1
|
O’Sullivan J, Patel S, Leventhal GE, Fitzgerald RS, Laserna-Mendieta EJ, Huseyin CE, Konstantinidou N, Rutherford E, Lavelle A, Dabbagh K, DeSantis TZ, Shanahan F, Temko A, Iwai S, Claesson MJ. Host-microbe multi-omics and succinotype profiling have prognostic value for future relapse in patients with inflammatory bowel disease. Gut Microbes 2025; 17:2450207. [PMID: 39812341 PMCID: PMC11740686 DOI: 10.1080/19490976.2025.2450207] [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: 07/16/2024] [Revised: 11/07/2024] [Accepted: 01/02/2025] [Indexed: 01/16/2025] Open
Abstract
Crohn's disease (CD) and ulcerative colitis (UC) are chronic relapsing inflammatory bowel disorders (IBD), the pathogenesis of which is uncertain but includes genetic susceptibility factors, immune-mediated tissue injury and environmental influences, most of which appear to act via the gut microbiome. We hypothesized that host-microbe alterations could be used to prognostically stratify patients experiencing relapses up to four years after endoscopy. We therefore examined multiple omics data, including published and new datasets, generated from paired inflamed and non-inflamed mucosal biopsies from 142 patients with IBD (54 CD; 88 UC) and from 34 control (non-diseased) biopsies. The relapse-predictive potential of 16S rRNA gene and transcript amplicons (standing and active microbiota) were investigated along with host transcriptomics, epigenomics and genetics. While standard single-omics analysis could not distinguish between patients who relapsed and those that remained in remission within four years of colonoscopy, we did find an association between the number of flares and a patient's succinotype. Our multi-omics machine learning approach was also able to predict relapse when combining features from the microbiome and human host. Therefore multi-omics, rather than single omics, better predicts relapse within 4 years of colonoscopy, while a patient's succinotype is associated with a higher frequency of relapses.
Collapse
Affiliation(s)
- Jill O’Sullivan
- School of Microbiology, University College Cork, Cork, Ireland
- APC Microbiome Ireland, University College Cork, Cork, Ireland
- SFI Centre for Research Training in Genomics Data Science, University of Galway, Galway, Ireland
| | - Shriram Patel
- School of Microbiology, University College Cork, Cork, Ireland
- APC Microbiome Ireland, University College Cork, Cork, Ireland
- SeqBiome Ltd, Cork, Ireland
| | | | - Rachel S. Fitzgerald
- School of Microbiology, University College Cork, Cork, Ireland
- APC Microbiome Ireland, University College Cork, Cork, Ireland
| | - Emilio J. Laserna-Mendieta
- School of Microbiology, University College Cork, Cork, Ireland
- APC Microbiome Ireland, University College Cork, Cork, Ireland
| | - Chloe E. Huseyin
- School of Microbiology, University College Cork, Cork, Ireland
- APC Microbiome Ireland, University College Cork, Cork, Ireland
| | - Nina Konstantinidou
- School of Microbiology, University College Cork, Cork, Ireland
- Department of Informatics, Second Genome Inc, South San Francisco, California, USA
| | - Erica Rutherford
- Department of Informatics, Second Genome Inc, South San Francisco, California, USA
| | - Aonghus Lavelle
- APC Microbiome Ireland, University College Cork, Cork, Ireland
- Department of Anatomy and Neuroscience, University College Cork, Cork, County Cork, Ireland
| | - Karim Dabbagh
- Department of Informatics, Second Genome Inc, South San Francisco, California, USA
| | - Todd Z. DeSantis
- Department of Informatics, Second Genome Inc, South San Francisco, California, USA
| | - Fergus Shanahan
- APC Microbiome Ireland, University College Cork, Cork, Ireland
- Department of Medicine, University College Cork, Cork, Ireland
| | - Andriy Temko
- Department of Electrical and Electronic Engineering, University College Cork, Cork, Ireland
| | - Shoko Iwai
- Department of Informatics, Second Genome Inc, South San Francisco, California, USA
| | - Marcus J. Claesson
- School of Microbiology, University College Cork, Cork, Ireland
- APC Microbiome Ireland, University College Cork, Cork, Ireland
| |
Collapse
|
2
|
Fernández-Pérez I, Jiménez-Balado J, Macias-Gómez A, Suárez-Pérez A, Vallverdú-Prats M, Pérez-Giraldo A, Viles-García M, Peris-Subiza J, Vidal-Notari S, Giralt-Steinhauer E, Guisado-Alonso D, Esteller M, Rodriguez-Campello A, Jiménez-Conde J, Ois A, Cuadrado-Godia E. Blood DNA Methylation Analysis Reveals a Distinctive Epigenetic Signature of Vasospasm in Aneurysmal Subarachnoid Hemorrhage. Transl Stroke Res 2025; 16:715-727. [PMID: 38649590 DOI: 10.1007/s12975-024-01252-x] [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: 02/27/2024] [Revised: 03/28/2024] [Accepted: 04/06/2024] [Indexed: 04/25/2024]
Abstract
Vasospasm is a potentially preventable cause of poor prognosis in patients with aneurysmal subarachnoid hemorrhage (aSAH). Epigenetics might provide insight on its molecular mechanisms. We aimed to analyze the association between differential DNA methylation (DNAm) and development of vasospasm. We conducted an epigenome-wide association study in 282 patients with aSAH admitted to our hospital. DNAm was assessed with the EPIC Illumina chip (> 850 K CpG sites) in whole-blood samples collected at hospital admission. We identified differentially methylated positions (DMPs) at the CpG level using Cox regression models adjusted for potential confounders, and then we used the DMP results to find differentially methylated regions (DMRs) and enriched biological pathways. A total of 145 patients (51%) experienced vasospasm. In the DMP analysis, we identified 31 CpGs associated with vasospasm at p-value < 10-5. One of them (cg26189827) was significant at the genome-wide level (p-value < 10-8), being hypermethylated in patients with vasospasm and annotated to SUGCT gene, mainly expressed in arteries. Region analysis revealed 13 DMRs, some of them annotated to interesting genes such as POU5F1, HLA-DPA1, RUFY1, and CYP1A1. Functional enrichment analysis showed the involvement of biological processes related to immunity, inflammatory response, oxidative stress, endothelial nitric oxide, and apoptosis. Our findings show, for the first time, a distinctive epigenetic signature of vasospasm in aSAH, establishing novel links with essential biological pathways, including inflammation, immune responses, and oxidative stress. Although further validation is required, our results provide a foundation for future research into the complex pathophysiology of vasospasm.
Collapse
Affiliation(s)
- Isabel Fernández-Pérez
- Neurology Department, Hospital del Mar, Barcelona, Catalunya, Spain
- Neurovascular Research Group, Hospital del Mar Medical Research Institute, C/Dr. Aiguader, 88, 08003, Barcelona, Catalunya, Spain
| | - Joan Jiménez-Balado
- Neurovascular Research Group, Hospital del Mar Medical Research Institute, C/Dr. Aiguader, 88, 08003, Barcelona, Catalunya, Spain.
| | - Adrià Macias-Gómez
- Neurology Department, Hospital del Mar, Barcelona, Catalunya, Spain
- Neurovascular Research Group, Hospital del Mar Medical Research Institute, C/Dr. Aiguader, 88, 08003, Barcelona, Catalunya, Spain
| | - Antoni Suárez-Pérez
- Neurology Department, Hospital del Mar, Barcelona, Catalunya, Spain
- Neurovascular Research Group, Hospital del Mar Medical Research Institute, C/Dr. Aiguader, 88, 08003, Barcelona, Catalunya, Spain
| | - Marta Vallverdú-Prats
- Neurovascular Research Group, Hospital del Mar Medical Research Institute, C/Dr. Aiguader, 88, 08003, Barcelona, Catalunya, Spain
| | | | - Marc Viles-García
- Neuroradiology Department, Hospital del Mar, Barcelona, Catalunya, Spain
| | | | | | - Eva Giralt-Steinhauer
- Neurology Department, Hospital del Mar, Barcelona, Catalunya, Spain
- Neurovascular Research Group, Hospital del Mar Medical Research Institute, C/Dr. Aiguader, 88, 08003, Barcelona, Catalunya, Spain
- Pompeu Fabra University, Barcelona, Catalunya, Spain
| | - Daniel Guisado-Alonso
- Neurology Department, Hospital del Mar, Barcelona, Catalunya, Spain
- Neurovascular Research Group, Hospital del Mar Medical Research Institute, C/Dr. Aiguader, 88, 08003, Barcelona, Catalunya, Spain
| | - Manel Esteller
- Cancer Epigenetics Group, Research Institute Against Leukemia Josep Carreras, Badalona, Catalunya, Spain
- Physiological Sciences Department, School of Medicine and Health Sciences, University of Barcelona, Barcelona, Catalunya, Spain
| | - Ana Rodriguez-Campello
- Neurology Department, Hospital del Mar, Barcelona, Catalunya, Spain
- Neurovascular Research Group, Hospital del Mar Medical Research Institute, C/Dr. Aiguader, 88, 08003, Barcelona, Catalunya, Spain
- Pompeu Fabra University, Barcelona, Catalunya, Spain
| | - Jordi Jiménez-Conde
- Neurology Department, Hospital del Mar, Barcelona, Catalunya, Spain
- Neurovascular Research Group, Hospital del Mar Medical Research Institute, C/Dr. Aiguader, 88, 08003, Barcelona, Catalunya, Spain
- Pompeu Fabra University, Barcelona, Catalunya, Spain
| | - Angel Ois
- Neurology Department, Hospital del Mar, Barcelona, Catalunya, Spain
- Neurovascular Research Group, Hospital del Mar Medical Research Institute, C/Dr. Aiguader, 88, 08003, Barcelona, Catalunya, Spain
- Pompeu Fabra University, Barcelona, Catalunya, Spain
| | - Elisa Cuadrado-Godia
- Neurology Department, Hospital del Mar, Barcelona, Catalunya, Spain
- Neurovascular Research Group, Hospital del Mar Medical Research Institute, C/Dr. Aiguader, 88, 08003, Barcelona, Catalunya, Spain
- Pompeu Fabra University, Barcelona, Catalunya, Spain
| |
Collapse
|
3
|
Li Y, Goodrich JM, Peterson KE, Song PX, Luo L. Uncertainty Quantification in Epigenetic Clocks via Conformalized Quantile Regression. Genet Epidemiol 2025; 49:e70008. [PMID: 40145332 PMCID: PMC11948180 DOI: 10.1002/gepi.70008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2024] [Revised: 01/20/2025] [Accepted: 02/21/2025] [Indexed: 03/28/2025]
Abstract
DNA methylation (DNAm) is a chemical modification of DNA that can be influenced by various factors, including age, the environment, and lifestyle. An epigenetic clock is a predictive tool that measures biological age based on DNAm levels. It can provide insights into an individual's biological age, which may differ from their chronological age. This difference, known as the epigenetic age acceleration, may reflect health status and the risk for age-related diseases. Moreover, epigenetic clocks are used in studies of aging to assess the effectiveness of antiaging interventions and to understand the underlying mechanisms of aging and disease. Various epigenetic clocks have been developed using samples from different populations, tissues, and cell types, typically by training high-dimensional linear regression models with an elastic net penalty. While these models can predict mean biological age based on DNAm with high precision, there is a lack of uncertainty quantification which is important for interpreting the precision of age estimations and for clinical decision-making. To understand the distribution of a biological age clock beyond its mean, we propose a general pipeline for training epigenetic clocks, based on an integration of high-dimensional quantile regression and conformal prediction, to effectively reveal population heterogeneity and construct prediction intervals. Our approach produces adaptive prediction intervals not only achieving nominal coverage but also accounting for the inherent variability across individuals. By using the data collected from 728 blood samples in 11 DNAm data sets from children, we find that our quantile regression-based prediction intervals are narrower than those derived from conventional mean regression-based epigenetic clocks. This observation demonstrates an improved statistical efficiency over the existing pipeline for training epigenetic clocks. In addition, the resulting intervals have a synchronized varying pattern to age acceleration, effectively revealing cellular evolutionary heterogeneity in age patterns in different developmental stages during individual childhoods and adolescent cohort. Our findings suggest that conformalized high-dimensional quantile regression can produce valid prediction intervals and uncover underlying population heterogeneity. Although our methodology focuses on the distribution of measures of biological aging in children, it is applicable to a broader range of age groups to improve understanding of epigenetic age beyond the mean. This inference-based toolbox could provide valuable insights for future applications of epigenetic interventions for age-related diseases.
