health, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Sabrina Llop
- Spanish Consortium for Research on Epidemiology and Public Health (CIBERESP), Instituto de Salud Carlos III, Madrid, Spain
- Epidemiology and Environmental Health Joint Research Unit, FISABIO-Universitat Jaume I-Universitat de València, Valencia, Spain
| | - Mariana F Fernández
- Spanish Consortium for Research on Epidemiology and Public Health (CIBERESP), Instituto de Salud Carlos III, Madrid, Spain
- Department of Radiology and Physical Medicine, Biomedical Research Center (CIBM), School of Medicine, University of Granada, Granada, Spain
- Instituto de Investigación Biosanitaria de Granada (ibs.GRANADA), Granada, Spain
| | - Martine Vrijheid
- ISGlobal, Barcelona, Spain
- Spanish Consortium for Research on Epidemiology and Public Health (CIBERESP), Instituto de Salud Carlos III, Madrid, Spain
- Universitat Pompeu Fabra, Barcelona, Spain
| | - Jesús Ibarluzea
- Spanish Consortium for Research on Epidemiology and Public Health (CIBERESP), Instituto de Salud Carlos III, Madrid, Spain
- Biogipuzkoa Health Research Institute, San Sebastian, Spain
- Department of Health of the Basque Government, Subdirectorate of Public Health of Gipuzkoa, San Sebastian, Spain
| | - Mònica Guxens
- ISGlobal, Barcelona, Spain
- Spanish Consortium for Research on Epidemiology and Public Health (CIBERESP), Instituto de Salud Carlos III, Madrid, Spain
- Universitat Pompeu Fabra, Barcelona, Spain
- Department of Child and Adolescent Psychiatry/Psychology, Erasmus MC, University Medical Centre, Rotterdam, The Netherlands
- ICREA, Barcelona, Spain
| | - Carmen Marsit
- Gangarosa Department of Environmental Health, Rollins School of Public Health, Emory University, Atlanta, GA, USA
| | - Mariona Bustamante
- ISGlobal, Barcelona, Spain
- Spanish Consortium for Research on Epidemiology and Public Health (CIBERESP), Instituto de Salud Carlos III, Madrid, Spain
- Universitat Pompeu Fabra, Barcelona, Spain
| | - Jose Ramon Bilbao
- Department of Genetics, Physical Anthropology and Animal Physiology, Biobizkaia Health Research Institute and University of the Basque Country (UPV/EHU), Leioa, Spain
- CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Madrid, Spain
| | - Nora Fernandez-Jimenez
- Department of Genetics, Physical Anthropology and Animal Physiology, Biobizkaia Health Research Institute and University of the Basque Country (UPV/EHU), Leioa, Spain.
| |
Collapse
|
27
|
Zakarya R, Chan YL, Wang B, Thorpe A, Xenaki D, Ho KF, Guo H, Chen H, Oliver BG, O'Neill C. Developmental air pollution exposure augments airway hyperreactivity, alters transcriptome, and DNA methylation in female adult progeny. Commun Biol 2025; 8:400. [PMID: 40057553 PMCID: PMC11890619 DOI: 10.1038/s42003-025-07835-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2024] [Accepted: 02/26/2025] [Indexed: 05/13/2025] Open
Abstract
Maternal exposure to particulate air pollution increases the incidence and severity of asthma in offspring, yet the mechanisms for this are unclear. Known susceptibility loci are a minor component of this effect. We interrogate a mouse allergic airway disease model to assess epigenetic associations between maternal air pollution exposure and asthma responses in offspring. Maternal air pollution exposure increased allergic airway disease severity in adult offspring associated with a suppressed transcriptomic response. Control progeny showed differential expression of 2842 genes across several important pathways, whilst air pollutant progeny showed an 80% reduction in differentially expressed genes and abrogation of many pathway associations. Whole genome CpG methylome analysis following allergen challenge detected differential methylation regions across the genome. Differentially methylated regions were markedly reduced in air pollutant offspring, and this was most evident in intronic regions and some transposable element classes. This study shows that asthma in adult offspring of PM2.5 exposed mothers had a markedly repressed transcriptomic response, a proportion of which was associated with identifiable changes in the lung's methylome. The results point to an epigenetic contribution to the severity of asthma in offspring of mothers exposed to particulate air pollution.
Collapse
Affiliation(s)
- Razia Zakarya
- School of Life Sciences, University of Technology Sydney, Sydney, Australia.
- Epigenetics of Chronic Disease Group, Woolcock Institute of Medical Research, Macquarie University, Sydney, Australia.
| | - Yik Lung Chan
- School of Life Sciences, University of Technology Sydney, Sydney, Australia
- Respiratory Cell and Molecular Biology Group, Woolcock Institute of Medical Research, Macquarie University, Sydney, Australia
| | - Baoming Wang
- School of Life Sciences, University of Technology Sydney, Sydney, Australia
- Respiratory Cell and Molecular Biology Group, Woolcock Institute of Medical Research, Macquarie University, Sydney, Australia
| | - Andrew Thorpe
- School of Life Sciences, University of Technology Sydney, Sydney, Australia
- Respiratory Cell and Molecular Biology Group, Woolcock Institute of Medical Research, Macquarie University, Sydney, Australia
| | - Dikaia Xenaki
- Respiratory Cell and Molecular Biology Group, Woolcock Institute of Medical Research, Macquarie University, Sydney, Australia
| | - Kin Fai Ho
- Jockey Club School of Public Health and Primary, The Chinese University of Hong Kong, Hong Kong Special Administrative Region of the People's Republic of China, Hong Kong, China
| | - Hai Guo
- Air Quality Studies, Department of Civil and Environmental Engineering, The Hong Kong Polytechnic University, Hong Kong, China
| | - Hui Chen
- School of Life Sciences, University of Technology Sydney, Sydney, Australia
| | - Brian G Oliver
- School of Life Sciences, University of Technology Sydney, Sydney, Australia.
- Respiratory Cell and Molecular Biology Group, Woolcock Institute of Medical Research, Macquarie University, Sydney, Australia.
| | - Christopher O'Neill
- School of Life Sciences, University of Technology Sydney, Sydney, Australia.
- Epigenetics of Chronic Disease Group, Woolcock Institute of Medical Research, Macquarie University, Sydney, Australia.
| |
Collapse
|
28
|
Zheng Z, Song Y, Li X, Luo T, Tan X. Dissecting the causal effects of smoking, alcohol consumption, and related DNA methylation markers on electrocardiographic indices. Clin Epigenetics 2025; 17:40. [PMID: 40038836 PMCID: PMC11881420 DOI: 10.1186/s13148-025-01851-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2024] [Accepted: 02/19/2025] [Indexed: 03/06/2025] Open
Abstract
BACKGROUND Tobacco and alcohol are recognized risk factors for heart disease, yet their causal effects on electrocardiogram (ECG) signaling and mechanisms remain unclear. Previous studies may be susceptible to confounding or bias, and this study dissected the genetic architecture linking tobacco and alcohol consumption with P-wave duration, PR interval, and QT interval. METHODS Utilizing genetic instruments for tobacco and alcohol consumption, associated methylation quantitative trait locus (mQTL), and summary-level GWAS data for ECG indices, we assessed heritability and genetic causal associations using linkage disequilibrium score regression and Mendelian randomization (MR) analysis. Fine mapping was performed via colocalization analysis and summary-data-based MR (SMR) to identify potential shared genetic variants. RESULTS A positive causal relationship was found between drinks per week (DrnkWk) and QT interval [β (95%CI): 1.06 (0.91, 5.05), P = 0.005], with causality substantiated through multiple robust MR models. Multivariable MR confirmed independence from smoking phenotypes. In epigenetic MR analyses, two alcohol-related CpG loci (cg03345232 and cg04605617) were causally associated with QT interval changes, with cg04605617 mapping to PLA2G2C gene significantly prolonging QT. The mQTL rs10916683 at cg04605617 is a strong eQTL for PLA2G2C. Additionally, cg03345232 shared a causal variant (rs12881206) with QT interval predisposition through colocalization analysis. SMR analysis did not identify shared putative functional genes passing the HEIDI test between DrnkWk and the QT interval. CONCLUSIONS There is a causal relationship between DrnkWk and QT interval prolongation, and targeting specific DNA methylation sites like cg04605617 mapped to PLA2G2C may provide novel targets for preventing QT interval prolongation.
Collapse
Affiliation(s)
- Zequn Zheng
- Department of Cardiology, First Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong, China
- Clinical Research Center, First Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong, China
- Institute of Innovation for Combined Medicine and Engineering, Ningbo Medical Center Lihuili Hospital, Ningbo University, Ningbo, Zhejiang, China
- Human Phenome institute of SUMC, Guangdong Engineering Research Center of Human Phenome, Chemistry and Chemical Engineering Guangdong Laboratory, Shantou, Guangdong, China
| | - Yongfei Song
- Institute of Innovation for Combined Medicine and Engineering, Ningbo Medical Center Lihuili Hospital, Ningbo University, Ningbo, Zhejiang, China
- Center for Molecular Medicine, Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Xinhan Li
- Department of Cardiology, Taizhou Hospital of Zhejiang Province affiliated to Wenzhou Medical University, Taizhou, Zhejiang, China
| | - Tao Luo
- Center of Stem Cell and Regenerative Medicine, and Bone Marrow Transplantation Center of the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Xuerui Tan
- Department of Cardiology, First Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong, China.
- Clinical Research Center, First Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong, China.
- Human Phenome institute of SUMC, Guangdong Engineering Research Center of Human Phenome, Chemistry and Chemical Engineering Guangdong Laboratory, Shantou, Guangdong, China.
| |
Collapse
|
29
|
Zhang W, Young JI, Gomez L, Schmidt MA, Lukacsovich D, Kunkle BW, Chen XS, Martin ER, Wang L. Blood DNA methylation signature for incident dementia: Evidence from longitudinal cohorts. Alzheimers Dement 2025; 21:e14496. [PMID: 40133250 PMCID: PMC11936765 DOI: 10.1002/alz.14496] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2024] [Revised: 11/26/2024] [Accepted: 12/01/2024] [Indexed: 03/27/2025]
Abstract
INTRODUCTION Distinguishing between molecular changes that precede dementia onset and those resulting from the disease is challenging with cross-sectional studies. METHODS We studied blood DNA methylation (DNAm) differences and incident dementia in two large longitudinal cohorts: the Offspring cohort of the Framingham Heart Study (FHS) and the Alzheimer's Disease Neuroimaging Initiative (ADNI) study. We analyzed blood DNAm samples from > 1000 cognitively unimpaired subjects. RESULTS Meta-analysis identified 44 CpGs and 44 differentially methylated regions consistently associated with time to dementia in both cohorts. Our integrative analysis identified early processes in dementia, such as immune responses and metabolic dysfunction. Furthermore, we developed a methylation-based risk score, which successfully predicted future cognitive decline in an independent validation set, even after accounting for age, sex, apolipoprotein E ε4, years of education, baseline diagnosis, and baseline Mini-Mental State Examination score. DISCUSSION DNAm offers a promising source as a biomarker for dementia risk assessment. HIGHLIGHTS Blood DNA methylation (DNAm) differences at individual CpGs and differentially methylated regions are significantly associated with incident dementia. Pathway analysis revealed DNAm differences associated with incident dementia are significantly enriched in biological pathways involved in immune responses and metabolic processes. Out-of-sample validation analysis demonstrated that a methylation-based risk score successfully predicted future cognitive decline in an independent dataset, even after accounting for age, sex, apolipoprotein E ε4, years of education, baseline diagnosis, and baseline Mini-Mental State Examination score.
Collapse
Affiliation(s)
- Wei Zhang
- Division of BiostatisticsDepartment of Public Health SciencesMiller School of MedicineUniversity of MiamiMiamiFloridaUSA
| | - Juan I. Young
- Dr. John T. Macdonald Foundation Department of Human GeneticsMiller School of MedicineUniversity of MiamiMiamiFloridaUSA
- John P. Hussman Institute for Human GenomicsMiller School of MedicineUniversity of MiamiMiamiFloridaUSA
| | - Lissette Gomez
- John P. Hussman Institute for Human GenomicsMiller School of MedicineUniversity of MiamiMiamiFloridaUSA
| | - Michael A. Schmidt
- Dr. John T. Macdonald Foundation Department of Human GeneticsMiller School of MedicineUniversity of MiamiMiamiFloridaUSA
- John P. Hussman Institute for Human GenomicsMiller School of MedicineUniversity of MiamiMiamiFloridaUSA
| | - David Lukacsovich
- Division of BiostatisticsDepartment of Public Health SciencesMiller School of MedicineUniversity of MiamiMiamiFloridaUSA
| | - Brian W. Kunkle
- Dr. John T. Macdonald Foundation Department of Human GeneticsMiller School of MedicineUniversity of MiamiMiamiFloridaUSA
- John P. Hussman Institute for Human GenomicsMiller School of MedicineUniversity of MiamiMiamiFloridaUSA
| | - X. Steven Chen
- Division of BiostatisticsDepartment of Public Health SciencesMiller School of MedicineUniversity of MiamiMiamiFloridaUSA
- Sylvester Comprehensive Cancer CenterMiller School of MedicineUniversity of MiamiMiamiFloridaUSA
| | - Eden R. Martin
- Dr. John T. Macdonald Foundation Department of Human GeneticsMiller School of MedicineUniversity of MiamiMiamiFloridaUSA
- John P. Hussman Institute for Human GenomicsMiller School of MedicineUniversity of MiamiMiamiFloridaUSA
| | - Lily Wang
- Division of BiostatisticsDepartment of Public Health SciencesMiller School of MedicineUniversity of MiamiMiamiFloridaUSA
- Dr. John T. Macdonald Foundation Department of Human GeneticsMiller School of MedicineUniversity of MiamiMiamiFloridaUSA
- John P. Hussman Institute for Human GenomicsMiller School of MedicineUniversity of MiamiMiamiFloridaUSA
- Sylvester Comprehensive Cancer CenterMiller School of MedicineUniversity of MiamiMiamiFloridaUSA
| |
Collapse
|
30
|
Xiong L, Zhang H, Guo Y, Tao Y. Identification of Potential Drug Targets for Immunoglobulin A Nephropathy: A Mendelian Randomization Study. Biomedicines 2025; 13:581. [PMID: 40149558 PMCID: PMC11940645 DOI: 10.3390/biomedicines13030581] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2025] [Revised: 02/14/2025] [Accepted: 02/21/2025] [Indexed: 03/29/2025] Open
Abstract
Background: The current pharmacological treatments for Immunoglobulin A nephropathy (IgAN) demonstrate limited effectiveness and may cause serious side effects. This study aimed to explore novel potential drug targets for IgAN. Methods: We utilized summarized data from a recent genome-wide association study on IgAN, cis-expression quantitative trait loci data for druggable genes obtained from the eQTLGen Consortium, and DNA methylation quantitative trait loci data derived from the GoDMC database. Two-sample Mendelian randomization (MR) analysis, Bayesian colocalization, and mediation analysis through a two-step MR approach were performed to investigate their causal relationships. Results: Two-sample MR and colocalization analyses demonstrated that the expression of HLA-DPA1 and C4A was associated with an increased risk of IgAN. In contrast, TUBB, CYP21A2, and C4B were associated with a decreased risk of IgAN. Mediation analysis revealed that the expression of HLA-DPA1 acted as a mediator in the potential causal relationship between three DNA methylation sites (cg01140143, cg08898074, and cg12168509) and IgAN, with mediated proportions of 33.74% (95% CI 1.64-73.27), 41.67% (95% CI 20.78-66.97), and 50.34% (95% CI 27.89-74.76), respectively. Conclusions: Several druggable genes and DNA methylation sites were identified to show potential causal associations with IgAN risk and may be targeted for drug development. Nevertheless, additional experimental validation is warranted to clarify the specific roles of DNA methylation and the identified druggable genes in the pathogenesis of IgAN.