Collapse
Affiliation(s)
- Yanping Li
- School of Statistics and Data ScienceNankai UniversityTianjinChina
| | - Jaclyn M. Goodrich
- Department of Environmental Health SciencesUniversity of MichiganAnn ArborMichiganUSA
| | - Karen E. Peterson
- Department of Nutritional SciencesUniversity of MichiganAnn ArborMichiganUSA
| | - Peter X.‐K. Song
- Department of BiostatisticsUniversity of MichiganAnn ArborMichiganUSA
| | - Lan Luo
- Department of Biostatistics and EpidemiologyRutgers UniversityPiscatawayNew JerseyUSA
| |
Collapse
|
4
|
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
|
5
|
Przybylowicz PK, Klepinowski T, Taryma-Leśniak O, Bińkowski J, Wojdacz TK. Evaluation of methylation changes in blood cells of COVID-19 patients as a biomarker of severity of the infection. BMC Infect Dis 2025; 25:778. [PMID: 40450204 DOI: 10.1186/s12879-025-11181-1] [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: 06/18/2024] [Accepted: 05/28/2025] [Indexed: 06/03/2025] Open
Abstract
BACKGROUND A number of studies have shown that methylomes of blood cells of COVID-19 patients display infection-related methylation changes. Those methylation abnormalities were associated with clinical outcomes of the infection and, thus, can potentially be utilized as biomarkers in clinical COVID-19 management. However, a number of parameters need to be assessed, before a clinical use of a biomarker candidate is proposed, with the most important one being reproducible detection in independent target populations. METHODS We benchmarked data reported in publications investigating genome-wide methylation changes occurring in blood cells during SARS-CoV-2 virus infection to assess potential applicability of these changes as biomarkers in clinical management of COVID-19 patients. RESULTS We identified nine studies, in which infection-related genome-wide methylation changes in blood cells of COVID-19 patients were reported and associations of those changes with outcomes of the infection was assessed. Our benchmarking analysis showed that each of those studies included patients with different clinical characteristics and each study utilized different symptoms assessment criteria. Most importantly, no uniform data analysis methodologies to identify virus-related methylation changes were used in the analyzed studies. Consequently, analyzed studies, except for one, which was based on uniform data analysis workflow applied across independent patient cohorts, reported sufficiently uniform results that would allow to propose the use of the identified infection-related methylation changes as biomarkers for clinical management of COVID-19 patients. CONCLUSIONS Due to lack of coherence between studies reporting SARS-CoV-2 associated methylation changes in blood, the potential use of already reported methylation changes as biomarkers for clinical management of COVID-19 patients needs to be further assessed in rigorously controlled studies. At the same time unified data analysis framework appears to be the most critical in development of biomarkers associated with the severity of SARS-CoV-2 infection.
Collapse
Affiliation(s)
- Patrycja K Przybylowicz
- Independent Clinical Epigenetics Laboratory, Pomeranian Medical University in Szczecin, Unii Lubelskiej 1, Szczecin, 71-252, Poland
| | - Tomasz Klepinowski
- Department of Neurosurgery, Pomeranian Medical University in Szczecin, Unii Lubelskiej 1, Szczecin, 71-252, Poland
| | - Olga Taryma-Leśniak
- Independent Clinical Epigenetics Laboratory, Pomeranian Medical University in Szczecin, Unii Lubelskiej 1, Szczecin, 71-252, Poland
| | - Jan Bińkowski
- Independent Clinical Epigenetics Laboratory, Pomeranian Medical University in Szczecin, Unii Lubelskiej 1, Szczecin, 71-252, Poland
| | - Tomasz K Wojdacz
- Independent Clinical Epigenetics Laboratory, Pomeranian Medical University in Szczecin, Unii Lubelskiej 1, Szczecin, 71-252, Poland.
| |
Collapse
|
6
|
Bozack A, Khodasevich D, Nwanaji-Enwerem JC, Gladish N, Shen H, Daredia S, Gamble M, Needham BL, Rehkopf DH, Cardenas A. One-carbon metabolism-related compounds are associated with epigenetic aging biomarkers: results from the cross-sectional National Health and Nutrition Examination Survey 1999-2002. Am J Clin Nutr 2025:S0002-9165(25)00317-X. [PMID: 40456316 DOI: 10.1016/j.ajcnut.2025.05.029] [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: 01/06/2025] [Revised: 05/05/2025] [Accepted: 05/27/2025] [Indexed: 06/11/2025] Open
Abstract
BACKGROUND One-carbon metabolism (OCM), a biochemical pathway dependent on micronutrients including B vitamins, plays an essential role in aging-related physiological processes. DNA methylation-based aging biomarkers may be influenced by OCM. OBJECTIVES This study investigated associations of OCM-related biomarkers with epigenetic aging biomarkers in the cross-sectional National Health and Nutrition Examination Survey (1999-2002). METHODS Blood DNA methylation was measured in adults aged ≥50 y. The following epigenetic aging biomarkers were included: Horvath1, Horvath2, Hannum, PhenoAge, GrimAge2, Dunedin Pace-of-Aging (DunedinPoAm), and DNA methylation telomere length (DNAmTL). We tested for associations of serum folate, red blood cell folate, vitamin B12, homocysteine (Hcy), and methylmalonic acid (MMA) concentrations with epigenetic age deviation (EAD) among 2346 participants with epigenetic and nutritional status biomarkers using adjusted survey-weighted general linear regression models. RESULTS A doubling of serum folate concentration was associated with -0.82 y (95% confidence interval: -1.40, -0.23) lower GrimAge EAD, -0.13 SDs (-0.22, -0.03) lower DunedinPoAm, and 0.02 kb (0.00, 0.04) greater DNAmTL EAD. Conversely, a doubling in Hcy concentration was associated with 1.05 y (0.06, 2.04) greater PhenoAge EAD, 1.93 y (1.16, 2.71) greater GrimAge2 EAD, and 0.26 SDs (0.10, 0.41) greater DunedinPoAm. Associations of serum folate with EAD were attenuated after adjusting for smoking status, alcohol intake, and estimated glomerular filtration rate. Furthermore, smoking modified the associations of Hcy with GrimAge2 EAD. Chronic kidney disease modified associations of B12 and MMA with Horvath1 and GrimAge2 EAD, respectively. CONCLUSIONS In a nationally representative sample of United States adults, higher concentration of folate, a carbon donor, was associated with lower EAD, and higher concentration of Hcys, an indicator of OCM deficiencies, was associated with greater EAD; however, some associations were influenced by smoking and renal function. Future research should focus on high-risk populations. Long-term randomized controlled trials are also needed to establish causality and investigate the clinical relevance of changes in EAD.
Collapse
Affiliation(s)
- Anne Bozack
- Department of Epidemiology and Population Health, Stanford University, Palo Alto, CA, United States.
| | - Dennis Khodasevich
- Department of Epidemiology and Population Health, Stanford University, Palo Alto, CA, United States
| | - Jamaji C Nwanaji-Enwerem
- Department of Emergency Medicine, Center for Health Justice, and Center of Excellence in Environmental Toxicology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Nicole Gladish
- Department of Epidemiology and Population Health, Stanford University, Palo Alto, CA, United States
| | - Hanyang Shen
- Department of Epidemiology and Population Health, Stanford University, Palo Alto, CA, United States
| | - Saher Daredia
- Department of Epidemiology, School of Public Health, University of California, Berkeley, Berkeley, CA, United States
| | - Mary Gamble
- Department of Environmental Health Sciences, Mailman School of Public Health, Columbia University, New York, NY, United States
| | - Belinda L Needham
- Department of Epidemiology, University of Michigan, Ann Arbor, MI, United States
| | - David H Rehkopf
- Department of Epidemiology and Population Health, Stanford University, Palo Alto, CA, United States; Department of Health Policy, Stanford University, Palo Alto, CA, United States; Department of Medicine - Primary Care and Population Health, Stanford University, Palo Alto, CA, United States; Department of Pediatrics, Stanford University, Palo Alto, CA, United States; Department of Sociology, Stanford University, Palo Alto, CA, United States
| | - Andres Cardenas
- Department of Epidemiology and Population Health, Stanford University, Palo Alto, CA, United States; Department of Pediatrics, Stanford University, Palo Alto, CA, United States
| |
Collapse
|
7
|
Sokolowska KE, Antoniewski J, Sobalska-Kwapis M, Strapagiel D, Marciniak W, Lubiński J, Wojdacz TK. Synergic effect of arsenic exposure related methylation changes in three cohorts exposed to levels of this toxicant. Int Arch Occup Environ Health 2025:10.1007/s00420-025-02147-6. [PMID: 40418341 DOI: 10.1007/s00420-025-02147-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2025] [Accepted: 04/24/2025] [Indexed: 05/27/2025]
Abstract
PURPOSE The results of studies assessing impact of arsenic exposure on methylome are to large extent inconsistent. To contribute to understanding of effect of arsenic exposure on methylome of the exposed cells, we assess the impact of low-level arsenic exposure on methylome of blood cells in three cohorts of exposed individuals. METHODS The Infinium MethylationEPIC array (Illumina Inc.) was used for genome-wide methylation profiling and robust linear regression to identify arsenic-related methylation changes in blood cells from healthy individuals with a 12-year cancer-free follow-up and breast cancer patients, sampled on average 4.29 years before diagnosis, as well as methylomics data from cord blood samples of Biomarkers of Exposure to Arsenic cohort. RESULTS Our analysis identified a 2,453 arsenic-associated methylation changes in blood from healthy individuals, 9,662 in breast cancer patients and 6,745 in cord blood samples. Similarly to previous studies methylation changes that we identified in each cohort, overlapped only to some extent. However, molecular processes linked to identified methylation changes were very similar in each of the cohorts. And included pathways that could be clearly associated with the adverse effects of arsenic exposure and specifically cancer in the cohort of cancer patients. Moreover, the genomic regions harboring identified in each cohort methylation changes were similar and predominantly included regions participating in regulation of gene transcription. CONCLUSION Overall, our findings show that specificity of arsenic related methylation changes is low but the impact of these changes on cell physiology is very similar across three cohorts we studded.
Collapse
Affiliation(s)
- Katarzyna Ewa Sokolowska
- Independent Clinical Epigenetics Laboratory, Pomeranian Medical University, Unii Lubelskiej 1, Szczecin, 71-252, Poland
| | - Jacek Antoniewski
- Independent Clinical Epigenetics Laboratory, Pomeranian Medical University, Unii Lubelskiej 1, Szczecin, 71-252, Poland
| | - Marta Sobalska-Kwapis
- Biobank Laboratory, Departament of Oncobiology and Epigenetics, Faculty of Biology and Environmental Protection, University of Lodz, Pomorska 139 St, Lodz, 90-235, Poland
| | - Dominik Strapagiel
- Biobank Laboratory, Departament of Oncobiology and Epigenetics, Faculty of Biology and Environmental Protection, University of Lodz, Pomorska 139 St, Lodz, 90-235, Poland
| | - Wojciech Marciniak
- Department of Genetics and Pathology, Pomeranian Medical University, Szczecin, Poland
- Read-Gene SA, Grzepnica, Poland
| | - Jan Lubiński
- Department of Genetics and Pathology, Pomeranian Medical University, Szczecin, Poland
- Read-Gene SA, Grzepnica, Poland
| | - Tomasz Kazimierz Wojdacz
- Independent Clinical Epigenetics Laboratory, Pomeranian Medical University, Unii Lubelskiej 1, Szczecin, 71-252, Poland.
| |
Collapse
|
8
|
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
|
9
|
Lv Y, Chen X, Jia Z, Wang Y, Zhu Z, Li C, Xu S, Li Y. Prenatal exposure to arsenic, umbilical cord blood DNA methylation, and child neurodevelopment: A prospective birth cohort study. ENVIRONMENTAL RESEARCH 2025; 280:121914. [PMID: 40398698 DOI: 10.1016/j.envres.2025.121914] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/30/2024] [Revised: 05/17/2025] [Accepted: 05/19/2025] [Indexed: 05/23/2025]
Abstract
Prenatal Exposure to Arsenic (As) is associated with child neurodevelopment disorders. However, the specific mechanisms involved remain unclear. This work intends to investigate the associations between prenatal As exposure and epigenome-wide Deoxyribo Nucleic Acid methylation (DNAm) and evaluate the role of DNAm in moderating the association between prenatal As exposure and child neurodevelopment. An As-related epigenome-wide DNAm association analysis was performed using robust linear models, and mediation analysis was further applied to explore potential DNAm mediators. Robust linear models were applied to perform an epigenome-wide association study (EWAS) for DNAm related to As exposure. Mediation analysis was subsequently conducted to explore potential DNAm mediators. The mental development index (MDI) score was found to be inversely associated with urinary As levels during the third trimester [β = -3.52, 95 % CI: -6.34, -0.71]. A total of 48 differential DNAm locations and 4 differentially methylated regions were found to be associated with urinary As concentration. Three cytidylyl phosphate guanosine positions (annotated to ARMC5, KIAA1217, and intergenic region, mediated proportion is around 30 %) mediated the association between urinary As and a reduction of MDI score (P < 0.05). Our findings indicated adverse effects of prenatal As exposure on child neurodevelopment, and specific DNAm played the role of partial mediator.
Collapse
Affiliation(s)
- Yiqing Lv
- Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection, State Key Laboratory of Environmental Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, PR China
| | - Xiaomei Chen
- Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection, State Key Laboratory of Environmental Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, PR China
| | - Zhenxian Jia
- Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection, State Key Laboratory of Environmental Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, PR China
| | - Yin Wang
- Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection, State Key Laboratory of Environmental Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, PR China
| | - Zhiqiang Zhu
- School of Environmental Science and Engineering, Hainan University, PR China
| | - Chengxi Li
- Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection, State Key Laboratory of Environmental Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, PR China
| | - Shunqing Xu
- School of Environmental Science and Engineering, Hainan University, PR China
| | - Yuanyuan Li
- Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection, State Key Laboratory of Environmental Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, PR China.
| |
Collapse
|
10
|
Lien JY, Hii LA, Su PH, Chen LY, Wen KC, Lai HC, Wang YC. Exploring potential methylation markers for ovarian cancer from cervical scraping samples. Hum Genomics 2025; 19:56. [PMID: 40382585 PMCID: PMC12085859 DOI: 10.1186/s40246-025-00763-4] [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: 02/07/2025] [Accepted: 04/25/2025] [Indexed: 05/20/2025] Open
Abstract
BACKGROUND Ovarian cancer has the highest mortality rate among gynecological cancers, making early detection crucial, as the five-year survival rate drops from 92% with early-stage diagnosis compared to 31% with late-stage diagnosis. Current diagnostic methods such as histopathological examination and detection of cancer antigen 125 and human epididymis protein 4 biomarkers are either invasive or lack specificity and sensitivity. However, the Papanicolaou (Pap) test, which is widely used for cervical cancer screening, shows the potential for detecting ovarian cancer by identifying tumor DNA in cervical scrapings. Since aberrant DNA methylation patterns are linked to cancer progression, DNA methylation offers a promising avenue for early diagnosis. Therefore, this study aimed to develop a methylation-based machine-learning model to stratify patients with ovarian cancer from the cervical scraping samples collected via Pap test. RESULTS Cervical scrapings were collected by gynecologists using conventional Pap smears. In total, 160 samples were collected: 95 normal, 37 benign, and 28 malignant. Methylation data were generated using the Illumina Infinium MethylationEPIC BeadChip array, which contains approximately 850,000 CpG loci. Methylation data were initially divided into training and testing sets in a 3:1 ratio comprising 120 and 40 samples, respectively. A two-step methylation-based model was trained using the training data for classification: a principal component analysis (PCA) model, consisting of 30 features, to classify samples as normal or tumor; then a gradient boosting model, containing 16 features, to further stratify tumor samples as benign or malignant. The two-step model achieved an accuracy of 0.88 and an F1-score of 0.86 on the testing data. Furthermore, an over-representation analysis was conducted to explore the functions associated with genes mapped from differentially methylated positions (DMPs) in comparisons between normal and tumor samples, as well as between benign and malignant samples. These results suggest that DMPs may be associated with olfactory transduction when comparing normal versus tumor samples, and immune regulation when comparing benign and malignant samples. CONCLUSIONS Our two-step model shows promise for predicting ovarian cancer and suggests that cervical scrapings may be a viable alternative for sample collection during screening.
Collapse
Affiliation(s)
- Ju-Yin Lien
- Institute of Biomedical Informatics, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Lu Ann Hii
- Institute of Biomedical Informatics, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Po-Hsuan Su
- College of Health Technology, National Taipei University of Nursing and Health Sciences, Taipei, Taiwan
| | - Lin-Yu Chen
- Department of Obstetrics and Gynecology, Shuang Ho Hospital, Taipei Medical University, New Taipei City, Taiwan
| | - Kuo-Chang Wen
- Department of Obstetrics and Gynecology, Shuang Ho Hospital, Taipei Medical University, New Taipei City, Taiwan
- Department of Obstetrics and Gynecology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Hung-Cheng Lai
- Department of Obstetrics and Gynecology, Shuang Ho Hospital, Taipei Medical University, New Taipei City, Taiwan.
- Department of Obstetrics and Gynecology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan.
- Translational Epigenetics Center, Shuang Ho Hospital, Taipei Medical University, New Taipei City, Taiwan.
- Department of Obstetrics and Gynecology, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan.
| | - Yu-Chao Wang
- Institute of Biomedical Informatics, National Yang Ming Chiao Tung University, Taipei, Taiwan.
- Digital Medicine and Smart Healthcare Research Center, National Yang Ming Chiao Tung University, Taipei, Taiwan.
| |
Collapse
|
11
|
Patrizi S, Vallese S, Barresi S, Cassandri M, Giovannoni I, Pedace L, Abballe L, Vinciarelli F, Antonacci C, Stracuzzi A, Mancini B, Russo I, Di Giannatale A, Rota R, Alaggio R, Locatelli F, Milano GM, Miele E. Age-linked DNA methylation and gene expression patterns in parameningeal head and neck alveolar rhabdomyosarcoma reveal CDK9 as a promising therapeutic target. Pharmacol Res 2025; 216:107767. [PMID: 40350107 DOI: 10.1016/j.phrs.2025.107767] [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: 04/03/2025] [Revised: 05/07/2025] [Accepted: 05/07/2025] [Indexed: 05/14/2025]
Abstract
BACKGROUND Alveolar rhabdomyosarcoma (ARMS) primarily affects children in the first decade of life, but it can also occur during adolescence, typically with a more favorable prognosis. This study aimed to explore differences in DNA methylation (DNAm) and gene expression profiles that may account for the worse prognosis in younger patients; and to investigate possible new therapeutic targets. METHODS We conducted whole-genome DNAm and transcriptome analyses on 10 parameningeal head and neck ARMS patients, including 4 patients under 1 year old and 6 over 10 years old. Among the differentially expressed genes, we focused on actionable therapeutic targets and confirmed their protein expression levels by immunohistochemistry. We validated the biological relevance of molecules of interest through functional experiments on rhabdomyosarcoma cell lines. RESULTS DNAm profiles did not significantly differ across age groups, while gene expression was the primary driver of observed differences. Several enriched pathways characterized younger patients with respect to older ones, including FAS, Integrin, PI3 kinase, and p53 by glucose deprivation. Among actionable molecules, cyclin dependent kinase 9 (CDK9) emerged as a promising therapy target, highly expressed in younger patients. Of note, CDK9 inhibitors specifically inhibit cell growth in bi- and three-dimensional ARMS cellular models, both as a monotherapy and in combination with BRD4 inhibitors. CONCLUSION Despite the small sample size, these findings suggest potential age-related molecular mechanisms and highlight candidate genes for further investigation as novel therapeutic targets. Notably, we identified CDK9 as a promising target, warranting further exploration in the context of ARMS treatment.