Collapse
Affiliation(s)
- Limei Xiong
- Division of Nephrology, Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu 610041, China; (L.X.)
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Chengdu 610041, China
| | - Hui Zhang
- Division of Nephrology, Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu 610041, China; (L.X.)
| | - Yannan Guo
- Division of Nephrology, Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu 610041, China; (L.X.)
| | - Yuhong Tao
- Division of Nephrology, Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu 610041, China; (L.X.)
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Chengdu 610041, China
| |
Collapse
|
31
|
Machado-Paula LA, Romanowska J, Lie RT, Hovey L, Doolittle B, Awotoye W, Dunlay L, Xie XJ, Zeng E, Butali A, Marazita ML, Murray JC, Moreno-Uribe LM, Petrin AL. Genetic-epigenetic interactions (meQTLs) in orofacial clefts etiology. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2025:2025.02.09.25321494. [PMID: 39990564 PMCID: PMC11844571 DOI: 10.1101/2025.02.09.25321494] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 02/25/2025]
Abstract
Objectives Nonsyndromic orofacial clefts (OFCs) etiology involves multiple genetic and environmental factors with over 60 identified risk loci; however, they account for only a minority of the estimated risk. Epigenetic factors such as differential DNA methylation (DNAm) are also associated with OFCs risk and can alter risk for different cleft types and modify OFCs penetrance. DNAm is a covalent addition of a methyl (CH3) group to the nucleotide cytosine that can lead to changes in expression of the targeted gene. DNAm can be affected by environmental influences and genetic variation via methylation quantitative loci (meQTLs). We hypothesize that aberrant DNAm and the resulting alterations in gene expression play a key role in the etiology of OFCs, and that certain common genetic variants that affect OFCs risk do so by influencing DNAm. Methods We used genotype from 10 cleft-associated SNPs and genome-wide DNA methylation data (Illumina 450K array) for 409 cases with OFCs and 456 controls and identified 23 cleft-associated meQTLs. We then used an independent cohort of 362 cleft-discordant sib pairs for replication. We used methylation-specific qPCR to measure methylation levels of each CpG site and combined genotypic and methylation data for an interaction analysis of each SNP-CpG pair using the R package MatrixeQTL in a linear model. We also performed a Paired T-test to analyze differences in DNA methylation between each member of the sibling pairs. Results We replicated 9 meQTLs, showing interactions between rs13041247 (MAFB) - cg18347630 (PLCG1) (P=0.04); rs227731 (NOG) - cg08592707 (PPM1E) (P=0.01); rs227731 (NOG) - cg10303698 (CUEDC1) (P=0.001); rs3758249 (FOXE1) - cg20308679 (FRZB) (P=0.04); rs8001641 (SPRY2) - cg19191560 (LGR4) (P=0.04); rs987525(8q24) - cg16561172(MYC) (P=0.00000963); rs7590268(THADA) - cg06873343 (TTYH3) (P=0.04); rs7078160 (VAX1) - cg09487139 (P=0.05); rs560426 (ABCA4/ARHGAP29) - cg25196715 (ABCA4/ARHGAP29) (P=0,03). Paired T-test showed significant differences for cg06873343 (TTYH3) (P=0.04); cg17103269 (LPIN3) (P=0.002), and cg19191560 (LGR4) (P=0.05). Conclusions Our results confirm previous evidence that some of the common non-coding variants detected through GWAS studies can influence the risk of OFCs via epigenetic mechanisms, such as DNAm, which can ultimately affect and regulate gene expression. Given the large prevalence of non-coding SNPs in most OFCs genome wide association studies, our findings can potentially address major knowledge gaps, like missing heritability, reduced penetrance, and variable expressivity associated with OFCs phenotypes.
Collapse
Affiliation(s)
- L A Machado-Paula
- University of Iowa College of Dentistry and Dental Clinics, Iowa City, IA, USA
| | | | - R T Lie
- University of Bergen, Bergen, Norway
| | - L Hovey
- University of Iowa College of Dentistry and Dental Clinics, Iowa City, IA, USA
| | - B Doolittle
- University of Iowa College of Dentistry and Dental Clinics, Iowa City, IA, USA
| | - W Awotoye
- University of Iowa College of Dentistry and Dental Clinics, Iowa City, IA, USA
| | - L Dunlay
- University of Iowa College of Dentistry and Dental Clinics, Iowa City, IA, USA
| | - X J Xie
- University of Iowa College of Dentistry and Dental Clinics, Iowa City, IA, USA
| | - E Zeng
- University of Iowa College of Dentistry and Dental Clinics, Iowa City, IA, USA
| | - A Butali
- University of Iowa College of Dentistry and Dental Clinics, Iowa City, IA, USA
- University of Iowa Carver College of Medicine, Iowa City, IA, USA
| | | | - J C Murray
- University of Iowa Carver College of Medicine, Iowa City, IA, USA
| | - L M Moreno-Uribe
- University of Iowa College of Dentistry and Dental Clinics, Iowa City, IA, USA
| | - A L Petrin
- University of Iowa College of Dentistry and Dental Clinics, Iowa City, IA, USA
| |
Collapse
|
32
|
Goldberg DC, Cloud C, Lee SM, Barnes B, Gruber S, Kim E, Pottekat A, Westphal MS, McAuliffe L, Majounie E, KalayilManian M, Zhu Q, Tran C, Hansen M, Stojakovic J, Parker JB, Kohli RM, Porecha R, Renke N, Zhou W. Scalable Screening of Ternary-Code DNA Methylation Dynamics Associated with Human Traits. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2024.05.17.594606. [PMID: 38826316 PMCID: PMC11142114 DOI: 10.1101/2024.05.17.594606] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/04/2024]
Abstract
Epigenome-wide association studies (EWAS) are transforming our understanding of the interplay between epigenetics and complex human traits and phenotypes. We introduce the Methylation Screening Array (MSA), a new iteration of the Infinium technology for scalable and quantitative screening of trait associations of nuanced ternary-code cytosine modifications in larger, more inclusive, and stratified human populations. MSA integrates EWAS, single-cell, and cell-type-resolved methylome profiles, covering diverse human traits and diseases. Our first MSA applications yield multiple biological insights: we revealed a previously unappreciated role of 5-hydroxymethylcytosine (5hmC) in trait associations and epigenetic clocks. We demonstrated that 5hmCs complement 5-methylcytosines (5mCs) in defining tissues and cells' epigenetic identities. In-depth analyses highlighted the cell type context of EWAS and GWAS hits. Using this platform, we conducted a comprehensive human 5hmC aging EWAS, discovering tissue-invariant and tissue-specific aging dynamics, including distinct tissue-specific rates of mitotic hyper- and hypomethylation rates. These findings chart a landscape of the complex interplay of the two forms of cytosine modifications in diverse human tissues and their roles in health and disease.
Collapse
Affiliation(s)
- David C Goldberg
- Center for Computational and Genomic Medicine, The Children’s Hospital of Philadelphia, PA, 19104, USA
| | - Cameron Cloud
- Center for Computational and Genomic Medicine, The Children’s Hospital of Philadelphia, PA, 19104, USA
| | - Sol Moe Lee
- Center for Computational and Genomic Medicine, The Children’s Hospital of Philadelphia, PA, 19104, USA
| | | | | | - Elliot Kim
- Center for Computational and Genomic Medicine, The Children’s Hospital of Philadelphia, PA, 19104, USA
| | | | | | | | | | | | | | | | | | | | - Jared B Parker
- Department of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Rahul M Kohli
- Department of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | | | | | - Wanding Zhou
- Center for Computational and Genomic Medicine, The Children’s Hospital of Philadelphia, PA, 19104, USA
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| |
Collapse
|
33
|
Song ZQ, Chen YQ, Xuan CH, Ni TT, Xu YP, Lu XY, Chen FR, Chen YH. Effect of smoking behaviour and related blood DNA methylation on visceral adipose tissues. Diabetes Obes Metab 2025; 27:619-628. [PMID: 39511847 DOI: 10.1111/dom.16054] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/19/2024] [Revised: 10/22/2024] [Accepted: 10/22/2024] [Indexed: 11/15/2024]
Abstract
BACKGROUND Recent studies have found that tobacco smoking is associated with fat distribution, yet limited research has focused on its relationship with visceral adipose tissues (VATs). Furthermore, the cellular and molecular mechanisms underlying the interactions among smoking, epigenetic modifications, and VATs remain unknown. METHOD We performed univariable Mendelian randomization (MR) analysis to elucidate the causal relationship between smoking behaviours and VATs, including epicardial and pericardial adipose tissue (EPAT), liver fat (LF), and pancreas fat (PF). This approach could minimize the impact of confounders and reverse causality through utilizing genetic variants to proxy the smoking behaviours. Mediation MR analysis were conducted to detect potential mediators. Additionally, summary-data-based MR (SMR) and colocalization analysis were performed to explore the association between smoking-related DNA methylation and VATs. RESULTS We identified a convincing association between smoking initiation and increased EPAT (beta: 0.15, 95% CI: 0.06, 0.23, p = 7.01 × 10-4) and LF area (beta: 0.15, 95% CI = 0.05, 0.24, p = 2.85 × 10-3), respectively. Further mediation analysis suggested type 2 diabetes mellitus (T2DM) as a potential mediator within these co-relationships. When further exploring the associations between the smoking related DNA methylation and VATs, we identified that WT1 methylation at cg05222924 was significantly linked to a lower EPAT area (beta: -0.12, 95% CI: -0.16, -0.06, PFDR = 2.24 × 10-3), while GPX1 methylation at cg18642234 facilitated the deposition of EPAT (beta: 0.15, 95% CI: 0.10, 0.20, PFDR = 1.66 × 10-4). CONCLUSION Our study uncovered a significant causal effect between smoking and VATs, with T2DM identified as a potential mediator. Further investigation into DNA methylation yielded novel insights into the pathogenic role of smoking on EPAT.
Collapse
Affiliation(s)
- Zheng-Qi Song
- Department of Cardiology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
- The First Clinical Medical College, Wenzhou Medical University, Wenzhou, China
| | - Yi-Qi Chen
- The Second Clinical Medical College, Wenzhou Medical University, Wenzhou, China
| | - Chen-Hao Xuan
- The First Clinical Medical College, Wenzhou Medical University, Wenzhou, China
| | - Tong-Tong Ni
- The First Clinical Medical College, Wenzhou Medical University, Wenzhou, China
| | - Yu-Peng Xu
- The First Clinical Medical College, Wenzhou Medical University, Wenzhou, China
| | - Xin-Yu Lu
- The First Clinical Medical College, Wenzhou Medical University, Wenzhou, China
| | - Fang-Ran Chen
- The First Clinical Medical College, Wenzhou Medical University, Wenzhou, China
| | - Yi-He Chen
- Department of Cardiology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| |
Collapse
|
34
|
Campa D, Gentiluomo M, Rizzato C. Genetic landscape for screening and early diagnosis of pancreatic ductal adenocarcinoma: is there a signature? Best Pract Res Clin Gastroenterol 2025; 74:101988. [PMID: 40210334 DOI: 10.1016/j.bpg.2025.101988] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/16/2025] [Accepted: 02/03/2025] [Indexed: 04/12/2025]
Abstract
The last 15 years have seen unprecedent advancement in genomics techniques such as dense single nucleotide variants (SNVs) arrays or next generation Sequencing. In parallel, new analytical methodologies have been developed to streamline data understanding and integration. These advances have been instrumental in identifying common genetic variants associated with pancreatic ductal adenocarcinoma (PDAC) risk. The role of the individual variants is rather small, and they have no clinical utility for screening or early detection. However, their combined effect computed though polygenic risk scores (PGS) are showing promising potentiality in PDAC risk prediction. There still caveats, and limitations that need to be properly addressed however it is foreseeable that the genetic background will become a powerful tool in PDAC prediction, leveraging the advantage that it has compared to other biomarkers: germline genetics is invariable from birth to death.
Collapse
|
35
|
Tost J, Ak-Aksoy S, Campa D, Corradi C, Farinella R, Ibáñez-Costa A, Dubrot J, Earl J, Melian EB, Kataki A, Kolnikova G, Madjarov G, Chaushevska M, Strnadel J, Tanić M, Tomas M, Dubovan P, Urbanova M, Buocikova V, Smolkova B. Leveraging epigenetic alterations in pancreatic ductal adenocarcinoma for clinical applications. Semin Cancer Biol 2025; 109:101-124. [PMID: 39863139 DOI: 10.1016/j.semcancer.2025.01.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2024] [Revised: 01/10/2025] [Accepted: 01/13/2025] [Indexed: 01/27/2025]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a highly aggressive malignancy characterized by late detection and poor prognosis. Recent research highlights the pivotal role of epigenetic alterations in driving PDAC development and progression. These changes, in conjunction with genetic mutations, contribute to the intricate molecular landscape of the disease. Specific modifications in DNA methylation, histone marks, and non-coding RNAs are emerging as robust predictors of disease progression and patient survival, offering the potential for more precise prognostic tools compared to conventional clinical staging. Moreover, the detection of epigenetic alterations in blood and other non-invasive samples holds promise for earlier diagnosis and improved management of PDAC. This review comprehensively summarises current epigenetic research in PDAC and identifies persisting challenges. These include the complex nature of epigenetic profiles, tumour heterogeneity, limited access to early-stage samples, and the need for highly sensitive liquid biopsy technologies. Addressing these challenges requires the standardisation of methodologies, integration of multi-omics data, and leveraging advanced computational tools such as machine learning and artificial intelligence. While resource-intensive, these efforts are essential for unravelling the functional consequences of epigenetic changes and translating this knowledge into clinical applications. By overcoming these hurdles, epigenetic research has the potential to revolutionise the management of PDAC and improve patient outcomes.
Collapse
Affiliation(s)
- Jorg Tost
- Centre National de Recherche en Génomique Humaine, CEA - Institut de Biologie François Jacob, University Paris - Saclay, Evry, France.
| | - Secil Ak-Aksoy
- Bursa Uludag University Faculty of Medicine, Medical Microbiology, Bursa 16059, Turkey.
| | - Daniele Campa
- Department of Biology, University of Pisa, via Derna 1, Pisa 56126, Italy.
| | - Chiara Corradi
- Department of Biology, University of Pisa, via Derna 1, Pisa 56126, Italy.
| | - Riccardo Farinella
- Department of Biology, University of Pisa, via Derna 1, Pisa 56126, Italy.
| | - Alejandro Ibáñez-Costa
- Maimonides Biomedical Research Institute of Cordoba (IMIBIC), Department of Cell Biology, Physiology, and Immunology, University of Cordoba, Reina Sofia University Hospital, Edificio IMIBIC, Avenida Men´endez Pidal s/n, Cordoba 14004, Spain.
| | - Juan Dubrot
- Solid Tumors Program, Cima Universidad de Navarra, Cancer Center Clínica Universidad de Navarra (CCUN), Pamplona, Spain.
| | - Julie Earl
- Biomarkers and Personalized Approach to Cancer (BIOPAC) Group, Ramón y Cajal Institute for Health Research (IRYCIS), Ctra Colmenar Viejo Km 9.100, CIBERONC, Madrid 28034, Spain.
| | - Emma Barreto Melian
- Biomarkers and Personalized Approach to Cancer (BIOPAC) Group, Ramón y Cajal Institute for Health Research (IRYCIS), Ctra Colmenar Viejo Km 9.100, CIBERONC, Madrid 28034, Spain
| | - Agapi Kataki
- A' Department of Propaedeutic Surgery, National and Kapodistrian University of Athens, Vas. Sofias 114, Athens 11527, Greece.
| | - Georgina Kolnikova
- Department of Pathology, National Cancer Institute in Bratislava, Klenova 1, Bratislava 83310, Slovakia.
| | - Gjorgji Madjarov
- Ss. Cyril and Methodius University - Faculty of Computer Science and Engineering, Rudjer Boshkovikj 16, Skopje 1000, Macedonia.
| | - Marija Chaushevska
- Ss. Cyril and Methodius University - Faculty of Computer Science and Engineering, Rudjer Boshkovikj 16, Skopje 1000, Macedonia; gMendel ApS, Fruebjergvej 3, Copenhagen 2100, Denmark.
| | - Jan Strnadel
- Biomedical Centre Martin, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, Martin 036 01, Slovakia.
| | - Miljana Tanić
- Experimental Oncology Department, Institute for Oncology and Radiology of Serbia, Serbia; UCL Cancer Institute, University College London, London WC1E 6DD, UK.
| | - Miroslav Tomas
- Department of Surgical Oncology, National Cancer Institute in Bratislava and Slovak Medical University in Bratislava, Klenova 1, Bratislava 83310, Slovakia.
| | - Peter Dubovan
- Department of Surgical Oncology, National Cancer Institute in Bratislava and Slovak Medical University in Bratislava, Klenova 1, Bratislava 83310, Slovakia.
| | - Maria Urbanova
- Biomedical Research Center, Slovak Academy of Sciences, Dubravska Cesta 9, Bratislava 84505, Slovakia.
| | - Verona Buocikova
- Biomedical Research Center, Slovak Academy of Sciences, Dubravska Cesta 9, Bratislava 84505, Slovakia.
| | - Bozena Smolkova
- Biomedical Research Center, Slovak Academy of Sciences, Dubravska Cesta 9, Bratislava 84505, Slovakia.
| |
Collapse
|
36
|
Marques IF, Domènech-Panicello C, Geurtsen ML, Hoang TT, Richmond R, Polinski K, Sirignano L, Page CM, Binter AC, Everson T, Burt A, Deuschle M, Gilles M, Streit F, Mumford SL, Magnus P, Reiss IKM, Vermeulen MJ, Witt SH, Chaves I, Yeung E, London SJ, Guxens M, Felix JF. Associations of maternal night shift work during pregnancy with DNA methylation in offspring: a meta-analysis in the PACE consortium. Clin Epigenetics 2025; 17:12. [PMID: 39844285 PMCID: PMC11756212 DOI: 10.1186/s13148-024-01810-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2024] [Accepted: 12/27/2024] [Indexed: 01/24/2025] Open
Abstract
BACKGROUND Night shift work during pregnancy has been associated with differential DNA methylation in placental tissue, but no studies have explored this association in cord blood. We aimed to examine associations of maternal night shift work with cord blood DNA methylation. METHODS A total of 4487 mother-newborn pairs from 7 studies were included. Maternal night shift work during pregnancy was ascertained via questionnaires and harmonized into "any" versus "no". DNA methylation was measured in cord blood using the Illumina Infinium Methylation arrays. Robust linear regression models adjusted for relevant confounders were run in the individual cohorts, and results were meta-analyzed. RESULTS Maternal night shift work during pregnancy ranged from 3.4% to 26.3%. Three CpGs were differentially methylated in relation to maternal night shift work during pregnancy at a false discovery rate adjusted P < 0.05: cg10945885 (estimate (β) 0.38%, standard error (SE) 0.07), cg00773359 (β 0.25%, SE 0.05), and cg21836426 (β - 0.29%, SE 0.05). Associations of the identified CpGs were found in previous literature for gestational age and childhood and adolescent BMI. In a mouse model of prenatal jet lag exposure, information on offspring DNA methylation of ten homologous genes annotated to the 16 CpGs with P < 1 × 10-5 in our analysis was available, of which eight were associated (enrichment P: 1.62 × 10-11). CONCLUSION Maternal night shift work during pregnancy was associated with newborn DNA methylation at 3 CpGs. Top findings overlapped with those in a mouse model of gestational jet lag. This work strengthens evidence that DNA methylation could be a marker or mediator of impacts of circadian rhythm disturbances.
Collapse
Affiliation(s)
- Irene F Marques
- Generation R Study Group, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands
- Department of Pediatrics, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands
| | - Carola Domènech-Panicello
- ISGlobal, Barcelona, Spain
- Universitat Pompeu Fabra (UPF), Barcelona, Spain
- CIBER Epidemiología y Salud Pública (CIBERESP), Instituto de Salud Carlos III, Madrid, Spain
| | - Madelon L Geurtsen
- Generation R Study Group, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands
- Department of Pediatrics, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands
| | - Thanh T Hoang
- Division of Intramural Research, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC, USA
- Department of Pediatrics, Division of Hematology-Oncology, Baylor College of Medicine, Houston, TX, USA
- Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX, USA
- Cancer and Hematology Center, Texas Children's Hospital, Houston, TX, USA
| | - Rebecca Richmond
- Medical Research Council Integrative Epidemiology Unit, University of Bristol, Bristol, UK
- Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
- NIHR Oxford Health Biomedical Research Centre, University of Oxford, Oxford, UK
| | - Kristen Polinski
- Division of Population Health Research, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, Bethesda, MD, USA
| | - Lea Sirignano
- Department of Genetic Epidemiology in Psychiatry, Medical Faculty Mannheim, Central Institute of Mental Health, Heidelberg University, Mannheim, Germany
| | - Christian M Page
- Centre for Fertility and Health, Norwegian Institute of Public Health, Oslo, Norway
- Department of Physical Health and Aging, Division for Physical and Mental Health, Norwegian Institute of Public Health, Oslo, Norway
| | - Anne-Claire Binter
- ISGlobal, Barcelona, Spain
- Universitat Pompeu Fabra (UPF), Barcelona, Spain
- CIBER Epidemiología y Salud Pública (CIBERESP), Instituto de Salud Carlos III, Madrid, Spain
| | - Todd Everson
- Gangarosa Department of Environmental Health, Rollins School of Public Health, Emory University, Atlanta, GA, USA
| | - Amber Burt
- Gangarosa Department of Environmental Health, Rollins School of Public Health, Emory University, Atlanta, GA, USA
| | - Michael Deuschle
- Department of Psychiatry and Psychotherapy, Medical Faculty Mannheim, Central Institute of Mental Health, Heidelberg University, Mannheim, Germany
| | - Maria Gilles
- Department of Psychiatry and Psychotherapy, Medical Faculty Mannheim, Central Institute of Mental Health, Heidelberg University, Mannheim, Germany
| | - Fabian Streit
- Department of Genetic Epidemiology in Psychiatry, Medical Faculty Mannheim, Central Institute of Mental Health, Heidelberg University, Mannheim, Germany
- Department of Psychiatry and Psychotherapy, Medical Faculty Mannheim, Central Institute of Mental Health, Heidelberg University, Mannheim, Germany
- Medical Faculty Mannheim, Hector Institute for Artificial Intelligence in Psychiatry, Central Institute of Mental Health, Heidelberg University, Mannheim, Germany
| | - Sunni L Mumford
- Division of Population Health Research, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, Bethesda, MD, USA
- Department of Biostatistics, Epidemiology and Informatics and Department of Obstetrics and Gynecology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Per Magnus
- Centre for Fertility and Health, Norwegian Institute of Public Health, Oslo, Norway
| | - Irwin K M Reiss
- Department of Neonatal and Pediatric Intensive Care, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands
| | - Marijn J Vermeulen
- Department of Neonatal and Pediatric Intensive Care, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands
| | - Stephanie H Witt
- Department of Genetic Epidemiology in Psychiatry, Medical Faculty Mannheim, Central Institute of Mental Health, Heidelberg University, Mannheim, Germany
| | - Inês Chaves
- Department of Molecular Genetics, Erasmus MC Cancer Institute, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands
| | - Edwina Yeung
- Division of Population Health Research, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, Bethesda, MD, USA
| | - Stephanie J London
- Division of Intramural Research, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC, USA
| | - Mònica Guxens
- ISGlobal, Barcelona, Spain
- Universitat Pompeu Fabra (UPF), Barcelona, Spain
- CIBER Epidemiología y Salud Pública (CIBERESP), Instituto de Salud Carlos III, Madrid, Spain
- Department of Child and Adolescent Psychiatry/Psychology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands
- ICREA, Barcelona, Spain
| | - Janine F Felix
- Generation R Study Group, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands.
- Department of Pediatrics, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands.
| |
Collapse
|
37
|
Zheng X, Tian Z, Che X, Zhang X, Xiang Y, Ge Z, Zhai Z, Ma Q, Pan J. DMRdb: a disease-centric Mendelian randomization database for systematically assessing causal relationships of diseases with genes, proteins, CpG sites, metabolites and other diseases. Nucleic Acids Res 2025; 53:D1363-D1371. [PMID: 39351893 PMCID: PMC11701675 DOI: 10.1093/nar/gkae853] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2024] [Revised: 09/10/2024] [Accepted: 09/18/2024] [Indexed: 01/18/2025] Open
Abstract
Exploring the causal relationships of diseases with genes, proteins, CpG sites, metabolites and other diseases is fundamental to the life sciences. However, large-scale research using Mendelian randomization (MR) analysis is currently lacking. To address this, we introduce DMRdb (http://www.inbirg.com/DMRdb/), a disease-centric Mendelian randomization database, designed to systematically assess causal relationships of diseases with genes, proteins, CpG sites, metabolites and other diseases. The database consists of three main components: (i) 6640 high-quality disease genome-wide association studies (GWASs) from public sources that were subjected to rigorous quality filtering and standardization; (ii) over 497 billion results from MR analyses involving 6640 disease GWAS datasets, 16 238 expression quantitative trait loci (eQTLs) data, 2564 protein quantitative trait loci (pQTLs) data, 12 000 methylation quantitative trait locus (meQTLs) data and 825 metabolites data and (iii) over 380 000 causal relationship pairs from 1223 literature sources relevant to MR analyses. A user-friendly online database was developed to allow users to query, search, and download all the results. In summary, we anticipate that DMRdb will be a valuable resource for advancing our understanding of disease mechanisms and identifying new biomarkers and therapeutic targets.