Collapse
Affiliation(s)
- Sara Patrizi
- Onco-Haematology, Cell and Gene Therapy, Bambino Gesù Children's Hospital, IRCCS, Rome 00165, Italy.
| | - Silvia Vallese
- Pathology Unit, Bambino Gesù Children's Hospital, IRCCS, Rome 00165, Italy
| | - Sabina Barresi
- Pathology Unit, Bambino Gesù Children's Hospital, IRCCS, Rome 00165, Italy
| | - Matteo Cassandri
- Onco-Haematology, Cell and Gene Therapy, Bambino Gesù Children's Hospital, IRCCS, Rome 00165, Italy
| | | | - Lucia Pedace
- Onco-Haematology, Cell and Gene Therapy, Bambino Gesù Children's Hospital, IRCCS, Rome 00165, Italy
| | - Luana Abballe
- Onco-Haematology, Cell and Gene Therapy, Bambino Gesù Children's Hospital, IRCCS, Rome 00165, Italy
| | - Flavia Vinciarelli
- Onco-Haematology, Cell and Gene Therapy, Bambino Gesù Children's Hospital, IRCCS, Rome 00165, Italy
| | - Celeste Antonacci
- Onco-Haematology, Cell and Gene Therapy, Bambino Gesù Children's Hospital, IRCCS, Rome 00165, Italy
| | | | - Barbara Mancini
- Pathology Unit, Bambino Gesù Children's Hospital, IRCCS, Rome 00165, Italy
| | - Ida Russo
- Onco-Haematology, Cell and Gene Therapy, Bambino Gesù Children's Hospital, IRCCS, Rome 00165, Italy
| | - Angela Di Giannatale
- Onco-Haematology, Cell and Gene Therapy, Bambino Gesù Children's Hospital, IRCCS, Rome 00165, Italy
| | - Rossella Rota
- Onco-Haematology, Cell and Gene Therapy, Bambino Gesù Children's Hospital, IRCCS, Rome 00165, Italy
| | - Rita Alaggio
- Pathology Unit, Bambino Gesù Children's Hospital, IRCCS, Rome 00165, Italy; Department of Medical-Surgical Sciences and Biotechnologies, Sapienza University of Rome, Rome 00185, Italy
| | - Franco Locatelli
- Onco-Haematology, Cell and Gene Therapy, Bambino Gesù Children's Hospital, IRCCS, Rome 00165, Italy; Department of Life Sciences and Public Health, Catholic University of the Sacred Heart, Rome 00168, Italy
| | - Giuseppe Maria Milano
- Onco-Haematology, Cell and Gene Therapy, Bambino Gesù Children's Hospital, IRCCS, Rome 00165, Italy
| | - Evelina Miele
- Onco-Haematology, Cell and Gene Therapy, Bambino Gesù Children's Hospital, IRCCS, Rome 00165, Italy.
| |
Collapse
|
12
|
Kuznetsov DV, Liu Y, Schowe AM, Czamara D, Instinske J, Pahnke CKL, Nöthen MM, Spinath FM, Binder EB, Diewald M, Forstner AJ, Kandler C, Mönkediek B. Genetic and environmental contributions to epigenetic aging across adolescence and young adulthood. Clin Epigenetics 2025; 17:78. [PMID: 40336042 PMCID: PMC12060359 DOI: 10.1186/s13148-025-01880-6] [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: 06/13/2024] [Accepted: 04/09/2025] [Indexed: 05/09/2025] Open
Abstract
BACKGROUND Epigenetic aging estimators commonly track chronological and biological aging, quantifying its accumulation (i.e., epigenetic age acceleration) or speed (i.e., epigenetic aging pace). Their scores reflect a combination of inherent biological programming and the impact of environmental factors, which are suggested to vary at different life stages. The transition from adolescence to adulthood is an important period in this regard, marked by an increasing and, then, stabilizing epigenetic aging variance. Whether this pattern arises from environmental influences or genetic factors is still uncertain. This study delves into understanding the genetic and environmental contributions to variance in epigenetic aging across these developmental stages. Using twin modeling, we analyzed four estimators of epigenetic aging, namely Horvath Acceleration, PedBE Acceleration, GrimAge Acceleration, and DunedinPACE, based on saliva samples collected at two timepoints approximately 2.5 years apart from 976 twins of four birth cohorts (aged about 9.5, 15.5, 21.5, and 27.5 years at first and 12, 18, 24, and 30 years at second measurement occasion). RESULTS Half to two-thirds (50-68%) of the differences in epigenetic aging were due to unique environmental factors, indicating the role of life experiences and epigenetic drift, besides measurement error. The remaining variance was explained by genetic (Horvath Acceleration: 24%; GrimAge Acceleration: 32%; DunedinPACE: 47%) and shared environmental factors (Horvath Acceleration: 26%; PedBE Acceleration: 47%). The genetic and shared environmental factors represented the primary sources of stable differences in corresponding epigenetic aging estimators over 2.5 years. Age moderation analyses revealed that the variance due to individually unique environmental sources was smaller in younger than in older cohorts in epigenetic aging estimators trained on chronological age (Horvath Acceleration: 47-49%; PedBE Acceleration: 33-68%). The variance due to genetic contributions, in turn, potentially increased across age groups for epigenetic aging estimators trained in adult samples (Horvath Acceleration: 18-39%; GrimAge Acceleration: 24-43%; DunedinPACE: 42-57%). CONCLUSIONS Transition to adulthood is a period of the increasing variance in epigenetic aging. Both environmental and genetic factors contribute to this trend. The degree of environmental and genetic contributions can be partially explained by the design of epigenetic aging estimators.
Collapse
Affiliation(s)
- Dmitry V Kuznetsov
- Bielefeld University, Bielefeld, Germany.
- Center for Environmental Neuroscience, Max Planck Institute for Human Development, Berlin, Germany.
| | - Yixuan Liu
- Bielefeld University, Bielefeld, Germany
| | - Alicia M Schowe
- Department Genes and Environment, Max Planck Institute of Psychiatry, Munich, Germany
- Graduate School of Systemic Neuroscience, Ludwig Maximilian University, Munich, Germany
| | - Darina Czamara
- Department Genes and Environment, Max Planck Institute of Psychiatry, Munich, Germany
| | | | - Charlotte K L Pahnke
- Institute of Human Genetics, School of Medicine and University Hospital Bonn, University of Bonn, Bonn, Germany
| | - Markus M Nöthen
- Institute of Human Genetics, School of Medicine and University Hospital Bonn, University of Bonn, Bonn, Germany
| | - Frank M Spinath
- Department of Psychology, Saarland University, Saarbrücken, Germany
| | - Elisabeth B Binder
- Department Genes and Environment, Max Planck Institute of Psychiatry, Munich, Germany
| | | | - Andreas J Forstner
- Institute of Human Genetics, School of Medicine and University Hospital Bonn, University of Bonn, Bonn, Germany
- Institute of Neuroscience and Medicine (INM-1), Research Center Jülich, Jülich, Germany
| | - Christian Kandler
- Bielefeld University, Bielefeld, Germany
- University of Bremen, Bremen, Germany
| | | |
Collapse
|
13
|
Qian Y, Peng Q, Qian Q, Gao X, Liu X, Li Y, Fan X, Cheng Y, Yuan N, Hadi S, Jin L, Wang S, Liu F. A methylation panel of 10 CpGs for accurate age inference via stepwise conditional epigenome-wide association study. Int J Legal Med 2025; 139:1193-1203. [PMID: 39633164 DOI: 10.1007/s00414-024-03365-2] [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: 05/29/2024] [Accepted: 10/31/2024] [Indexed: 12/07/2024]
Abstract
Estimating individual age from DNA methylation at age associated CpG sites may provide key information facilitating forensic investigations. Systematic marker screening and feature selection play a critical role in ensuring the performance of the final prediction model. In the discovery stage, we screened for 811876 CpGs from whole blood of 2664 Chinese individuals ranging from 18 to 83 years of age based on a stepwise conditional epigenome-wide association study (SCEWAS). The SCEWAS identified 28 CpGs showing genome-wide significant and independent effects. Further restricting this panel to 10 most informative CpGs showed a tolerable loss of information. A linear model consisting of these 10 CpGs could explain 93% of the age variance (R2 = 0.93) in the training set (n = 2664). In an independent test set of Chinese individuals (n = 648), this model also provided highly accurate predictions (R2 = 0.85, mean absolute deviation, MAD = 3.20 years). The model was additionally validated in a public dataset of multiple ancestral origins (86 Europeans, 14 Asians, and 273 Africans) and the prediction accuracy reduced significantly (R2 = 0.85, MAD = 6.21 years), as might be expected due to different genomic backgrounds, sample sizes, and age ranges. Our 10 CpG model also outperformed the recently proposed 9-CpG model constructed in 390 Chinese males (R2 = 0.79 in test set). We also demonstrated that our SCEWAS approach outperformed the traditional EWAS and the elastic net approach in obtaining a small set of most age informative CpGs. Overall, our systematic genome-wide feature selection identified a small panel of 10 CpGs for accurate age estimation with high potential in forensic applications.
Collapse
Affiliation(s)
- Yu Qian
- Key Laboratory of Genomic and Precision Medicine, Beijing Institute of Genomics, China National Center for Bioinformation, Chinese Academy of Sciences, Beijing, China
- Beijing No.8 High School, Beijing, China
| | - Qianqian Peng
- CAS Key Laboratory of Computational Biology, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, 320 Yue Yang Road, Shanghai, 200031, China
| | - Qili Qian
- CAS Key Laboratory of Computational Biology, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, 320 Yue Yang Road, Shanghai, 200031, China
| | - Xingjian Gao
- National Clinical Research Center of Kidney Diseases, Jinling Hospital, Nanjing, Jiangsu, China
| | - Xinxuan Liu
- Key Laboratory of Genomic and Precision Medicine, Beijing Institute of Genomics, China National Center for Bioinformation, Chinese Academy of Sciences, Beijing, China
| | - Yi Li
- CAS Key Laboratory of Computational Biology, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, 320 Yue Yang Road, Shanghai, 200031, China
| | - Xiu Fan
- Key Laboratory of Genomic and Precision Medicine, Beijing Institute of Genomics, China National Center for Bioinformation, Chinese Academy of Sciences, Beijing, China
| | - Yuan Cheng
- Key Laboratory of Genomic and Precision Medicine, Beijing Institute of Genomics, China National Center for Bioinformation, Chinese Academy of Sciences, Beijing, China
| | - Na Yuan
- Key Laboratory of Genomic and Precision Medicine, Beijing Institute of Genomics, China National Center for Bioinformation, Chinese Academy of Sciences, Beijing, China
| | - Sibte Hadi
- Department of Forensic Sciences, College of Criminal Justice, Naif Arab University of Security Sciences, Riyadh, 11452, Kingdom of Saudi Arabia
| | - Li Jin
- Human Phenome Institute, Fudan University, Shanghai, China
- Taizhou Institute of Health Sciences, Fudan University, Taizhou, Jiangsu, China
- State Key Laboratory of Genetic Engineering and Ministry of Education Key Laboratory of Contemporary Anthropology, Collaborative Innovation Center for Genetics and Development, School of Life Sciences, Fudan University, Shanghai, China
| | - Sijia Wang
- CAS Key Laboratory of Computational Biology, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, 320 Yue Yang Road, Shanghai, 200031, China
- Center for Excellence in Animal Evolution and Genetics, Chinese Academy of Sciences, Kunming, 650223, China
| | - Fan Liu
- Department of Forensic Sciences, College of Criminal Justice, Naif Arab University of Security Sciences, Riyadh, 11452, Kingdom of Saudi Arabia.
| |
Collapse
|
14
|
deSteiguer AJ, Raffington L, Sabhlok A, Tanksley P, Tucker-Drob EM, Harden KP. Stability of Aging- and Cognition-Related Methylation Profile Scores Across Two Waves in Children and Adolescents. Child Dev 2025; 96:1189-1206. [PMID: 40171752 DOI: 10.1111/cdev.14239] [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: 01/24/2024] [Revised: 01/13/2025] [Accepted: 02/12/2025] [Indexed: 04/04/2025]
Abstract
DNA-methylation profile scores (MPSs) index biology relevant for lifelong physical and cognitive health, but information on their longitudinal stability in childhood is lacking. Using two waves of data collected from 2014 to 2022 (Mlag between waves = 2.41 years) from N = 407 participants (Mage = 12.05 years, 51% female, 60% White), test-retest correlations were estimated for four salivary MPSs related to aging (PhenoAgeAccel, GrimAgeAccel, DunedinPACE), and cognitive function (Epigenetic-g). MPSs varied in longitudinal stability (test-retest rs = 0.38 to 0.76). MPSs did not differ in children exposed to the COVID-19 pandemic, but race-ethnic and sex differences were apparent. Further research is necessary to understand which environmental perturbations impact DNA-methylation trajectories and when children are most sensitive to those impacts.
Collapse
Affiliation(s)
- Abby J deSteiguer
- Department of Psychology, University of Texas at Austin, Austin, Texas, USA
| | - Laurel Raffington
- Max Planck Research Group Biosocial-Biology, Social Disparities, and Development, Max Planck Institute for Human Development, Berlin, Germany
| | - Aditi Sabhlok
- Department of Psychology, University of Texas at Austin, Austin, Texas, USA
| | - Peter Tanksley
- Population Research Center, The University of Texas at Austin, Austin, Texas, USA
| | - Elliot M Tucker-Drob
- Department of Psychology, University of Texas at Austin, Austin, Texas, USA
- Population Research Center, The University of Texas at Austin, Austin, Texas, USA
| | - K Paige Harden
- Department of Psychology, University of Texas at Austin, Austin, Texas, USA
- Population Research Center, The University of Texas at Austin, Austin, Texas, USA
| |
Collapse
|
15
|
Hill RA, Gibbons A, Suwakulsiri W, Taseska A, Darke H, Malhotra A, Yee H, Fahey M, Hunt RW, Lim I, Palmer K, Sundram S. Investigating the impact of severe maternal SARS-CoV-2 infection on infant DNA methylation and neurodevelopment. Mol Psychiatry 2025; 30:1976-1984. [PMID: 39478169 DOI: 10.1038/s41380-024-02808-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Revised: 10/17/2024] [Accepted: 10/21/2024] [Indexed: 04/24/2025]
Abstract
Maternal infections during pregnancy can increase the risk to offspring of developing a neurodevelopmental disorder. Given the global prevalence and severity of infection with Severe Acute Respiratory Syndrome related Coronavirus 2 (SARS-CoV-2), the objective of this study was to determine if in utero exposure to severe maternal SARS-CoV-2 infection alters infant neurodevelopmental outcomes at 12 months and to identify potential biological markers of adverse infant outcomes. Mother-infant dyads exposed to severe SARS-CoV-2 infection (requiring hospitalization) during pregnancy and age and sociodemographic matched control dyads were recruited from Monash Medical Centre, Australia in 2021/22 and prospectively assessed over 12 months. Maternal serum cytokine levels and Edinburgh Postnatal Depression Scale (EPDS) scores were assessed at birth. DNA methylation was assessed from infant buccal swabs at birth (Illumina EPIC BeadChip). Infant neurodevelopmental outcomes at 12 months were assessed using the Ages and Stages Questionnaire (ASQ-3). Mothers exposed to severe SARS-CoV-2 exhibited elevated serum IL-6 and IL-17A and higher EPDS scores than controls at birth. Infants exposed to severe SARS-CoV-2 in utero demonstrated over 3000 significant differentially methylated sites within their genomes compared to non-exposed (adjusted p-value < 0.05), including genes highly relevant to ASD and synaptic pathways. At 12 months, severe SARS-CoV-2 exposed infants scored lower on the ASQ-3 than non-exposed infants, and communication and problem-solving scores negatively correlated with maternal IL-6 levels at birth. DNA methylation changes therefore unveil potential mechanisms linking infection exposure to delayed neurodevelopment and maternal serum IL-6 levels may be a potential biomarker of child developmental delay. Mothers exposed to severe SARS-CoV-2 infections show elevated pro-inflammatory cytokines. Infants exposed in utero to severe SARS-CoV-2 infection show altered DNA methylation at birth and delayed development at 12 months of age. Created in Biorender.com.
Collapse
Affiliation(s)
- Rachel A Hill
- Department of Psychiatry, Monash University, Clayton, Vic, Australia.
| | - Andrew Gibbons
- Department of Psychiatry, Monash University, Clayton, Vic, Australia
| | | | - Angela Taseska
- Department of Psychiatry, Monash University, Clayton, Vic, Australia
| | - Hayley Darke
- Department of Psychiatry, Monash University, Clayton, Vic, Australia
| | - Atul Malhotra
- Department of Paediatrics, Monash University, Clayton, Vic, Australia
- Monash Children's Hospital, Clayton, Vic, Australia
| | - Hnin Yee
- Department of Psychiatry, Monash University, Clayton, Vic, Australia
| | - Michael Fahey
- Department of Paediatrics, Monash University, Clayton, Vic, Australia
- Monash Children's Hospital, Clayton, Vic, Australia
| | - Rod W Hunt
- Department of Paediatrics, Monash University, Clayton, Vic, Australia
- Monash Children's Hospital, Clayton, Vic, Australia
- Clinical Sciences, Murdoch Children's Research Institute, Parkville, Vic, Australia
| | - Izaak Lim
- Department of Psychiatry, Monash University, Clayton, Vic, Australia
| | - Kirsten Palmer
- Monash Women's, Monash Health, Clayton, Vic, Australia
- Department of Obstetrics and Gynaecology, Monash University, Clayton, Vic, Australia
| | - Suresh Sundram
- Department of Psychiatry, Monash University, Clayton, Vic, Australia.
- Mental Health Program, Monash Health, Clayton, Vic, Australia.
| |
Collapse
|
16
|
Jyoti J, Gronau I, Cakir E, Hütt MT, Lerchl A, Meyer V. 5G-exposed human skin cells do not respond with altered gene expression and methylation profiles. PNAS NEXUS 2025; 4:pgaf127. [PMID: 40365161 PMCID: PMC12070386 DOI: 10.1093/pnasnexus/pgaf127] [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: 07/04/2024] [Accepted: 03/31/2025] [Indexed: 05/15/2025]
Abstract
Due to the ever-increasing wirelessly transmitted data, the development of new transmission standards and higher frequencies in the 5G band is required. Despite basic biophysical considerations that argue against health effects, there is public concern about this technology. Because the skin penetration depth at these frequencies is only 1 mm or less, we exposed fibroblasts and keratinocytes to electromagnetic fields up to ten times the permissible limits, for 2 and 48 h in a fully blinded experimental design. Sham-exposed cells served as negative, and UV-exposed cells as positive controls. Differences in gene expression and methylation due to exposure were small and not higher than expected by chance. These data strongly support the assessment that there is no evidence for exposure-induced damage to human skin cells.
Collapse
Affiliation(s)
- Jyoti Jyoti
- School of Science, Constructor University, Bremen 28759, Germany
| | - Isabel Gronau
- School of Science, Constructor University, Bremen 28759, Germany
| | - Eda Cakir
- School of Science, Constructor University, Bremen 28759, Germany
| | | | - Alexander Lerchl
- School of Science, Constructor University, Bremen 28759, Germany
| | - Vivian Meyer
- School of Science, Constructor University, Bremen 28759, Germany
- Department of Biology and Environmental Sciences, Carl von Ossietzky Universität Oldenburg, Oldenburg 26129, Germany
| |
Collapse
|
17
|
Mori MP, Lozoya OA, Brooks AM, Bortner CD, Nadalutti CA, Ryback B, Rickard BP, Overchuk M, Rizvi I, Rogasevskaia T, Huang KT, Hasan P, Hajnóczky G, Santos JH. Mitochondrial membrane hyperpolarization modulates nuclear DNA methylation and gene expression through phospholipid remodeling. Nat Commun 2025; 16:4029. [PMID: 40301431 PMCID: PMC12041266 DOI: 10.1038/s41467-025-59427-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2024] [Accepted: 04/23/2025] [Indexed: 05/01/2025] Open
Abstract
Maintenance of the mitochondrial inner membrane potential (ΔΨm) is critical for many aspects of mitochondrial function. While ΔΨm loss and its consequences are well studied, little is known about the effects of mitochondrial hyperpolarization. In this study, we used cells deleted of ATP5IF1 (IF1), a natural inhibitor of the hydrolytic activity of the ATP synthase, as a genetic model of increased resting ΔΨm. We found that the nuclear DNA hypermethylates when the ΔΨm is chronically high, regulating the transcription of mitochondrial, carbohydrate and lipid genes. These effects can be reversed by decreasing the ΔΨm and recapitulated in wild-type (WT) cells exposed to environmental chemicals that cause hyperpolarization. Surprisingly, phospholipid changes, but not redox or metabolic alterations, linked the ΔΨm to the epigenome. Sorted hyperpolarized WT and ovarian cancer cells naturally depleted of IF1 also showed phospholipid remodeling, indicating this as an adaptation to mitochondrial hyperpolarization. These data provide a new framework for how mitochondria can impact epigenetics and cellular biology to influence health outcomes, including through chemical exposures and in disease states.