Collapse
Affiliation(s)
- Xiao Zheng
- Basic Medicine Research and Innovation Center for Novel Target and Therapeutic Intervention, Ministry of Education, College of Pharmacy, and Precision Medicine Center, the Second Affiliated Hospital, Chongqing Medical University, Chongqing 400016, China
| | - Zhihao Tian
- Basic Medicine Research and Innovation Center for Novel Target and Therapeutic Intervention, Ministry of Education, College of Pharmacy, and Precision Medicine Center, the Second Affiliated Hospital, Chongqing Medical University, Chongqing 400016, China
| | - Xiaohui Che
- Basic Medicine Research and Innovation Center for Novel Target and Therapeutic Intervention, Ministry of Education, College of Pharmacy, and Precision Medicine Center, the Second Affiliated Hospital, Chongqing Medical University, Chongqing 400016, China
| | - Xu Zhang
- Basic Medicine Research and Innovation Center for Novel Target and Therapeutic Intervention, Ministry of Education, College of Pharmacy, and Precision Medicine Center, the Second Affiliated Hospital, Chongqing Medical University, Chongqing 400016, China
| | - Yu Xiang
- Basic Medicine Research and Innovation Center for Novel Target and Therapeutic Intervention, Ministry of Education, College of Pharmacy, and Precision Medicine Center, the Second Affiliated Hospital, Chongqing Medical University, Chongqing 400016, China
| | - Zhijian Ge
- Basic Medicine Research and Innovation Center for Novel Target and Therapeutic Intervention, Ministry of Education, College of Pharmacy, and Precision Medicine Center, the Second Affiliated Hospital, Chongqing Medical University, Chongqing 400016, China
| | - Zhaoyu Zhai
- Basic Medicine Research and Innovation Center for Novel Target and Therapeutic Intervention, Ministry of Education, College of Pharmacy, and Precision Medicine Center, the Second Affiliated Hospital, Chongqing Medical University, Chongqing 400016, China
| | - Qinfeng Ma
- Basic Medicine Research and Innovation Center for Novel Target and Therapeutic Intervention, Ministry of Education, College of Pharmacy, and Precision Medicine Center, the Second Affiliated Hospital, Chongqing Medical University, Chongqing 400016, China
| | - Jianbo Pan
- Basic Medicine Research and Innovation Center for Novel Target and Therapeutic Intervention, Ministry of Education, College of Pharmacy, and Precision Medicine Center, the Second Affiliated Hospital, Chongqing Medical University, Chongqing 400016, China
| |
Collapse
|
38
|
Cao S, Zeng Y, Pang K, Chen M, Guo R, Wu N, Fang C, Deng H, Zhang X, Xie X, Ouyang W, Yang H. Unraveling the causal impact of smoking and its DNA methylation signatures on cardiovascular disease: Mendelian randomization and colocalization analysis. Clin Epigenetics 2025; 17:1. [PMID: 39748436 PMCID: PMC11694376 DOI: 10.1186/s13148-024-01808-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2024] [Accepted: 12/23/2024] [Indexed: 01/04/2025] Open
Abstract
BACKGROUND To explore the mechanisms linking smoking to cardiovascular diseases (CVDs) from an epigenetic perspective. METHODS Mendelian Randomization (MR) analysis was performed to assess the causal effects of smoking behavior and DNA methylation levels at smoking-related CpG sites on nine CVDs, including aortic aneurysm, atrial fibrillation, coronary atherosclerosis, coronary heart disease, heart failure, intracerebral hemorrhage, ischemic stroke, myocardial infarction, subarachnoid hemorrhage. Colocalization analysis was used to further identify key smoking-related CpG sites from the MR causal estimates. Reactome enrichment analysis was used to elucidate the potential mechanisms. RESULTS MR analysis indicates that smoking behaviors are significantly associated with an increased risk of nine CVDs (OR > 1, P < 0.05). Through MR and colocalization analysis, five key smoking-related CpG sites were ultimately determined. DNA methylation alteration at cg25313468 (located in the TSS1500 region of REST) is simultaneously associated with the risk of atrial fibrillation, coronary atherosclerosis, coronary heart disease, and myocardial infarction. Additionally, cg21647257 (located in the TSS200 region of CLIP3) is associated with the risk of atrial fibrillation; cg06197751 (located in SGEF gene body) and cg07520810 (located in ARID5B gene body) are associated with the risk of coronary atherosclerosis; cg16822035 (located in MCF2L gene body) is associated with the risk of myocardial infarction. Enrichment analysis suggests that phosphatase and tensin homologue (PTEN) may be involved in the downstream mechanisms of cg25313468 (REST). CONCLUSION This study uncovers the relationship between smoking, DNA methylation, and CVDs, providing new insights into the pathogenic effect of smoking on CVDs from an epigenetic perspective.
Collapse
Affiliation(s)
- Si Cao
- Department of Anesthesiology, Third Xiangya Hospital, Central South University, Changsha, 410013, Hunan, China
| | - Youjie Zeng
- Department of Anesthesiology, Third Xiangya Hospital, Central South University, Changsha, 410013, Hunan, China
| | - Ke Pang
- Department of Anesthesiology, Third Xiangya Hospital, Central South University, Changsha, 410013, Hunan, China
| | - Minghua Chen
- Department of Anesthesiology, Third Xiangya Hospital, Central South University, Changsha, 410013, Hunan, China
| | - Ren Guo
- Department of Pharmacy, Third Xiangya Hospital, Central South University, Changsha, 410013, Hunan, China
| | - Nayiyuan Wu
- The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University/Hunan Cancer Hospital, Changsha, Hunan, China
| | - Chao Fang
- The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University/Hunan Cancer Hospital, Changsha, Hunan, China
| | - Huiyin Deng
- Department of Anesthesiology, Third Xiangya Hospital, Central South University, Changsha, 410013, Hunan, China
| | - Xiaoyi Zhang
- Department of Medicine, Jacobi Medical Center, Albert Einstein College of Medicine, Bronx, NY, 10461, USA
| | - Xiaohui Xie
- Department of Medicine, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Wen Ouyang
- Department of Anesthesiology, Third Xiangya Hospital, Central South University, Changsha, 410013, Hunan, China
| | - Heng Yang
- Department of Neurology, Third Xiangya Hospital, Central South University, 138 Tongzipo Road, Yuelu District, Changsha, 410013, Hunan, China.
| |
Collapse
|
39
|
Tang Z, Chen Y, Ouyang Y, Peng Y, Man X. COVID-19 related epigenetic changes and atopic dermatitis: An exploratory analysis. World Allergy Organ J 2025; 18:101022. [PMID: 39867872 PMCID: PMC11758953 DOI: 10.1016/j.waojou.2024.101022] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2024] [Revised: 09/13/2024] [Accepted: 12/06/2024] [Indexed: 01/28/2025] Open
Abstract
Background While epidemiological data suggest a connection between atopic dermatitis (AD) and COVID-19, the molecular mechanisms underlying this relationship remain unclear. Objective To investigate whether COVID-19-related CpGs may contribute to AD development and whether this association is mediated through the regulation of specific genes' expression. Methods We combined Mendelian randomization and transcriptome analysis for data-driven explorations. Results Among the 172 CpGs -associated with COVID-19 infection, merely 3 of them exhibited significant impacts on the risk of AD, including cg04543273, cg11916609, and cg10636246. In the following analysis of the causal effects of CpGs and their related gene expression, cg04543273 inhibited LMAN2 expression. However, there was not a significant impact of cg11916609 and cg10636246 on the expression of their corresponding genes. Besides, transcriptome analysis suggested that LMAN2 expression was significantly upregulated among the COVID-19-infected population, and LMAN2 expression was obviously correlated with Type 2 helper cells across different post-infection time points. Conclusion Overall, this study provides new insights of the COVID-19-related onset and exacerbation of AD-COVID-19-related epigenetic changes and their regulatory impact on transcription. A novel role of LMAN2 was proposed in the relationship between viral infection and AD. More studies are warranted to further explore the mechanism of LMAN2-related immunopathology.
Collapse
Affiliation(s)
- Zhenwei Tang
- Department of Dermatology, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yu Chen
- Clinical Medicine Eight-year Program, Xiangya Hospital, Central South University, Changsha, China
| | - Yuzhen Ouyang
- Clinical Medicine Eight-year Program, Xiangya Hospital, Central South University, Changsha, China
| | - Yu Peng
- Department of Rheumatology, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Xiaoyong Man
- Department of Dermatology, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| |
Collapse
|
40
|
Ji A, Sui Y, Xue X, Ji X, Shi W, Shi Y, Terkeltaub R, Dalbeth N, Takei R, Yan F, Sun M, Li M, Lu J, Cui L, Liu Z, Wang C, Li X, Han L, Fang Z, Sun W, Liang Y, He Y, Zheng G, Wang X, Wang J, Zhang H, Pang L, Qi H, Li Y, Cheng Z, Li Z, Xiao J, Zeng C, Merriman TR, Qu H, Fang X, Li C. Novel Genetic Loci in Early-Onset Gout Derived From Whole-Genome Sequencing of an Adolescent Gout Cohort. Arthritis Rheumatol 2025; 77:107-115. [PMID: 39118347 DOI: 10.1002/art.42969] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Revised: 08/01/2024] [Accepted: 08/05/2024] [Indexed: 08/10/2024]
Abstract
OBJECTIVE Mechanisms underlying the adolescent-onset and early-onset gout are unclear. This study aimed to discover variants associated with early-onset gout. METHODS We conducted whole-genome sequencing in a discovery adolescent-onset gout cohort of 905 individuals (gout onset 12 to 19 years) to discover common and low-frequency single-nucleotide variants (SNVs) associated with gout. Candidate common SNVs were genotyped in an early-onset gout cohort of 2,834 individuals (gout onset ≤30 years old), and meta-analysis was performed with the discovery and replication cohorts to identify loci associated with early-onset gout. Transcriptome and epigenomic analyses, quantitative real-time polymerase chain reaction and RNA sequencing in human peripheral blood leukocytes, and knock-down experiments in human THP-1 macrophage cells investigated the regulation and function of candidate gene RCOR1. RESULTS In addition to ABCG2, a urate transporter previously linked to pediatric-onset and early-onset gout, we identified two novel loci (Pmeta < 5.0 × 10-8): rs12887440 (RCOR1) and rs35213808 (FSTL5-MIR4454). Additionally, we found associations at ABCG2 and SLC22A12 that were driven by low-frequency SNVs. SNVs in RCOR1 were linked to elevated blood leukocyte messenger RNA levels. THP-1 macrophage culture studies revealed the potential of decreased RCOR1 to suppress gouty inflammation. CONCLUSION This is the first comprehensive genetic characterization of adolescent-onset gout. The identified risk loci of early-onset gout mediate inflammatory responsiveness to crystals that could mediate gouty arthritis. This study will contribute to risk prediction and therapeutic interventions to prevent adolescent-onset gout.
Collapse
Affiliation(s)
- Aichang Ji
- Affiliated Hospital of Qingdao University, Qingdao, China
| | - Yang Sui
- China National Center for Bioinformation, Beijing Institute of Genomics, Chinese Academy of Sciences, and University of Chinese Academy of Sciences, Beijing, China
| | - Xiaomei Xue
- Affiliated Hospital of Qingdao University, Qingdao, China
| | - Xiapeng Ji
- Affiliated Hospital of Qingdao University, Qingdao, China
| | - Wenrui Shi
- China National Center for Bioinformation, Beijing Institute of Genomics, Chinese Academy of Sciences, and University of Chinese Academy of Sciences, Beijing, China
| | - Yongyong Shi
- Affiliated Hospital of Qingdao University and Biomedical Sciences Institute of Qingdao University (Qingdao Branch of SJTU Bio-X Institutes), Qingdao University, Qingdao, China, and Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Collaborative Innovation Center for Brain Science, Shanghai Jiao Tong University, Shanghai, China
| | | | | | - Riku Takei
- Asia Pacific Gout Consortium and University of Alabama at Birmingham
| | - Fei Yan
- Affiliated Hospital of Qingdao University, Qingdao, China
| | - Mingshu Sun
- Shandong Provincial Clinical Research Center for Immune Diseases and Gout & Shandong Provincial Key Laboratory of Metabolic Diseases, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Maichao Li
- Affiliated Hospital of Qingdao University, Qingdao, China
| | - Jie Lu
- Affiliated Hospital of Qingdao University, Qingdao, China
| | - Lingling Cui
- Affiliated Hospital of Qingdao University, Qingdao, China
| | - Zhen Liu
- Affiliated Hospital of Qingdao University, Qingdao, China
| | - Can Wang
- Affiliated Hospital of Qingdao University, Qingdao, China
| | - Xinde Li
- Affiliated Hospital of Qingdao University, Qingdao, China
| | - Lin Han
- Affiliated Hospital of Qingdao University, Qingdao, China
| | - Zhanjie Fang
- China National Center for Bioinformation, Beijing Institute of Genomics, Chinese Academy of Sciences, and University of Chinese Academy of Sciences, Beijing, China
| | - Wenyan Sun
- Affiliated Hospital of Qingdao University, Qingdao, China
| | - Yue Liang
- China National Center for Bioinformation, Beijing Institute of Genomics, Chinese Academy of Sciences, and University of Chinese Academy of Sciences, Beijing, China
| | - Yuwei He
- Affiliated Hospital of Qingdao University, Qingdao, China
| | - Guangmin Zheng
- China National Center for Bioinformation, Beijing Institute of Genomics, Chinese Academy of Sciences, and University of Chinese Academy of Sciences, Beijing, China
| | - Xuefeng Wang
- Affiliated Hospital of Qingdao University, Qingdao, China
| | - Jiayi Wang
- Development Center for Medical Science & Technology, National Health Commission of the People's Republic of China, Beijing, China
| | - Hui Zhang
- Institute of Metabolic Diseases, Qingdao University, Qingdao, China
| | - Lei Pang
- Affiliated Hospital of Qingdao University, Qingdao, China
| | - Han Qi
- Affiliated Hospital of Qingdao University, Qingdao, China
| | - Yushuang Li
- Affiliated Hospital of Qingdao University, Qingdao, China
| | - Zan Cheng
- Affiliated Hospital of Qingdao University, Qingdao, China
| | - Zhiqiang Li
- The Biomedical Sciences Institute and The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, Shandong, China
| | - Jingfa Xiao
- China National Center for Bioinformation, Beijing Institute of Genomics, Chinese Academy of Sciences, and University of Chinese Academy of Sciences, Beijing, China
| | - Changqing Zeng
- China National Center for Bioinformation, Beijing Institute of Genomics, Chinese Academy of Sciences, and University of Chinese Academy of Sciences, Beijing, China
| | - Tony R Merriman
- Asia Pacific Gout Consortium, University of Alabama at Birmingham, Institute of Metabolic Diseases, Qingdao University, Qingdao, China, and University of Otago, Dunedin, New Zealand
| | - Hongzhu Qu
- China National Center for Bioinformation, Beijing Institute of Genomics, Chinese Academy of Sciences, University of Chinese Academy of Sciences, and Beijing Key Laboratory of Genome and Precision Medicine Technologies, Beijing, China
| | - Xiangdong Fang
- China National Center for Bioinformation, Beijing Institute of Genomics, Chinese Academy of Sciences, University of Chinese Academy of Sciences, and Beijing Key Laboratory of Genome and Precision Medicine Technologies, Beijing, China
| | - Changgui Li
- The Affiliated Hospital of Qingdao University, Qingdao, China, Asia Pacific Gout Consortium, and Institute of Metabolic Diseases, Qingdao University, Qingdao, China
| |
Collapse
|
41
|
Singh M, Dolan CV, Lapato DM, Hottenga JJ, Pool R, Verhulst B, Boomsma DI, Breeze CE, de Geus EJC, Hemani G, Min JL, Peterson RE, Maes HHM, van Dongen J, Neale MC. Unidirectional and bidirectional causation between smoking and blood DNA methylation: evidence from twin-based Mendelian randomisation. Eur J Epidemiol 2025; 40:55-69. [PMID: 39786687 PMCID: PMC11799127 DOI: 10.1007/s10654-024-01187-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2024] [Accepted: 11/22/2024] [Indexed: 01/12/2025]
Abstract
Cigarette smoking is associated with numerous differentially-methylated genomic loci in multiple human tissues. These associations are often assumed to reflect the causal effects of smoking on DNA methylation (DNAm), which may underpin some of the adverse health sequelae of smoking. However, prior causal analyses with Mendelian Randomisation (MR) have found limited support for such effects. Here, we apply an integrated approach combining MR with twin causal models to examine causality between smoking and blood DNAm in the Netherlands Twin Register (N = 2577). Analyses revealed potential causal effects of current smoking on DNAm at > 500 sites in/near genes enriched for functional pathways relevant to known biological effects of smoking (e.g., hemopoiesis, cell- and neuro-development, and immune regulation). Notably, we also found evidence of reverse and bidirectional causation at several DNAm sites, suggesting that variation in DNAm at these sites may influence smoking liability. Seventeen of the loci with putative effects of DNAm on smoking showed highly specific enrichment for gene-regulatory functional elements in the brain, while the top three sites annotated to genes involved in G protein-coupled receptor signalling and innate immune response. These novel findings are partly attributable to the analyses of current smoking in twin models, rather than lifetime smoking typically examined in MR studies, as well as the increased statistical power achieved using multiallelic/polygenic scores as instrumental variables while controlling for potential horizontal pleiotropy. This study highlights the value of twin studies with genotypic and DNAm data for investigating causal relationships of DNAm with health and disease.