Collapse
Affiliation(s)
- Mateus Prates Mori
- Mechanistic Toxicology Branch, Division of Translational Toxicology, National Institute of Environmental Health Sciences (NIEHS), National Institutes of Health (NIH), Durham, NC, USA
| | - Oswaldo A Lozoya
- Genome Integrity and Structural Biology Laboratory, National Institute of Environmental Health Sciences (NIEHS), National Institutes of Health (NIH), Durham, NC, USA
| | - Ashley M Brooks
- Biostatistics and Computational Biology Branch, Integrative Bioinformatics Support Group, National Institute of Environmental Health Sciences (NIEHS), National Institutes of Health (NIH), Durham, NC, USA
| | - Carl D Bortner
- Flow Cytometry Center, National Institute of Environmental Health Sciences (NIEHS), National Institutes of Health (NIH), Durham, NC, USA
| | - Cristina A Nadalutti
- Mechanistic Toxicology Branch, Division of Translational Toxicology, National Institute of Environmental Health Sciences (NIEHS), National Institutes of Health (NIH), Durham, NC, USA
| | - Birgitta Ryback
- Dana Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Brittany P Rickard
- Curriculum in Toxicology & Environmental Medicine, University of North Carolina (UNC), Chapel Hill, NC, USA
| | - Marta Overchuk
- Department of Biomedical Engineering, North Carolina State University, Raleigh, NC, USA
| | - Imran Rizvi
- Department of Biomedical Engineering, North Carolina State University, Raleigh, NC, USA
- Lineberger Comprehensive Cancer Center, UNC, Chapel Hill, NC, USA
| | | | - Kai Ting Huang
- MitoCare Center, Department of Pathology and Genomic Medicine, Thomas Jefferson University, Philadelphia, PA, USA
| | - Prottoy Hasan
- MitoCare Center, Department of Pathology and Genomic Medicine, Thomas Jefferson University, Philadelphia, PA, USA
| | - György Hajnóczky
- MitoCare Center, Department of Pathology and Genomic Medicine, Thomas Jefferson University, Philadelphia, PA, USA
| | - Janine H Santos
- Mechanistic Toxicology Branch, Division of Translational Toxicology, National Institute of Environmental Health Sciences (NIEHS), National Institutes of Health (NIH), Durham, NC, USA.
| |
Collapse
|
18
|
Dong Z, Zhao K, Gu H, Yang W, Zhang X. Profiling of Circulating Cell-free DNA Methylation Patterns Identifies Aberrant Methylated CTBP1 Promotor Sites for Prediction of Alzheimer's Disease. J Integr Neurosci 2025; 24:36527. [PMID: 40302267 DOI: 10.31083/jin36527] [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: 12/22/2024] [Revised: 02/14/2025] [Accepted: 02/25/2025] [Indexed: 05/02/2025] Open
Abstract
BACKGROUND Alzheimer's disease (AD) is the most common neurodegenerative disease affecting the elderly, with its diagnosis at early stages crucial for effective intervention. Recent evidence increasingly supports the role of epigenetic alterations in AD pathogenesis, highlighting the need for innovative biomarkers that reflect these changes. This study aimed to characterize the genome-wide DNA methylation profiles of cell-free DNA in peripheral blood for potential biomarkers associated with AD. METHODS The Illumina Infinium array was utilized to detect the methylation patterns of circulating cell-free DNA from AD patients and healthy controls. The R Bioconductor Linear Models for Microarray Data (LIMMA) package was employed to identify methylation variable positions (MVPs), and Probe Lasso was used to pinpoint differentially methylated regions (DMRs) linked to AD. Bioinformatics enrichment analysis of the annotated genes was performed using EnrichR. A second cohort was recruited to validate the methylation changes at the C-terminal binding protein1 (CTBP1) promoter cytosine-phosphate-guanine (CpG) sites via pyrosequencing. Additionally, microarray data from the Gene Expression Omnibus (GEO) database were analyzed to further validate gene expression and immune infiltration. RESULTS A unique DNA methylation landscape in peripheral blood was characterized for AD patients and 4335 MVPs showed significant differential methylation (p < 0.01). Functional annotation and pathway enrichment analysis underscored processes and pathways inherent in the nervous system. Probe Lasso identified 68 DMRs annotated to 10 genes, with hypermethylation of CpG islands in the CTBP1 TSS1500 promoter showing significant differences when AD and controls were compared (p < 0.01), with an area under the receiver operating characteristic (ROC) curve (AUC) of 0.779. Analysis of immune cell infiltration revealed CTBP1 expression is significantly correlated with altered distribution of immune cells (p < 0.001), underscoring its potential role in modulating immune responses in AD. Moreover, CTBP1 expression levels significantly varied across multiple GEO datasets. CONCLUSIONS AD displays distinct DNA methylation patterns in peripheral blood and CTBP1 promoter hypermethylation represents a promising potential biomarker for AD diagnosis.
Collapse
Affiliation(s)
- Zhiwu Dong
- Department of Laboratory Medicine, Shanghai Second People's Hospital, 200011 Shanghai, China
| | - Kewen Zhao
- Department of Pathophysiology, Shanghai Jiao Tong University School of Medicine, 200025 Shanghai, China
| | - Hongjun Gu
- Department of Geriatric Medicine, Shanghai Jinshan District Hospital of Integrated Traditional Chinese and Western Medicine, 201501 Shanghai, China
| | - Wenwei Yang
- Department of Laboratory Medicine, Shanghai Second People's Hospital, 200011 Shanghai, China
| | - Xin Zhang
- Department of Laboratory Medicine, Shanghai Second People's Hospital, 200011 Shanghai, China
| |
Collapse
|
19
|
Zarandooz S, Raffington L. Applying blood-derived epigenetic algorithms to saliva: cross-tissue similarity of DNA-methylation indices of aging, physiology, and cognition. Clin Epigenetics 2025; 17:61. [PMID: 40270051 PMCID: PMC12016411 DOI: 10.1186/s13148-025-01868-2] [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: 11/03/2024] [Accepted: 03/29/2025] [Indexed: 04/25/2025] Open
Abstract
BACKGROUND Epigenetic algorithms of aging, health, and cognition, based on DNA-methylation (DNAm) patterns, are prominent tools for measuring biological age and have been linked to age-related diseases, cognitive decline, and mortality. While most of these methylation profile scores (MPSs) are developed in blood tissue, there is growing interest in using less invasive tissues like saliva. The aim of the current study is to probe the cross-tissue intraclass correlation coefficients (ICCs) of MPSs developed in blood applied to saliva DNAm from the same people. While our primary focus is on MPSs that were previously found to be robustly correlated with social determinants of health, including second- and third-generation clocks and MPSs of physiology and cognition, we also report ICC values for first-generation clocks to enable comparison across metrics. We pooled three publicly available datasets that had both saliva and blood DNAm from the same individuals (total n = 107, aged 5-74 years), corrected MPSs for cell composition within each tissue, and computed the cross-tissue ICCs. RESULTS We found that after correcting for cell composition, saliva-blood cross-tissue ICCs were moderate for second- and third-generation indices of aging and MPSs of physiology and cognition. Specifically, PCGrimAge had the highest ICC (0.76), followed by PCPhenoAge (0.72), a measure of cognitive performance (Epigenetic-g, 0.69), DunedinPACE (0.68), PCGrimAge Acceleration (0.67), PCPhenoAge Acceleration (0.66), an MPS of hs-CRP (0.58), and BMI (0.54). These ICCs appear lower than previous reports on within-tissue ICCs (saliva ICCs range from 0.67 to 0.85, blood ICCs range from 0.73 to 0.93). Cross-tissue ICCs values for first-generation biological age acceleration measures were poor, ranging from 0.19 to 0.25. CONCLUSIONS Our findings suggest that applying second- and third-generation MPSs of biological age acceleration and related phenotypes developed in blood to saliva DNAm results in moderate cross-tissue similarity and the precise cross-tissue correspondence differs by measure. While the degree of cross-tissue similarity of several MPSs may suffice for some research settings, it may not be suitable in clinical or commercial applications. Collection of both blood and saliva DNAm samples is necessary to validate existing algorithms and to customize MPSs in saliva DNAm.
Collapse
Affiliation(s)
- Sepideh Zarandooz
- Max Planck Research Group Biosocial - Biology, Social Disparities, and Development, Max Planck Institute for Human Development, Berlin, Germany
| | - Laurel Raffington
- Max Planck Research Group Biosocial - Biology, Social Disparities, and Development, Max Planck Institute for Human Development, Berlin, Germany.
| |
Collapse
|
20
|
Tay JH, Chew YE, Wang W, Lim ZM, Guan L, Dorajoo R, Kennedy BK, Brooke R, Gordevicius J, Horvath S, Sandalova E, Maier AB. DNAm age differences between infinium methylationEPICv1 vs EPICv2 in buffy coat, PBMC, and saliva samples. Commun Biol 2025; 8:654. [PMID: 40269264 PMCID: PMC12019316 DOI: 10.1038/s42003-025-08021-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: 12/20/2024] [Accepted: 03/31/2025] [Indexed: 04/25/2025] Open
Abstract
This study aims to evaluate differences between Infinium MethylationEPIC (EPICv1) and Infinium MethylationEPICv2 (EPICv2) arrays in estimating DNAm age with eleven DNAm clocks using buffy coat, peripheral blood mononuclear cell (PBMC), and saliva from 16 healthy middle-aged individuals. DNAm ages were estimated using six principal component-based (PC) clocks (PCHorvath1, PCHorvath2, PCHannum, PCPhenoAge, PCGrimAge, and PCDNAmTL) and five non-PC clocks (DunedinPACE, DNAmFit, YingCausAge, YingAdaptAge, and YingDamAge) across all biological samples. Agreement between arrays was assessed using Spearman correlation, Bland-Altman plots, and Wilcoxon Signed-Rank test. The 16 individuals with median age of 48 [43.5;53.8] years, were predominantly female, Chinese and non-smokers. High correlations (ρ > 0.8) were observed between EPICv1 and EPICv2 except for DunedinPACE, YingDamAge and YingAdaptAge. PC-based clocks showed lower systematic bias (MAPE:0.118-8.98%) compared to non-PC-based clocks (MAPE:5.31-21.2%). Saliva samples demonstrated greatest variability between arrays. EPICv2 introduces systematic biases especially in non-PC-based clocks and between different biological samples.
Collapse
Affiliation(s)
- Jian Hua Tay
- Healthy Longevity Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117456, Singapore
- NUS Academy for Healthy Longevity, National University of Singapore, 10 Medical Drive, 117597, Singapore, Singapore
| | - Yi Ern Chew
- Healthy Longevity Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117456, Singapore
- NUS Academy for Healthy Longevity, National University of Singapore, 10 Medical Drive, 117597, Singapore, Singapore
| | - Weilan Wang
- Healthy Longevity Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117456, Singapore
- NUS Academy for Healthy Longevity, National University of Singapore, 10 Medical Drive, 117597, Singapore, Singapore
| | - Zhi Meng Lim
- Healthy Longevity Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117456, Singapore
- Centre for Healthy Longevity, National University Health System (NUHS), Singapore, 159964, Singapore
| | - Lihuan Guan
- Healthy Longevity Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117456, Singapore
- NUS Academy for Healthy Longevity, National University of Singapore, 10 Medical Drive, 117597, Singapore, Singapore
| | - Rajkumar Dorajoo
- Genome Institute of Singapore, Agency for Science, Technology and Research, Singapore, 138672, Singapore
- Department of Paediatrics, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 119228, Singapore
| | - Brian K Kennedy
- Healthy Longevity Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117456, Singapore
- Centre for Healthy Longevity, National University Health System (NUHS), Singapore, 159964, Singapore
| | - Robert Brooke
- Epigenetic Clock Development Foundation, Torrance, CA, USA
| | | | - Steve Horvath
- Epigenetic Clock Development Foundation, Torrance, CA, USA
- Altos Labs, San Diego Institute of Science, San Diego, CA, USA
| | - Elena Sandalova
- Healthy Longevity Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117456, Singapore
| | - Andrea B Maier
- Healthy Longevity Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117456, Singapore.