Collapse
Affiliation(s)
- Madhurbain Singh
- Department of Psychiatry, Virginia Institute for Psychiatric and Behavioral Genetics, Virginia Commonwealth University, 800 E. Leigh St., Suite 100, Richmond, VA, 23298, USA.
- Department of Human and Molecular Genetics, Virginia Commonwealth University, Richmond, VA, USA.
- Department of Biological Psychology, Vrije Universiteit (VU) Amsterdam, Van Der Boechorststraat 7, 1081 BT, Amsterdam, The Netherlands.
| | - Conor V Dolan
- Department of Biological Psychology, Vrije Universiteit (VU) Amsterdam, Van Der Boechorststraat 7, 1081 BT, Amsterdam, The Netherlands
- Amsterdam Public Health Research Institute, Amsterdam, The Netherlands
| | - Dana M Lapato
- Department of Psychiatry, Virginia Institute for Psychiatric and Behavioral Genetics, Virginia Commonwealth University, 800 E. Leigh St., Suite 100, Richmond, VA, 23298, USA
- Department of Human and Molecular Genetics, Virginia Commonwealth University, Richmond, VA, USA
| | - Jouke-Jan Hottenga
- Department of Biological Psychology, Vrije Universiteit (VU) Amsterdam, Van Der Boechorststraat 7, 1081 BT, Amsterdam, The Netherlands
- Amsterdam Public Health Research Institute, Amsterdam, The Netherlands
| | - René Pool
- Department of Biological Psychology, Vrije Universiteit (VU) Amsterdam, Van Der Boechorststraat 7, 1081 BT, Amsterdam, The Netherlands
- Amsterdam Public Health Research Institute, Amsterdam, The Netherlands
| | - Brad Verhulst
- Department of Psychiatry and Behavioral Sciences, Texas A&M University, College Station, TX, USA
| | - Dorret I Boomsma
- Department of Biological Psychology, Vrije Universiteit (VU) Amsterdam, Van Der Boechorststraat 7, 1081 BT, Amsterdam, The Netherlands
- Amsterdam Public Health Research Institute, Amsterdam, The Netherlands
- Department of Complex Trait Genetics, Center for Neurogenomics and Cognitive Research, Vrije Universiteit (VU) Amsterdam, Amsterdam, The Netherlands
| | - Charles E Breeze
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Department Health and Human Services, Bethesda, MD, USA
- UCL Cancer Institute, University College London, London, UK
| | - Eco J C de Geus
- Department of Biological Psychology, Vrije Universiteit (VU) Amsterdam, Van Der Boechorststraat 7, 1081 BT, Amsterdam, The Netherlands
- Amsterdam Public Health Research Institute, Amsterdam, The Netherlands
| | - Gibran Hemani
- MRC Integrative Epidemiology Unit, University of Bristol, Bristol, UK
| | - Josine L Min
- MRC Integrative Epidemiology Unit, University of Bristol, Bristol, UK
| | - Roseann E Peterson
- Department of Psychiatry, Virginia Institute for Psychiatric and Behavioral Genetics, Virginia Commonwealth University, 800 E. Leigh St., Suite 100, Richmond, VA, 23298, USA
- Department of Psychiatry and Behavioral Sciences, SUNY Downstate Health Sciences University, Brooklyn, NY, USA
- Institute for Genomics in Health, SUNY Downstate Health Sciences University, Brooklyn, NY, USA
| | - Hermine H M Maes
- Department of Psychiatry, Virginia Institute for Psychiatric and Behavioral Genetics, Virginia Commonwealth University, 800 E. Leigh St., Suite 100, Richmond, VA, 23298, USA
- Department of Human and Molecular Genetics, Virginia Commonwealth University, Richmond, VA, USA
| | - Jenny van Dongen
- Department of Biological Psychology, Vrije Universiteit (VU) Amsterdam, Van Der Boechorststraat 7, 1081 BT, Amsterdam, The Netherlands.
- Amsterdam Public Health Research Institute, Amsterdam, The Netherlands.
| | - Michael C Neale
- Department of Psychiatry, Virginia Institute for Psychiatric and Behavioral Genetics, Virginia Commonwealth University, 800 E. Leigh St., Suite 100, Richmond, VA, 23298, USA.
- Department of Human and Molecular Genetics, Virginia Commonwealth University, Richmond, VA, USA.
- Department of Biological Psychology, Vrije Universiteit (VU) Amsterdam, Van Der Boechorststraat 7, 1081 BT, Amsterdam, The Netherlands.
| |
Collapse
|
42
|
Cosin-Tomas M, Hoang T, Qi C, Monasso GS, Langdon R, Kebede Merid S, Calas L, de Prado-Bert P, Richmond R, Jaddoe VV, Duijts L, Wright J, Annesi-Maesano I, Grazuleviciene R, Karachaliou M, Koppelman GH, Melén E, Gruzieva O, Vrijheid M, Yousefi P, Felix JF, London SJ, Bustamante M. Association of exposure to second-hand smoke during childhood with blood DNA methylation. ENVIRONMENT INTERNATIONAL 2025; 195:109204. [PMID: 39693780 DOI: 10.1016/j.envint.2024.109204] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/30/2024] [Revised: 11/11/2024] [Accepted: 12/09/2024] [Indexed: 12/20/2024]
Abstract
INTRODUCTION By recent estimates, 40% of children worldwide are exposed to second-hand smoke (SHS), which has been associated with adverse health outcomes. While numerous studies have linked maternal smoking during pregnancy (MSDP) to widespread differences in child blood DNA methylation (DNAm), research specifically examining postnatal SHS exposure remains sparse. To address this gap, we conducted epigenome-wide meta-analyses to identify associations of postnatal SHS and child blood DNAm. METHODS Six cohorts from the Pregnancy And Childhood Epigenetics (PACE) Consortium (total N = 2,695), with SHS data and child blood DNAm (aged 7-9 years) measured with the Illumina 450K array were included in the meta-analysis. Linear regression models adjusted for covariates were fitted to examine the association between the number of household smokers in postnatal life (0, 1, 2+) and child blood DNAm. Sensitivity models without adjusting for MSDP and restricted to mothers who did not smoke during pregnancy were evaluated. RESULTS Our analysis revealed significant associations (False Discovery Rate < 0.05) between household postnatal SHS exposure and DNAm at 11 CpGs in exposed children. Nine CpGs were mapped to genes (MYO1G, FAM184B, CTDSPL2, LTBP3, PDE10A, and FIBCD1), while 2 CpGs were located in open sea regions. Notably, all except 2 CpGs (mapped to FIBCD1 and CTDSPL2) have previously been linked to either personal smoking habits or in utero exposure to smoking. The models restricted to non-smoking mothers provided similar results. Importantly, several of these CpGs and their associated genes are implicated in conditions exacerbated by or directly linked to SHS. CONCLUSIONS Our findings highlight the potential biological effects of SHS on blood DNAm. These findings support further research on epigenetic factors mediating deleterious effects of SHS on child health and call for public health policies aimed at reducing exposure, particularly in environments where children are present.
Collapse
Affiliation(s)
- Marta Cosin-Tomas
- ISGlobal, Barcelona, Catalonia, Spain; Universitat Pompeu Fabra (UPF), Barcelona, Catalonia, Spain; Centro de investigación biomédica en red en epidemiología y salud pública (CIBERESP), Madrid, Spain.
| | - Thanh Hoang
- Division of Intramural Research, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC, USA; Department of Pediatrics, Division of Hematology-Oncology, Baylor College of Medicine, Houston, TX, USA; Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX, USA; Cancer and Hematology Center, Texas Children's Hospital, Houston, TX, USA
| | - Cancan Qi
- Groningen Research Institute for Asthma and COPD, Groningen, the Netherlands; Department of Pediatric Pulmonology and Pediatric Allergology, Beatrix Children's Hospital, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands; Microbiome Medicine Center, Department of Laboratory Medicine, ZhuJiang Hospital, Southern Medical University, Guangzhou, China
| | - Giulietta S Monasso
- The Generation R Study Group, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands; Department of Pediatrics, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands
| | - Ryan Langdon
- Medical Research Council Integrative Epidemiology Unit at the University of Bristol, University of Bristol, Bristol, UK; Population Health Science, Bristol Medical School, University of Bristol, Bristol, UK
| | - Simon Kebede Merid
- Department of Clinical Science and Education, Södersjukhuset, Karolinska Institutet, Stockholm, Sweden
| | - Lucinda Calas
- Université Paris Cité and Université Sorbonne Paris Nord, Inserm, INRAE, Center for Research in Epidemiology and StatisticS (CRESS), F-75004, Paris, France
| | - Paula de Prado-Bert
- ISGlobal, Barcelona, Catalonia, Spain; Universitat Pompeu Fabra (UPF), Barcelona, Catalonia, Spain; Centro de investigación biomédica en red en epidemiología y salud pública (CIBERESP), Madrid, Spain
| | - Rebecca Richmond
- Medical Research Council Integrative Epidemiology Unit at the University of Bristol, University of Bristol, Bristol, UK; Population Health Science, Bristol Medical School, University of Bristol, Bristol, UK
| | - Vincent Vw Jaddoe
- The Generation R Study Group, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands; Department of Pediatrics, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands
| | - Liesbeth Duijts
- The Generation R Study Group, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands; Department of Pediatrics, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands; Department of Neonatal and Pediatric Intensive Care, division of Neonatology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands
| | - John Wright
- Bradford Institute for Health Research, Bradford Teaching Hospitals NHS Foundation Trust, Bradford, UK
| | - Isabella Annesi-Maesano
- Université Paris Cité and Université Sorbonne Paris Nord, Inserm, INRAE, Center for Research in Epidemiology and StatisticS (CRESS), F-75004, Paris, France
| | | | - Marianna Karachaliou
- ISGlobal, Barcelona, Catalonia, Spain; Clinic of preventive and Social Medicine, Medical School, University of Crete, Iraklio, Greece
| | - Gerard H Koppelman
- Groningen Research Institute for Asthma and COPD, Groningen, the Netherlands; Department of Pediatric Pulmonology and Pediatric Allergology, Beatrix Children's Hospital, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Erik Melén
- Department of Clinical Science and Education, Södersjukhuset, Karolinska Institutet, Stockholm, Sweden; Sachs' Children's Hospital, Stockholm, Sweden
| | - Olena Gruzieva
- Centre for Occupational and Environmental Medicine, Region Stockholm, Stockholm, Sweden; Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Martine Vrijheid
- ISGlobal, Barcelona, Catalonia, Spain; Universitat Pompeu Fabra (UPF), Barcelona, Catalonia, Spain; Centro de investigación biomédica en red en epidemiología y salud pública (CIBERESP), Madrid, Spain
| | - Paul Yousefi
- Medical Research Council Integrative Epidemiology Unit at the University of Bristol, University of Bristol, Bristol, UK; Population Health Science, Bristol Medical School, University of Bristol, Bristol, UK; NIHR Bristol Biomedical Research Centre, University Hospitals Bristol and Weston NHS Foundation Trust and University of Bristol, Bristol, UK
| | - Janine F Felix
- The Generation R Study Group, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands; Department of Pediatrics, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands.
| | - Stephanie J London
- Division of Intramural Research, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC, USA
| | - Mariona Bustamante
- ISGlobal, Barcelona, Catalonia, Spain; Universitat Pompeu Fabra (UPF), Barcelona, Catalonia, Spain; Centro de investigación biomédica en red en epidemiología y salud pública (CIBERESP), Madrid, Spain
| |
Collapse
|
43
|
Waterfield S, Yousefi P, Suderman M. DNA methylation models of protein abundance across the lifecourse. Clin Epigenetics 2024; 16:189. [PMID: 39709440 DOI: 10.1186/s13148-024-01802-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Accepted: 12/13/2024] [Indexed: 12/23/2024] Open
Abstract
BACKGROUND Multiple studies have shown that DNA methylation (DNAm) models of protein abundance can be informative about exposure, phenotype and disease risk. Here we investigate and provide descriptive details of the capacity of DNAm to capture non-genetic variation in protein abundance across the lifecourse. METHODS We evaluated the performance of 14 previously published DNAm models of protein abundance (episcores) in peripheral blood from a large adult population using the Avon Longitudinal Study of Parents and Children (ALSPAC) at ages 7-24 and their mothers antenatally and in middle age (N range = 145-1464). New age-specific episcores were trained in ALSPAC and evaluated at different ages. In all instances, episcore-protein associations were evaluated with and without adjustment for genetics. The association between longitudinal protein stability and longitudinal episcore projection was also evaluated, as was sex-specificity of episcores derived solely in female participants. FINDINGS Of the 14 Gadd episcores, 10 generated estimates associated with abundance in middle age, 9 at age 24, and none at age 9. Eight of these episcores explained variation beyond genotype in adulthood (6 at age 24; 7 at midlife). At age 9, the abundances of 22 proteins could be modelled by DNAm, 7 beyond genotype of which one trained model generated informative estimates at ages 24 and in middle age. At age 24, 31 proteins could be modelled by DNAm, 19 beyond genotype, of which 5 trained models generated informative estimates at age 9 and 8 in middle age. In middle age, 23 proteins could be modelled, 13 beyond genotype, of which 3 were informative at age 9 and 7 at age 24. INTERPRETATION We observed that episcores performed better at older ages than in children with several episcores capturing non-genetic variation at all ages.