- Department of Human Movement Sciences, @AgeAmsterdam, Faculty of Behavioural and Movement Sciences, Vrije Universiteit Amsterdam, Amsterdam Movement Sciences, Amsterdam, Netherlands.
- NUS Academy for Healthy Longevity, National University of Singapore, 10 Medical Drive, 117597, Singapore, Singapore.
| |
Collapse
|
21
|
Pathak GA, Pietrzak RH, Lacobelle A, Overstreet C, Wendt FR, Deak JD, Friligkou E, Nunez YZ, Montalvo-Ortiz JL, Levey DF, Kranzler HR, Gelernter J, Polimanti R. Epigenetic and genetic profiling of comorbidity patterns among substance dependence diagnoses. Mol Psychiatry 2025:10.1038/s41380-025-03031-y. [PMID: 40247127 DOI: 10.1038/s41380-025-03031-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/16/2024] [Revised: 04/08/2025] [Accepted: 04/10/2025] [Indexed: 04/19/2025]
Abstract
This study investigated the genetic and epigenetic mechanisms underlying the comorbidity of five substance dependence diagnoses (SDs; alcohol, AD; cannabis, CaD; cocaine, CoD; opioid, OD; tobacco, TD). A latent class analysis (LCA) was performed on 22,668 individuals from six cohorts to identify comorbid DSM-IV SD patterns. In subsets of this sample, we tested SD-latent classes with respect to polygenic overlap of psychiatric and psychosocial traits in 7659 individuals of European descent and epigenome-wide changes in 886 individuals of African, European, and Admixed-American descents. The LCA identified four latent classes related to SD comorbidities: AD + TD, CoD + TD, AD + CoD + OD + TD (i.e., polysubstance addiction, PSU), and TD. In the epigenome-wide association analysis, SPATA4 cg02833127 was associated with CoD + TD, AD + TD, and PSU latent classes. AD + TD latent class was also associated with CpG sites located on ARID1B, NOTCH1, SERTAD4, and SIN3B, while additional epigenome-wide significant associations with CoD + TD latent class were observed in ANO6 and MOV10 genes. PSU-latent class was also associated with a differentially methylated region in LDB1. We also observed shared polygenic score (PGS) associations for PSU, AD + TD, and CoD + TD latent classes (i.e., attention-deficit hyperactivity disorder, anxiety, educational attainment, and schizophrenia PGS). In contrast, TD-latent class was exclusively associated with posttraumatic stress disorder-PGS. Other specific associations were observed for PSU-latent class (subjective wellbeing-PGS and neuroticism-PGS) and AD + TD-latent class (bipolar disorder-PGS). In conclusion, we identified shared and unique genetic and epigenetic mechanisms underlying SD comorbidity patterns. These findings highlight the importance of modeling the co-occurrence of SD diagnoses when investigating the molecular basis of addiction-related traits.
Collapse
Affiliation(s)
- Gita A Pathak
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA
- U.S Department of Veteran Affairs Connecticut Healthcare System, West Haven, CT, USA
| | - Robert H Pietrzak
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA
- U.S Department of Veteran Affairs Connecticut Healthcare System, West Haven, CT, USA
- Department of Social and Behavioral Sciences, Yale School of Public Health, New Haven, CT, USA
| | - AnnMarie Lacobelle
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA
- U.S Department of Veteran Affairs Connecticut Healthcare System, West Haven, CT, USA
| | - Cassie Overstreet
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA
- U.S Department of Veteran Affairs Connecticut Healthcare System, West Haven, CT, USA
| | - Frank R Wendt
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA
- U.S Department of Veteran Affairs Connecticut Healthcare System, West Haven, CT, USA
- Regeneron Genetics Center, Regeneron Pharmaceuticals, Tarrytown, NY, USA
| | - Joseph D Deak
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA
- U.S Department of Veteran Affairs Connecticut Healthcare System, West Haven, CT, USA
| | - Eleni Friligkou
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA
- U.S Department of Veteran Affairs Connecticut Healthcare System, West Haven, CT, USA
| | - Yaira Z Nunez
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA
- U.S Department of Veteran Affairs Connecticut Healthcare System, West Haven, CT, USA
| | - Janitza L Montalvo-Ortiz
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA
- U.S Department of Veteran Affairs Connecticut Healthcare System, West Haven, CT, USA
| | - Daniel F Levey
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA
- U.S Department of Veteran Affairs Connecticut Healthcare System, West Haven, CT, USA
| | - Henry R Kranzler
- Center for Studies of Addiction, University of Pennsylvania Perelman School of Medicine and the Mental Illness Research, Education and Clinical Center, Crescenz VAMC, Philadelphia, PA, USA
| | - Joel Gelernter
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA
- U.S Department of Veteran Affairs Connecticut Healthcare System, West Haven, CT, USA
| | - Renato Polimanti
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA.
- U.S Department of Veteran Affairs Connecticut Healthcare System, West Haven, CT, USA.
| |
Collapse
|
22
|
Tsai YH, Lai YH, Chen SJ, Cheng YC, Pai TW. DNA Methylation Biomarker Discovery for Colorectal Cancer Diagnosis Assistance Through Integrated Analysis. Cancer Inform 2025; 24:11769351251324545. [PMID: 40291817 PMCID: PMC12033546 DOI: 10.1177/11769351251324545] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2024] [Accepted: 02/13/2025] [Indexed: 04/30/2025] Open
Abstract
Objective This study aimed to identify biomarkers for colorectal cancer (CRC) with representative gene functions and high classification accuracy in tissue and blood samples. Methods We integrated CRC DNA methylation profiles from The Cancer Genome Atlas and comorbidity patterns of CRC to select biomarker candidates. We clustered these candidates near the promoter regions into multiple functional groups based on their functional annotations. To validate the selected biomarkers, we applied 3 machine learning techniques to construct models and compare their prediction performances. Results The 10 screened genes showed significant methylation differences in both tissue and blood samples. Our test results showed that 3-gene combinations achieved outstanding classification performance. Selecting 3 representative biomarkers from different genetic functional clusters, the combination of ADHFE1, ADAMTS5, and MIR129-2 exhibited the best performance across the 3 prediction models, achieving a Matthews correlation coefficient > .85 and an F1-score of .9. Conclusions Using integrated DNA methylation analysis, we identified 3 CRC-related biomarkers with remarkable classification performance. These biomarkers can be used to design a practical clinical toolkit for CRC diagnosis assistance and may also serve as candidate biomarkers for further clinical experiments through liquid biopsies.
Collapse
Affiliation(s)
- Yi-Hsuan Tsai
- Department of Computer Science and Information Engineering, National Taipei University of Technology, Taipei, Taiwan
| | - Yi-Husan Lai
- Department of Product Development, ACT Genomics Co., Ltd., Taipei, Taiwan
| | - Shu-Jen Chen
- Department of Product Development, ACT Genomics Co., Ltd., Taipei, Taiwan
| | - Yi-Chiao Cheng
- Division of Colon and Rectal Surgery, Department of Surgery, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Tun-Wen Pai
- Department of Computer Science and Information Engineering, National Taipei University of Technology, Taipei, Taiwan
| |
Collapse
|
23
|
Cordero AIH, Li X, Yang CX, Ambalavanan A, MacIsaac JL, Kobor MS, Doiron D, Tan W, Bourbeau J, Sin DD, Duan Q, Leung JM. Cannabis smoking is associated with persistent epigenome-wide disruptions despite smoking cessation. BMC Pulm Med 2025; 25:168. [PMID: 40205553 PMCID: PMC11980083 DOI: 10.1186/s12890-025-03634-9] [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: 02/05/2025] [Accepted: 03/28/2025] [Indexed: 04/11/2025] Open
Abstract
BACKGROUND The use of cannabis has been associated with both therapeutic and harmful effects. As with cigarette smoking, cannabis smoking may affect the epigenetic regulation (e.g., DNA methylation) of gene expression which could result in long term health effects. The study of DNA methylation in cannabis smoking has to date been restricted to young adults and there remains yet no evaluation of whether cannabis smoking cessation can reverse epigenetic disturbances. Here, we aimed to investigate the relationship between genome-wide DNA methylation and cannabis smoking. METHODS We used peripheral blood from a subset of older adults within the Canadian Cohort of Obstructive Lung Disease (CanCOLD) cohort (n = 93) to conduct an epigenome-wide DNA methylation analysis that identified differential methylated positions (DMPs) associated with cannabis smoking at a false discovery rate < 0.05. Using these DMPs, we then identified differentially methylated genes (DMGs) that enriched pathways associated with both former and current cannabis smoking status. RESULTS We found DMPs corresponding to 12,115 DMGs and 10,806 DMGs that distinguished the current and former cannabis smoking groups, respectively, from the never cannabis smoking group. 5,915 of these DMGs were shared between the current and former cannabis smoking groups. 50 enriched pathways were also shared between the current and former cannabis smoking groups, which were heavily represented by multiple aging- and cancer-related pathways. CONCLUSIONS Our findings indicate that in older adults, cannabis smoking is linked with epigenome-wide disruptions, many of which persist despite cannabis smoking cessation. Epigenetic modulation of genes associated with aging and cancer that remains even after quitting cannabis should serve as a caution that there may be long-lasting epigenetic injury with cannabis smoking. TRIAL REGISTRATION NCT00920348.
Collapse
Affiliation(s)
- Ana I Hernandez Cordero
- Centre for Heart Lung Innovation, St. Paul'S Hospital and University of British Columbia, Room 166, 1081 Burrard Street, Vancouver, BC, V6Z 1Y6, Canada
- Edwin S. H. Leong Healthy Aging Program, Faculty of Medicine, University of British Columbia, Vancouver, Canada
| | - Xuan Li
- Centre for Heart Lung Innovation, St. Paul'S Hospital and University of British Columbia, Room 166, 1081 Burrard Street, Vancouver, BC, V6Z 1Y6, Canada
| | - Chen Xi Yang
- Centre for Heart Lung Innovation, St. Paul'S Hospital and University of British Columbia, Room 166, 1081 Burrard Street, Vancouver, BC, V6Z 1Y6, Canada
| | - Amirtha Ambalavanan
- Department of Biomedical and Molecular Sciences, School of Medicine, Queen's University, Kingston, Canada
| | - Julie L MacIsaac
- Edwin S. H. Leong Healthy Aging Program, Faculty of Medicine, University of British Columbia, Vancouver, Canada
- Centre for Molecular Medicine and Therapeutics, University of British Columbia, Vancouver, Canada
| | - Michael S Kobor
- Edwin S. H. Leong Healthy Aging Program, Faculty of Medicine, University of British Columbia, Vancouver, Canada
- Centre for Molecular Medicine and Therapeutics, University of British Columbia, Vancouver, Canada
| | - Dany Doiron
- McGill University Health Centre, McGill University, Montreal, Canada
| | - Wan Tan
- Centre for Heart Lung Innovation, St. Paul'S Hospital and University of British Columbia, Room 166, 1081 Burrard Street, Vancouver, BC, V6Z 1Y6, Canada
| | - Jean Bourbeau
- McGill University Health Centre, McGill University, Montreal, Canada
| | - Don D Sin
- Centre for Heart Lung Innovation, St. Paul'S Hospital and University of British Columbia, Room 166, 1081 Burrard Street, Vancouver, BC, V6Z 1Y6, Canada
- Edwin S. H. Leong Healthy Aging Program, Faculty of Medicine, University of British Columbia, Vancouver, Canada
- Division of Respiratory Medicine, Department of Medicine, University of British Columbia, Vancouver, Canada
| | - Qingling Duan
- Department of Biomedical and Molecular Sciences, School of Medicine, Queen's University, Kingston, Canada
- School of Computing, Queen's University, Kingston, Canada
| | - Janice M Leung
- Centre for Heart Lung Innovation, St. Paul'S Hospital and University of British Columbia, Room 166, 1081 Burrard Street, Vancouver, BC, V6Z 1Y6, Canada.