Collapse
Affiliation(s)
- Scott Waterfield
- MRC Integrative Epidemiology Unit, University of Bristol, Bristol, UK.
- Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK.
- Cancer Research UK Integrative Cancer Epidemiology Programme, University of Bristol, Bristol, UK.
- MRC Integrative Epidemiology Unit, Oakfield House, University of Bristol, Oakfield Grove, Bristol, BS8 2BN, UK.
| | - Paul Yousefi
- MRC Integrative Epidemiology Unit, University of Bristol, Bristol, UK
- Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
| | - Matt Suderman
- MRC Integrative Epidemiology Unit, University of Bristol, Bristol, UK
- Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
| |
Collapse
|
44
|
Großbach A, Suderman MJ, Hüls A, Lussier AA, Smith ADAC, Walton E, Dunn EC, Simpkin AJ. Maximizing insights from longitudinal epigenetic age data: simulations, applications, and practical guidance. Clin Epigenetics 2024; 16:187. [PMID: 39707425 DOI: 10.1186/s13148-024-01784-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2024] [Accepted: 11/15/2024] [Indexed: 12/23/2024] Open
Abstract
BACKGROUND Epigenetic age (EA) is an age estimate, developed using DNA methylation (DNAm) states of selected CpG sites across the genome. Although EA and chronological age are highly correlated, EA may not increase uniformly with time. Departures, known as epigenetic age acceleration (EAA), are common and have been linked to various traits and future disease risk. Limited by available data, most studies investigating these relationships have been cross-sectional, using a single EA measurement. However, the recent growth in longitudinal DNAm studies has led to analyses of associations with EA over time. These studies differ in (1) their choice of model; (2) the primary outcome (EA vs. EAA); and (3) in their use of chronological age or age-independent time variables to account for the temporal dynamic. We evaluated the robustness of each approach using simulations and tested our results in two real-world examples, using biological sex and birthweight as predictors of longitudinal EA. RESULTS Our simulations showed most accurate effect sizes in a linear mixed model or generalized estimating equation, using chronological age as the time variable. The use of EA versus EAA as an outcome did not strongly impact estimates. Applying the optimal model in real-world data uncovered advanced GrimAge in individuals assigned male at birth that decelerates over time. CONCLUSION Our results can serve as a guide for forthcoming longitudinal EA studies, aiding in methodological decisions that may determine whether an association is accurately estimated, overestimated, or potentially overlooked.
Collapse
Affiliation(s)
- Anna Großbach
- School of Mathematical and Statistical Sciences, University of Galway, Galway, Ireland.
- The SFI Centre for Research Training in Genomics Data Science, Galway, Ireland.
| | - Matthew J Suderman
- MRC Integrative Epidemiology Unit, Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
| | - Anke Hüls
- Department of Epidemiology, Rollins School of Public Health, Emory University, Atlanta, GA, USA
- Gangarosa Department of Environmental Health, Rollins School of Public Health, Emory University, Atlanta, GA, USA
- Department of Biostatistics and Bioinformatics, Rollins School of Public Health, Emory University, Atlanta, GA, USA
| | - Alexandre A Lussier
- Psychiatric and Neurodevelopmental Genetics Unit, Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA
- Department of Psychiatry, Harvard Medical School, Boston, MA, USA
- Stanley Center for Psychiatric Research, The Broad Institute of Harvard and MIT, Cambridge, MA, USA
| | - Andrew D A C Smith
- Mathematics and Statistics Research Group, University of the West of England, Bristol, UK
| | - Esther Walton
- Department of Psychology, University of Bath, Bath, UK
| | - Erin C Dunn
- Psychiatric and Neurodevelopmental Genetics Unit, Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA
- Department of Psychiatry, Harvard Medical School, Boston, MA, USA
- Stanley Center for Psychiatric Research, The Broad Institute of Harvard and MIT, Cambridge, MA, USA
- Department of Sociology, College of Liberal Arts, Purdue University, West Lafayette, IN, USA
| | - Andrew J Simpkin
- School of Mathematical and Statistical Sciences, University of Galway, Galway, Ireland
- The SFI Centre for Research Training in Genomics Data Science, Galway, Ireland
| |
Collapse
|
45
|
Katrinli S, Wani AH, Maihofer AX, Ratanatharathorn A, Daskalakis NP, Montalvo-Ortiz J, Núñez-Ríos DL, Zannas AS, Zhao X, Aiello AE, Ashley-Koch AE, Avetyan D, Baker DG, Beckham JC, Boks MP, Brick LA, Bromet E, Champagne FA, Chen CY, Dalvie S, Dennis MF, Fatumo S, Fortier C, Galea S, Garrett ME, Geuze E, Grant G, Hauser MA, Hayes JP, Hemmings SMJ, Huber BR, Jajoo A, Jansen S, Kessler RC, Kimbrel NA, King AP, Kleinman JE, Koen N, Koenen KC, Kuan PF, Liberzon I, Linnstaedt SD, Lori A, Luft BJ, Luykx JJ, Marx CE, McLean SA, Mehta D, Milberg W, Miller MW, Mufford MS, Musanabaganwa C, Mutabaruka J, Mutesa L, Nemeroff CB, Nugent NR, Orcutt HK, Qin XJ, Rauch SAM, Ressler KJ, Risbrough VB, Rutembesa E, Rutten BPF, Seedat S, Stein DJ, Stein MB, Toikumo S, Ursano RJ, Uwineza A, Verfaellie MH, Vermetten E, Vinkers CH, Ware EB, Wildman DE, Wolf EJ, Young RM, Zhao Y, van den Heuvel LL, Uddin M, Nievergelt CM, Smith AK, Logue MW. Epigenome-wide association studies identify novel DNA methylation sites associated with PTSD: a meta-analysis of 23 military and civilian cohorts. Genome Med 2024; 16:147. [PMID: 39696436 PMCID: PMC11658418 DOI: 10.1186/s13073-024-01417-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2024] [Accepted: 11/29/2024] [Indexed: 12/20/2024] Open
Abstract
BACKGROUND The occurrence of post-traumatic stress disorder (PTSD) following a traumatic event is associated with biological differences that can represent the susceptibility to PTSD, the impact of trauma, or the sequelae of PTSD itself. These effects include differences in DNA methylation (DNAm), an important form of epigenetic gene regulation, at multiple CpG loci across the genome. Moreover, these effects can be shared or specific to both central and peripheral tissues. Here, we aim to identify blood DNAm differences associated with PTSD and characterize the underlying biological mechanisms by examining the extent to which they mirror associations across multiple brain regions. METHODS As the Psychiatric Genomics Consortium (PGC) PTSD Epigenetics Workgroup, we conducted the largest cross-sectional meta-analysis of epigenome-wide association studies (EWASs) of PTSD to date, involving 5077 participants (2156 PTSD cases and 2921 trauma-exposed controls) from 23 civilian and military studies. PTSD diagnosis assessments were harmonized following the standardized guidelines established by the PGC-PTSD Workgroup. DNAm was assayed from blood using Illumina HumanMethylation450 or MethylationEPIC (850 K) BeadChips. Within each cohort, DNA methylation was regressed on PTSD, sex (if applicable), age, blood cell proportions, and ancestry. An inverse variance-weighted meta-analysis was performed. We conducted replication analyses in tissue from multiple brain regions, neuronal nuclei, and a cellular model of prolonged stress. RESULTS We identified 11 CpG sites associated with PTSD in the overall meta-analysis (1.44e - 09 < p < 5.30e - 08), as well as 14 associated in analyses of specific strata (military vs civilian cohort, sex, and ancestry), including CpGs in AHRR and CDC42BPB. Many of these loci exhibit blood-brain correlation in methylation levels and cross-tissue associations with PTSD in multiple brain regions. Out of 9 CpGs annotated to a gene expressed in blood, methylation levels at 5 CpGs showed significant correlations with the expression levels of their respective annotated genes. CONCLUSIONS This study identifies 11 PTSD-associated CpGs and leverages data from postmortem brain samples, GWAS, and genome-wide expression data to interpret the biology underlying these associations and prioritize genes whose regulation differs in those with PTSD.
Collapse
Affiliation(s)
- Seyma Katrinli
- Department of Gynecology and Obstetrics, Emory University, Atlanta, GA, USA
| | - Agaz H Wani
- Genomics Program, College of Public Health, University of South Florida, Tampa, FL, USA
| | - Adam X Maihofer
- Department of Psychiatry, University of California San Diego, La Jolla, CA, San Diego, USA
- Center of Excellence for Stress and Mental Health, Veterans Affairs San Diego Healthcare System, San Diego, CA, USA
- Research Service, Veterans Affairs San Diego Healthcare System, San Diego, CA, USA
| | - Andrew Ratanatharathorn
- Department of Epidemiology, Columbia University Mailmain School of Public Health, New York, NY, USA
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Nikolaos P Daskalakis
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Psychiatry, Harvard Medical School, Boston, MA, USA
- Center of Excellence in Depression and Anxiety Disorders, McLean Hospital, Belmont, MA, USA
| | - Janitza Montalvo-Ortiz
- U.S. Department of Veterans Affairs National Center of Posttraumatic Stress Disorder, Clinical Neurosciences Division, West Haven, CT, USA
- VA Connecticut Healthcare System, West Haven, CT, USA
- Department of Psychiatry, Yale School of Medicine, New Haven, CT, USA
| | - Diana L Núñez-Ríos
- U.S. Department of Veterans Affairs National Center of Posttraumatic Stress Disorder, Clinical Neurosciences Division, West Haven, CT, USA
- VA Connecticut Healthcare System, West Haven, CT, USA
- Department of Psychiatry, Yale School of Medicine, New Haven, CT, USA
| | - Anthony S Zannas
- Carolina Stress Initiative, University of North Carolina at Chapel Hill, NC, Chapel Hill, USA
- Department of Genetics, University of North Carolina at Chapel Hill, NC, Chapel Hill, USA
- Department of Psychiatry, University of North Carolina at Chapel Hill, NC, Chapel Hill, USA
- Institute for Trauma Recovery, University of North Carolina at Chapel Hill, NC, Chapel Hill, USA
| | - Xiang Zhao
- Department of Biostatistics, Boston University School of Public Health, Boston, MA, USA
| | - Allison E Aiello
- Robert N. Butler Columbia Aging Center, Department of Epidemiology, Columbia University, New York, NY, USA
| | - Allison E Ashley-Koch
- Duke Molecular Physiology Institute, Duke University Medical Center, Durham, NC, USA
| | - Diana Avetyan
- Department of Psychiatry, University of California San Diego, La Jolla, CA, San Diego, USA
| | - Dewleen G Baker
- Department of Psychiatry, University of California San Diego, La Jolla, CA, San Diego, USA
- Center of Excellence for Stress and Mental Health, Veterans Affairs San Diego Healthcare System, San Diego, CA, USA
- Psychiatry Service, Veterans Affairs San Diego Healthcare System, San Diego, CA, USA
| | - Jean C Beckham
- Department of Psychiatry and Behavioral Sciences, Duke University School of Medicine, Durham, NC, USA
- Durham VA Health Care System, Researcg, Durham, NC, USA
- Genetics Research Laboratory, VA Mid-Atlantic Mental Illness Research Education, and Clinical Center (MIRECC), Durham, NC, USA
| | - Marco P Boks
- Department of Psychiatry, Brain Center University Medical Center Utrecht, Utrecht, UT, NL, Netherlands
| | - Leslie A Brick
- Department of Psychiatry and Human Behavior, Alpert Medical School of Brown University, Providence, RI, USA
| | - Evelyn Bromet
- Epidemiology Research Group, State University of New York at Stony Brook, Stony Brook, NY, USA
| | - Frances A Champagne
- Department of Psychology, The University of Texas at Austin, Austin, TX, USA
| | - Chia-Yen Chen
- Biogen Inc, Translational Sciences, Cambridge, MA, USA
| | - Shareefa Dalvie
- Department of Pathology, University of Cape Town, Western Province, Cape Town, ZA, South Africa
- Division of Human Genetics, University of Cape Town, Western Province, Cape Town, ZA, South Africa
| | - Michelle F Dennis
- Department of Psychiatry and Behavioral Sciences, Duke University School of Medicine, Durham, NC, USA
- Durham VA Health Care System, Researcg, Durham, NC, USA
- Genetics Research Laboratory, VA Mid-Atlantic Mental Illness Research Education, and Clinical Center (MIRECC), Durham, NC, USA
| | - Segun Fatumo
- MRC/UVRI and London School of Hygiene and Tropical Medicine, The African Computational Genomics (TACG) Research Group, Entebbe, Wakiso, Uganda
| | - Catherine Fortier
- Department of Psychiatry, Harvard Medical School, Boston, MA, USA
- Translational Research Center for TBI and Stress Disorders (TRACTS)/Geriatric Research Education and Clinical Center (GRECC), VA Boston Healthcare System, Boston, MA, USA
| | - Sandro Galea
- School of Public Health, Boston University, Boston, MA, USA
| | - Melanie E Garrett
- Duke Molecular Physiology Institute, Duke University Medical Center, Durham, NC, USA
| | - Elbert Geuze
- Brain Research and Innovation Centre, Netherlands Ministry of Defence, Utrecht, UT, NL, Netherlands
- Department of Psychiatry, UMC Utrecht Brain Center Rudolf Magnus, Utrecht, UT, Netherlands
| | - Gerald Grant
- Department of Neurosurgery, Duke University School of Medicine, Durham, NC, USA
| | - Michael A Hauser
- Department of Medicine, Duke University School of Medicine, Durham, NC, USA
| | - Jasmeet P Hayes
- Department of Psychology, The Ohio State University, Columbus, OH, USA
| | - Sian M J Hemmings
- Department of Psychiatry, Faculty of Medicine and Health Sciences, Stellenbosch University, Western Cape, Cape Town, ZA, South Africa
- SAMRC Genomics of Brain Disorders Research Unit, Stellenbosch University, Western Cape, Cape Town, ZA, South Africa
| | - Bertrand Russel Huber
- Department of Neurology, Boston University School of Medicine, Boston, MA, USA
- Pathology and Laboratory Medicine, VA Boston Healthcare System, Boston, MA, USA
| | - Aarti Jajoo
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Psychiatry, Harvard Medical School, Boston, MA, USA
- McLean Hospital, Belmont, MA, USA
| | - Stefan Jansen
- College of Medicine and Health Sciences, University of Rwanda, Kigali, RW, Rwanda
| | - Ronald C Kessler
- Department of Health Care Policy, Harvard Medical School, Boston, MA, USA
| | - Nathan A Kimbrel
- Department of Psychiatry and Behavioral Sciences, Duke University School of Medicine, Durham, NC, USA
- Genetics Research Laboratory, VA Mid-Atlantic Mental Illness Research Education, and Clinical Center (MIRECC), Durham, NC, USA
- Mental Health Service Line, Durham VA Health Care System, Durham, NC, USA
| | - Anthony P King
- Institute for Behavioral Medicine Research, The Ohio State University College of Medicine, Columbus, OH, USA
- Psychiatry & Behavioral Health, The Ohio State University College of Medicine, Columbus, OH, USA
| | - Joel E Kleinman
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Lieber Institute for Brain Development, Baltimore, MD, USA
| | - Nastassja Koen
- Department of Psychiatry & Mental Health, University of Cape Town, Western Province, Cape Town, ZA, South Africa
- Neuroscience Institute, University of Cape Town, Western Province, Cape Town, ZA, South Africa
- SA MRC Unit on Risk & Resilience in Mental Disorders, University of Cape Town, Western Province, Cape Town, ZA, South Africa
| | - Karestan C Koenen
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Psychiatric and Neurodevelopmental Genetics Unit (PNGU), Massachusetts General Hospital, Boston, MA, USA
| | - Pei-Fen Kuan
- Department of Applied Mathematics and Statistics, Stony Brook University, Stony Brook, NY, USA
| | - Israel Liberzon
- Department of Psychiatry and Behavioral Sciences, Texas A&M University College of Medicine, Bryan, TX, USA
| | - Sarah D Linnstaedt
- Department of Anesthesiology, University of North Carolina at Chapel Hill, NC, Chapel Hill, USA
- UNC Institute for Trauma Recovery, University of North Carolina at Chapel Hill, NC, Chapel Hill, USA
| | - Adriana Lori
- Department of Psychiatry and Behavioral Sciences, Emory University, Atlanta, GA, USA
| | - Benjamin J Luft
- Department of Medicine, Stony Brook University, Stony Brook, NY, USA
| | - Jurjen J Luykx
- Amsterdam Neuroscience Research Institute Stress & Sleep Program, Amsterdam University Medical Center, Amsterdam, NH, Netherlands
- Amsterdam Public Health Research Institute, Mental Health Program, Amsterdam University Medical Center, Amsterdam, NH, Netherlands
- Department of Psychiatry, Amsterdam University Medical Center, Amsterdam, NH, Netherlands
| | - Christine E Marx
- Department of Psychiatry and Behavioral Sciences, Duke University School of Medicine, Durham, NC, USA
- Durham VA Health Care System, Durham, NC, USA
- VA Mid-Atlantic Mental Illness Research Education, and Clinical Center (MIRECC), Durham, NC, USA
| | - Samuel A McLean
- Department of Psychiatry, UNC Institute for Trauma Recovery, NC, Chapel Hill, USA
| | - Divya Mehta
- Centre for Genomics and Personalised Health, Queensland University of Technology, Kelvin Grove, QLD, AU, Brisbane, Australia
- School of Biomedical Sciences, Queensland University of Technology, Kelvin Grove, QLD, AU, Brisbane, Australia
| | | | - Mark W Miller
- Biomedical Genetics & Psychiatry, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
- National Center for PTSD, VA Boston Healthcare System, Boston, MA, USA
| | - Mary S Mufford
- Department of Psychiatry and Mental Health, University of Cape Town, Western Province, Cape Town, ZA, South Africa
| | - Clarisse Musanabaganwa
- Research Innovation and Data Science Division, Rwanda Biomedical Center, Kigali, Rwanda
- Center of Human Genetics, University of Rwanda, Kigali, RW, Rwanda
| | - Jean Mutabaruka
- Department of Clinical Psychology, University of Rwanda, Huye, RW, Rwanda
| | - Leon Mutesa
- College of Medicine and Health Sciences, University of Rwanda, Kigali, RW, Rwanda
- Center for Human Genetics, University of Rwanda, Kigali, RW, Rwanda
| | - Charles B Nemeroff
- Department of Psychology, The University of Texas at Austin, Austin, TX, USA
- Department of Psychiatry and Behavioral Sciences, The University of Texas at Austin, Austin, TX, USA
| | - Nicole R Nugent
- Department of Emergency Medicine, Alpert Brown Medical School, Providence, RI, USA
- Department of Pediatrics, Alpert Brown Medical School, Providence, RI, USA
- Department of Psychiatry and Human Behavior, Alpert Brown Medical School, Providence, RI, USA
| | - Holly K Orcutt
- Department of Psychology, Northern Illinois University, DeKalb, IL, USA
| | - Xue-Jun Qin
- Duke Molecular Physiology Institute, Duke University, Durham, NC, USA
| | - Sheila A M Rauch
- Department of Psychiatry & Behavioral Sciences, Emory University, Atlanta, GA, USA
- Joseph Maxwell Cleland Atlanta Veterans Affairs Healthcare System, Atlanta, GA, USA
| | - Kerry J Ressler
- Department of Psychiatry, Harvard Medical School, Boston, MA, USA
- McLean Hospital, Belmont, MA, USA
- Department of Psychiatry and Behavioral Sciences, Emory University, Atlanta, GA, USA
| | - Victoria B Risbrough
- Department of Psychiatry, University of California San Diego, La Jolla, CA, San Diego, USA
- Center of Excellence for Stress and Mental Health, Veterans Affairs San Diego Healthcare System, San Diego, CA, USA
- Research Service, Veterans Affairs San Diego Healthcare System, San Diego, CA, USA
| | | | - Bart P F Rutten
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience, Maastricht Universitair Medisch Centrum, Maastricht, Limburg, NL, Netherlands
| | - Soraya Seedat
- Department of Psychiatry, Faculty of Medicine and Health Sciences, Stellenbosch University, Western Cape, Cape Town, ZA, South Africa
- SA MRC Extramural Genomics of Brain Disorders Research Unit, Stellenbosch University, Western Cape, Cape Town, ZA, South Africa
| | - Dan J Stein
- Department of Psychiatry & Mental Health, University of Cape Town, Western Province, Cape Town, ZA, South Africa
- Neuroscience Institute, University of Cape Town, Western Province, Cape Town, ZA, South Africa
- SA MRC Unit on Risk & Resilience in Mental Disorders, University of Cape Town, Western Province, Cape Town, ZA, South Africa
| | - Murray B Stein
- Department of Psychiatry, University of California San Diego, La Jolla, CA, San Diego, USA
- Psychiatry Service, Veterans Affairs San Diego Healthcare System, San Diego, CA, USA
- School of Public Health, University of California San Diego, CA, La Jolla, USA
| | - Sylvanus Toikumo
- Department of Psychiatry, Faculty of Medicine and Health Sciences, Stellenbosch University, Western Cape, Cape Town, ZA, South Africa
- SA MRC Genomics of Brain Disorders Research Unit, Stellenbosch University, Western Cape, Cape Town, ZA, South Africa
| | - Robert J Ursano
- Department of Psychiatry, Center for the Study of Traumatic Stress, Uniformed Services University, Bethesda, MD, USA
| | - Annette Uwineza
- College of Medicine and Health Sciences, University of Rwanda, Kigali, Rwanda
| | - Mieke H Verfaellie
- Department of Psychiatry, Boston University School of Medicine, Boston, MA, USA
- Memory Disorders Research Center, VA Boston Healthcare System, Boston, MA, USA
| | - Eric Vermetten
- Department of Psychiatry, Leiden University Medical Center, Leiden, ZH, NL, Netherlands
- Department of Psychiatry, New York University School of Medicine, New York, NY, USA
| | - Christiaan H Vinkers
- Department of Anatomy and Neurosciences, Amsterdam UMC Location Vrije Universiteit Amsterdam, Amsterdam, Holland, Netherlands
- Department of Psychiatry, Amsterdam, UMC Location Vrije Universiteit Amsterdam, Amsterdam, Holland, Netherlands
- Amsterdam University Medical Center, Amsterdam Neuroscience Research Institute, Stress & Sleep Program, MoodPsychosisAmsterdam, Holland, AnxietyNL, Netherlands
| | - Erin B Ware
- Survey Research Center, University of Michigan, Ann Arbor, MI, USA
| | - Derek E Wildman
- College of Public Health, University of South Florida, Tampa, FL, USA
- Genomics Program, University of South Florida, Tampa, FL, USA
| | - Erika J Wolf
- National Center for PTSD, VA Boston Healthcare System, Boston, MA, USA
- Department of Psychiatry, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
| | - Ross McD Young
- School of Clinical Sciences, Queensland University of Technology, Kelvin Grove, QLD, AU, Brisbane, Australia
- University of the Sunshine Coast, The Chancellory Sippy Downs, QLD, AU, Buderim, Australia
| | - Ying Zhao
- Institute for Trauma Recovery, University of North Carolina at Chapel Hill, NC, Chapel Hill, USA
- Department of Anesthesiology, University of North Carolina at Chapel Hill, NC, Chapel Hill, USA
| | - Leigh L van den Heuvel
- Department of Psychiatry, Faculty of Medicine and Health Sciences, Stellenbosch University, Western Cape, Cape Town, ZA, South Africa
- SA MRC Extramural Genomics of Brain Disorders Research Unit, Stellenbosch University, Western Cape, Cape Town, ZA, South Africa
| | - Monica Uddin
- University of South Florida College of Public Health, Genomics Program, Tampa, FL, USA
| | - Caroline M Nievergelt
- Department of Psychiatry, University of California San Diego, La Jolla, CA, San Diego, USA
- Center of Excellence for Stress and Mental Health, Veterans Affairs San Diego Healthcare System, San Diego, CA, USA
- Research Service, Veterans Affairs San Diego Healthcare System, San Diego, CA, USA
| | - Alicia K Smith
- Department of Gynecology and Obstetrics, Emory University, Atlanta, GA, USA
- Department of Psychiatry and Behavioral Sciences, Emory University, Atlanta, GA, USA
- Department of Human Genetics, Emory University, Atlanta, GA, USA
| | - Mark W Logue
- Department of Biostatistics, Boston University School of Public Health, Boston, MA, USA.
- Biomedical Genetics & Psychiatry, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA.
- National Center for PTSD, VA Boston Healthcare System, Boston, MA, USA.
| |
Collapse
|
46
|
Liu C, Joehanes R, Ma J, Xie J, Yang J, Wang M, Huan T, Hwang SJ, Wen J, Sun Q, Cumhur DY, Heard-Costa NL, Orchard P, Carson AP, Raffield LM, Reiner A, Li Y, O'Connor G, Murabito JM, Munson P, Levy D. Integrating Whole Genome and Transcriptome Sequencing to Characterize the Genetic Architecture of Isoform Variation and its Implications for Health and Disease. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.12.04.24318434. [PMID: 39677465 PMCID: PMC11643148 DOI: 10.1101/2024.12.04.24318434] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2024]
Abstract
We created a comprehensive whole blood splice variation quantitative trait locus (sQTL) resource by analyzing isoform expression ratio (isoform-to-gene) in Framingham Heart Study (FHS) participants (discovery: n=2,622; validation: n=1,094) with whole genome (WGS) and transcriptome sequencing (RNA-seq) data. External replication was conducted using WGS and RNA-seq from the Jackson Heart Study (JHS, n=1,020). We identified over 3.5 million cis -sQTL-isoform pairs ( p <5e-8), comprising 1,176,624 cis -sQTL variants and 10,883 isoform transcripts from 4,971 sGenes, with significant change in isoform-to-gene ratio due to allelic variation. We validated 61% of these pairs in the FHS validation sample ( p <1e-4). External validation ( p <1e-4) in JHS for the top 10,000 and 100,000 most significant cis -sQTL-isoform pairs was 88% and 69%, respectively, while overall pairs validated at 23%. For 20% of cis -sQTLs in the FHS discovery sample, allelic variation did not significantly correlate with overall gene expression. sQTLs are enriched in splice donor and acceptor sites, as well as in GWAS SNPs, methylation QTLs, and protein QTLs. We detailed several sentinel cis -sQTLs influencing alternative splicing, with potential causal effects on cardiovascular disease risk. Notably, rs12898397 (T>C) affects splicing of ULK3 , lowering levels of the full-length transcript ENST00000440863.7 and increasing levels of the truncated transcript ENST00000569437.5, encoding proteins of different lengths. Mendelian randomization analysis demonstrated that a lower ratio of the full-length isoform is causally associated with lower diastolic blood pressure and reduced lymphocyte percentages. This sQTL resource provides valuable insights into how transcriptomic variation may influence health outcomes.
Collapse
|
47
|
McDonnell E, Orr SE, Barter MJ, Rux D, Brumwell A, Wrobel N, Murphy L, Overman LM, Sorial AK, Young DA, Soul J, Rice SJ. The methylomic landscape of human articular cartilage development contains epigenetic signatures of osteoarthritis risk. Am J Hum Genet 2024; 111:2756-2772. [PMID: 39579763 PMCID: PMC11639090 DOI: 10.1016/j.ajhg.2024.10.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2024] [Revised: 10/21/2024] [Accepted: 10/25/2024] [Indexed: 11/25/2024] Open
Abstract
Increasing evidence is emerging to link age-associated complex musculoskeletal diseases, including osteoarthritis (OA), to developmental factors. Multiple studies have shown a functional role for DNA methylation in the genetic mechanisms of OA risk using articular cartilage samples taken from aged individuals, yet knowledge of temporal changes to the methylome during human cartilage development is limited. We quantified DNA methylation at ∼700,000 individual CpGs across the epigenome of developing human chondrocytes in 72 samples ranging from 7 to 21 post-conception weeks. We identified significant changes in 3% of all CpGs and >8,200 developmental differentially methylated regions. We further identified 24 loci at which OA genetic variants colocalize with methylation quantitative trait loci. Through integrating developmental and mature human chondrocyte datasets, we find evidence for functional effects exerted solely in development or throughout the life course. This will have profound impacts on future approaches to translating genetic pathways for therapeutic intervention.
Collapse
Affiliation(s)
- Euan McDonnell
- Computational Biology Facility, University of Liverpool, MerseyBio, Crown Street, Liverpool, UK
| | - Sarah E Orr
- Biosciences Institute, Newcastle University, Central Parkway, Newcastle upon Tyne, UK
| | - Matthew J Barter
- Biosciences Institute, Newcastle University, Central Parkway, Newcastle upon Tyne, UK
| | - Danielle Rux
- Orthopedic Surgery, UConn Health, Farmington, CT, USA
| | - Abby Brumwell
- Biosciences Institute, Newcastle University, Central Parkway, Newcastle upon Tyne, UK
| | - Nicola Wrobel
- Edinburgh Clinical Research Facility, University of Edinburgh, Edinburgh, UK
| | - Lee Murphy
- Edinburgh Clinical Research Facility, University of Edinburgh, Edinburgh, UK
| | - Lynne M Overman
- Human Developmental Biology Resource, Newcastle University, International Centre for Life, Central Parkway, Newcastle upon Tyne, UK
| | - Antony K Sorial
- Biosciences Institute, Newcastle University, Central Parkway, Newcastle upon Tyne, UK
| | - David A Young
- Biosciences Institute, Newcastle University, Central Parkway, Newcastle upon Tyne, UK
| | - Jamie Soul
- Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool, UK.
| | - Sarah J Rice
- Biosciences Institute, Newcastle University, Central Parkway, Newcastle upon Tyne, UK.
| |
Collapse
|
48
|
Ciantar J, Marttila S, Rajić S, Kostiniuk D, Mishra PP, Lyytikäinen LP, Mononen N, Kleber ME, März W, Kähönen M, Raitakari O, Lehtimäki T, Raitoharju E. Identification and functional characterisation of DNA methylation differences between East- and West-originating Finns. Epigenetics 2024; 19:2397297. [PMID: 39217505 PMCID: PMC11382697 DOI: 10.1080/15592294.2024.2397297] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Revised: 08/14/2024] [Accepted: 08/22/2024] [Indexed: 09/04/2024] Open
Abstract
Eastern and Western Finns show a striking difference in coronary heart disease-related mortality; genetics is a known contributor for this discrepancy. Here, we discuss the potential role of DNA methylation in mediating the discrepancy in cardiometabolic disease-risk phenotypes between the sub-populations. We used data from the Young Finns Study (n = 969) to compare the genome-wide DNA methylation levels of East- and West-originating Finns. We identified 21 differentially methylated loci (FDR < 0.05; Δβ >2.5%) and 7 regions (smoothed FDR < 0.05; CpGs ≥ 5). Methylation at all loci and regions associates with genetic variants (p < 5 × 10-8). Independently of genetics, methylation at 11 loci and 4 regions associates with transcript expression, including genes encoding zinc finger proteins. Similarly, methylation at 5 loci and 4 regions associates with cardiometabolic disease-risk phenotypes including triglycerides, glucose, cholesterol, as well as insulin treatment. This analysis was also performed in LURIC (n = 2371), a German cardiovascular patient cohort, and results replicated for the association of methylation at cg26740318 and DMR_11p15 with diabetes-related phenotypes and methylation at DMR_22q13 with triglyceride levels. Our results indicate that DNA methylation differences between East and West Finns may have a functional role in mediating the cardiometabolic disease discrepancy between the sub-populations.
Collapse
Affiliation(s)
- Joanna Ciantar
- Molecular Epidemiology (MOLE), Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Saara Marttila
- Molecular Epidemiology (MOLE), Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
- Gerontology Research Center, Tampere University, Tampere, Finland
- Tays Research Services, Wellbeing Services County of Pirkanmaa, Tampere University Hospital, Tampere, Finland
| | - Sonja Rajić
- Molecular Epidemiology (MOLE), Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Daria Kostiniuk
- Molecular Epidemiology (MOLE), Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Pashupati P Mishra
- Department of Clinical Chemistry, Tays Research Services, Fimlab Laboratories, and Finnish Cardiovascular Research Center, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Leo-Pekka Lyytikäinen
- Department of Clinical Chemistry, Tays Research Services, Fimlab Laboratories, and Finnish Cardiovascular Research Center, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Nina Mononen
- Department of Clinical Chemistry, Tays Research Services, Fimlab Laboratories, and Finnish Cardiovascular Research Center, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Marcus E Kleber
- Vth Department of Medicine (Nephrology, Hypertensiology, Endocrinology, Diabetology, Rheumatology), Medical Faculty of Mannheim, Heidelberg University, Mannheim, Germany
- SYNLAB MVZ Humangenetik Mannheim, Mannheim, Germany
| | - Winfried März
- Vth Department of Medicine (Nephrology, Hypertensiology, Endocrinology, Diabetology, Rheumatology), Medical Faculty of Mannheim, Heidelberg University, Mannheim, Germany
- Synlab Academy, SYNLAB Holding Deutschland GmbH, Mannheim, Germany
| | - Mika Kähönen
- Department of Clinical Physiology, Tampere University Hospital and Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Olli Raitakari
- Centre for Population Health Research, University of Turku and Turku University Hospital, Turku, Finland
- Research Centre of Applied and Preventive Cardiovascular Medicine, University of Turku, Turku, Finland
- Department of Clinical Physiology and Nuclear Medicine, Turku University Hospital, Turku, Finland
| | - Terho Lehtimäki
- Department of Clinical Chemistry, Tays Research Services, Fimlab Laboratories, and Finnish Cardiovascular Research Center, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
- Finnish Cardiovascular Research Center Tampere, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Emma Raitoharju
- Molecular Epidemiology (MOLE), Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
- Finnish Cardiovascular Research Center Tampere, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
- Fimlab Laboratories, Tampere, Finland
| |
Collapse
|
49
|
Wang J, Hu J, Qin D, Han D, Hu J. A multi-omics Mendelian randomization identifies putatively causal genes and DNA methylation sites for asthma. World Allergy Organ J 2024; 17:101008. [PMID: 39720783 PMCID: PMC11667005 DOI: 10.1016/j.waojou.2024.101008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2024] [Revised: 08/02/2024] [Accepted: 11/12/2024] [Indexed: 12/26/2024] Open
Abstract
Background Asthma is a global chronic respiratory disease with complex pathogenesis. While current therapies offer some relief, they often fall short in effectively managing symptoms and preventing exacerbations for numerous patients. Thus, understanding its mechanisms and discovering new drug targets remains a pressing need for better treatment. Methods Using the GEO dataset, we screened differentially expressed genes (DEGs) in asthma patients' blood. Employing Summary Data-based Mendelian Randomization (SMR) and Two-Sample Mendelian Randomization (TSMR), we pinpointed asthma causal genes, causal DNA methylation sites, and methylation sites affecting gene expression, cross validated with at least 2 large-scale GWAS from each source. We utilized colocalization for genetic associations, meta-analysis for data integration, two-step MR for methylation-gene-asthma mediation mechanism. Druggability was evaluated using Open Target, virtual screening, and docking. Results Among the 954 DEGs found in asthma patients' blood, increased expression of CEP95 (discovery, OR_SMR = 0.94, 95% CI: 0.91-0.97), RBM6 (discovery, OR_SMR = 0.97, 95% CI: 0.95-0.99), and ITPKB (discovery, OR_SMR = 0.82, 95% CI: 0.74-0.92) in the blood decreased the risk of asthma, higher levels of HOXB-AS1 (discovery, OR_SMR = 1.05, 95% CI: 1.03-1.07), ETS1 (discovery, OR_SMR = 1.62, 95% CI: 1.29-2.04), and JAK2 (discovery, OR_SMR = 1.13, 95% CI: 1.06-1.21) in the blood increased the risk of asthma. Additionally, a total of 8 methylation sites on ITPKB, ETS1, and JAK2 were identified to influence asthma. An increase in methylation at site cg16265553 raised the risk of asthma partially by suppressing ITPKB expression. Similarly, increased methylation at cg13661497 reduced the asthma risk totally by suppressing JAK2 expression. The impact of CEP95, HOXB-AS1, and RBM6 expressions on asthma was further confirmed in lung tissues. Except for HOXB-AS1, all the other genes were potential druggable targets. Conclusion Our study highlighted that specific gene expressions and methylation sites significantly influence asthma risk and revealed a potential methylation-to-gene-to-asthma mechanism. This provided pivotal evidence for future targeted functional studies and the development of preventive and treatment strategies.
Collapse
Affiliation(s)
- Jia Wang
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang, China
| | - Jinxin Hu
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang, China
- Liaoning Key Laboratory of Research and Application of Animal Models for Environmental and Metabolic Diseases, Medical Research Center, Shengjing Hospital of China Medical University, Shenyang, China
| | - Dan Qin
- Research Center of Traditional Chinese Medicine Information Engineering, Beijing University of Chinese Medicine, Beijing, China
| | - Dan Han
- Department of Neonatology, The First Hospital of China Medical University, Shenyang, China
| | - Jiapeng Hu
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang, China
| |
Collapse
|
50
|
Zhang W, Young JI, Gomez L, Schmidt MA, Lukacsovich D, Varma A, Chen XS, Kunkle B, Martin ER, Wang L. Critical evaluation of the reliability of DNA methylation probes on the Illumina MethylationEPIC v1.0 BeadChip microarrays. Epigenetics 2024; 19:2333660. [PMID: 38564759 PMCID: PMC10989698 DOI: 10.1080/15592294.2024.2333660] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2023] [Accepted: 03/18/2024] [Indexed: 04/04/2024] Open
Abstract
DNA methylation (DNAm) plays a crucial role in a number of complex diseases. However, the reliability of DNAm levels measured using Illumina arrays varies across different probes. Previous research primarily assessed probe reliability by comparing duplicate samples between the 450k-450k or 450k-EPIC platforms, with limited investigations on Illumina EPIC v1.0 arrays. We conducted a comprehensive assessment of the EPIC v1.0 array probe reliability using 69 blood DNA samples, each measured twice, generated by the Alzheimer's Disease Neuroimaging Initiative study. We observed higher reliability in probes with average methylation beta values of 0.2 to 0.8, and lower reliability in type I probes or those within the promoter and CpG island regions. Importantly, we found that probe reliability has significant implications in the analyses of Epigenome-wide Association Studies (EWAS). Higher reliability is associated with more consistent effect sizes in different studies, the identification of differentially methylated regions (DMRs) and methylation quantitative trait locus (mQTLs), and significant correlations with downstream gene expression. Moreover, blood DNAm measurements obtained from probes with higher reliability are more likely to show concordance with brain DNAm measurements. Our findings, which provide crucial reliability information for probes on the EPIC v1.0 array, will serve as a valuable resource for future DNAm studies.
Collapse
Affiliation(s)
- Wei Zhang
- Division of Biostatistics, Department of Public Health Sciences, University of Miami, Miller School of Medicine, Miami, FL, USA
| | - Juan I. Young
- Dr. John T MacDonald Foundation Department of Human Genetics, University of Miami, Miller School of Medicine, Miami, FL, USA
- John P. Hussman Institute for Human Genomics, the University of Miami Miller School of Medicine, Miami, FL, USA
| | - Lissette Gomez
- John P. Hussman Institute for Human Genomics, the University of Miami Miller School of Medicine, Miami, FL, USA
| | - Michael A. Schmidt
- John P. Hussman Institute for Human Genomics, the University of Miami Miller School of Medicine, Miami, FL, USA
| | - David Lukacsovich
- Division of Biostatistics, Department of Public Health Sciences, University of Miami, Miller School of Medicine, Miami, FL, USA
| | - Achintya Varma
- John P. Hussman Institute for Human Genomics, the University of Miami Miller School of Medicine, Miami, FL, USA
| | - X. Steven Chen
- Division of Biostatistics, Department of Public Health Sciences, University of Miami, Miller School of Medicine, Miami, FL, USA
- Sylvester Comprehensive Cancer Center, University of Miami, Miller School of Medicine, Miami, FL, USA
| | - Brian Kunkle
- Dr. John T MacDonald Foundation Department of Human Genetics, University of Miami, Miller School of Medicine, Miami, FL, USA
- John P. Hussman Institute for Human Genomics, the University of Miami Miller School of Medicine, Miami, FL, USA
| | - Eden R. Martin
- Dr. John T MacDonald Foundation Department of Human Genetics, University of Miami, Miller School of Medicine, Miami, FL, USA
- John P. Hussman Institute for Human Genomics, the University of Miami Miller School of Medicine, Miami, FL, USA
| | - Lily Wang
- Division of Biostatistics, Department of Public Health Sciences, University of Miami, Miller School of Medicine, Miami, FL, USA
- Dr. John T MacDonald Foundation Department of Human Genetics, University of Miami, Miller School of Medicine, Miami, FL, USA
- John P. Hussman Institute for Human Genomics, the University of Miami Miller School of Medicine, Miami, FL, USA
- Sylvester Comprehensive Cancer Center, University of Miami, Miller School of Medicine, Miami, FL, USA
| |
Collapse
|