- Edwin S. H. Leong Healthy Aging Program, Faculty of Medicine, University of British Columbia, Vancouver, Canada.
- Division of Respiratory Medicine, Department of Medicine, University of British Columbia, Vancouver, Canada.
| |
Collapse
|
24
|
Macías M, Alba-Linares JJ, Acha B, Blanco-Luquin I, Fernández AF, Álvarez-Jiménez J, Urdánoz-Casado A, Roldan M, Robles M, Cabezon-Arteta E, Alcolea D, de Gordoa JSR, Corroza J, Cabello C, Erro ME, Jericó I, Fraga MF, Mendioroz M. Advancing Personalized Medicine in Alzheimer's Disease: Liquid Biopsy Epigenomics Unveil APOE ε4-Linked Methylation Signatures. Int J Mol Sci 2025; 26:3419. [PMID: 40244264 PMCID: PMC11989983 DOI: 10.3390/ijms26073419] [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: 01/28/2025] [Revised: 03/31/2025] [Accepted: 04/03/2025] [Indexed: 04/18/2025] Open
Abstract
Recent studies show that patients with Alzheimer's disease (AD) harbor specific methylation marks in the brain that, if accessible, could be used as epigenetic biomarkers. Liquid biopsy enables the study of circulating cell-free DNA (cfDNA) fragments originated from dead cells, including neurons affected by neurodegenerative processes. Here, we isolated and epigenetically characterized plasma cfDNA from 35 patients with AD and 35 cognitively healthy controls by using the Infinium® MethylationEPIC BeadChip array. Bioinformatics analysis was performed to identify differential methylation positions (DMPs) and regions (DMRs), including APOE ε4 genotype stratified analysis. Plasma pTau181 (Simoa) and cerebrospinal fluid (CSF) core biomarkers (Fujirebio) were also measured and correlated with differential methylation marks. Validation was performed with bisulfite pyrosequencing and bisulfite cloning sequencing. Epigenome-wide cfDNA analysis identified 102 DMPs associated with AD status. Most DMPs correlated with clinical cognitive and functional tests including 60% for Mini-Mental State Examination (MMSE) and 80% for Global Deterioration Scale (GDS), and with AD blood and CSF biomarkers. In silico functional analysis connected 30 DMPs to neurological processes, identifying key regulators such as SPTBN4 and APOE genes. Several DMRs were annotated to genes previously reported to harbor epigenetic brain changes in AD (HKR1, ZNF154, HOXA5, TRIM40, ATG16L2, ADAMST2) and were linked to APOE ε4 genotypes. Notably, a DMR in the HKR1 gene, previously shown to be hypermethylated in the AD hippocampus, was validated in cfDNA from an orthogonal perspective. These results support the feasibility of studying cfDNA to identify potential epigenetic biomarkers in AD. Thus, liquid biopsy could improve non-invasive AD diagnosis and aid personalized medicine by detecting epigenetic brain markers in blood.
Collapse
Affiliation(s)
- Mónica Macías
- Neuroepigenetics Unit, Navarrabiomed, Hospital Universitario de Navarra, Universidad Pública de Navarra, Navarra Institute for Health Research (IdiSNA), 31008 Pamplona, Spain
| | - Juan José Alba-Linares
- Cancer Epigenetics and Nanomedicine Laboratory, Nanomaterials and Nanotechnology Research Center (CINN CSIC), 33940 El Entrego, Spain
- Health Research Institute of Asturias (ISPA FINBA), University of Oviedo, 33011 Oviedo, Spain
- Institute of Oncology of Asturias (IUOPA), University of Oviedo, 33006 Oviedo, Spain
- Rare Diseases CIBER (CIBERER) of the Carlos III Health Institute (ISCIII), 28029 Madrid, Spain
| | - Blanca Acha
- Neuroepigenetics Unit, Navarrabiomed, Hospital Universitario de Navarra, Universidad Pública de Navarra, Navarra Institute for Health Research (IdiSNA), 31008 Pamplona, Spain
| | - Idoia Blanco-Luquin
- Neuroepigenetics Unit, Navarrabiomed, Hospital Universitario de Navarra, Universidad Pública de Navarra, Navarra Institute for Health Research (IdiSNA), 31008 Pamplona, Spain
| | - Agustín F. Fernández
- Cancer Epigenetics and Nanomedicine Laboratory, Nanomaterials and Nanotechnology Research Center (CINN CSIC), 33940 El Entrego, Spain
- Health Research Institute of Asturias (ISPA FINBA), University of Oviedo, 33011 Oviedo, Spain
- Institute of Oncology of Asturias (IUOPA), University of Oviedo, 33006 Oviedo, Spain
- Rare Diseases CIBER (CIBERER) of the Carlos III Health Institute (ISCIII), 28029 Madrid, Spain
| | - Johana Álvarez-Jiménez
- Neuroepigenetics Unit, Navarrabiomed, Hospital Universitario de Navarra, Universidad Pública de Navarra, Navarra Institute for Health Research (IdiSNA), 31008 Pamplona, Spain
| | - Amaya Urdánoz-Casado
- Neuroepigenetics Unit, Navarrabiomed, Hospital Universitario de Navarra, Universidad Pública de Navarra, Navarra Institute for Health Research (IdiSNA), 31008 Pamplona, Spain
| | - Miren Roldan
- Neuroepigenetics Unit, Navarrabiomed, Hospital Universitario de Navarra, Universidad Pública de Navarra, Navarra Institute for Health Research (IdiSNA), 31008 Pamplona, Spain
| | - Maitane Robles
- Neuroepigenetics Unit, Navarrabiomed, Hospital Universitario de Navarra, Universidad Pública de Navarra, Navarra Institute for Health Research (IdiSNA), 31008 Pamplona, Spain
| | - Eneko Cabezon-Arteta
- Neuroepigenetics Unit, Navarrabiomed, Hospital Universitario de Navarra, Universidad Pública de Navarra, Navarra Institute for Health Research (IdiSNA), 31008 Pamplona, Spain
| | - Daniel Alcolea
- Department of Neurology, Institut d’Investigacions Biomèdiques Sant Pau (IIB Sant Pau), Hospital de la Santa Creu i Sant Pau, Universitat Autònoma de Barcelona, 08025 Barcelona, Spain
- Centro de Investigación Biomédica en Red en Enfermedades Neurodegenerativas, CIBERNED, 28029 Madrid, Spain
| | - Javier Sánchez Ruiz de Gordoa
- Neuroepigenetics Unit, Navarrabiomed, Hospital Universitario de Navarra, Universidad Pública de Navarra, Navarra Institute for Health Research (IdiSNA), 31008 Pamplona, Spain
- Neurology Department, Hospital Universitario de Navarra, Universidad Pública de Navarra, Navarra Institute for Health Research (IdiSNA), 31008 Pamplona, Spain
| | - Jon Corroza
- Neurology Department, Hospital Universitario de Navarra, Universidad Pública de Navarra, Navarra Institute for Health Research (IdiSNA), 31008 Pamplona, Spain
| | - Carolina Cabello
- Neuroepigenetics Unit, Navarrabiomed, Hospital Universitario de Navarra, Universidad Pública de Navarra, Navarra Institute for Health Research (IdiSNA), 31008 Pamplona, Spain
- Neurology Department, Hospital Universitario de Navarra, Universidad Pública de Navarra, Navarra Institute for Health Research (IdiSNA), 31008 Pamplona, Spain
| | - María Elena Erro
- Neuroepigenetics Unit, Navarrabiomed, Hospital Universitario de Navarra, Universidad Pública de Navarra, Navarra Institute for Health Research (IdiSNA), 31008 Pamplona, Spain
- Neurology Department, Hospital Universitario de Navarra, Universidad Pública de Navarra, Navarra Institute for Health Research (IdiSNA), 31008 Pamplona, Spain
| | - Ivonne Jericó
- Neurology Department, Hospital Universitario de Navarra, Universidad Pública de Navarra, Navarra Institute for Health Research (IdiSNA), 31008 Pamplona, Spain
| | - Mario F. Fraga
- Cancer Epigenetics and Nanomedicine Laboratory, Nanomaterials and Nanotechnology Research Center (CINN CSIC), 33940 El Entrego, Spain
- Health Research Institute of Asturias (ISPA FINBA), University of Oviedo, 33011 Oviedo, Spain
- Institute of Oncology of Asturias (IUOPA), University of Oviedo, 33006 Oviedo, Spain
- Rare Diseases CIBER (CIBERER) of the Carlos III Health Institute (ISCIII), 28029 Madrid, Spain
- Department of Organisms and Systems Biology (B.O.S.), University of Oviedo, 33006 Oviedo, Spain
| | - Maite Mendioroz
- Neuroepigenetics Unit, Navarrabiomed, Hospital Universitario de Navarra, Universidad Pública de Navarra, Navarra Institute for Health Research (IdiSNA), 31008 Pamplona, Spain
- Neurology Department, Hospital Universitario de Navarra, Universidad Pública de Navarra, Navarra Institute for Health Research (IdiSNA), 31008 Pamplona, Spain
| |
Collapse
|
25
|
Luo Q, Teschendorff AE. Cell-type-specific subtyping of epigenomes improves prognostic stratification of cancer. Genome Med 2025; 17:34. [PMID: 40181447 PMCID: PMC11967111 DOI: 10.1186/s13073-025-01453-5] [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: 08/15/2024] [Accepted: 03/10/2025] [Indexed: 04/05/2025] Open
Abstract
BACKGROUND Most molecular classifications of cancer are based on bulk-tissue profiles that measure an average over many distinct cell types. As such, cancer subtypes inferred from transcriptomic or epigenetic data are strongly influenced by cell-type composition and do not necessarily reflect subtypes defined by cell-type-specific cancer-associated alterations, which could lead to suboptimal cancer classifications. METHODS To address this problem, we here propose the novel concept of cell-type-specific combinatorial clustering (CELTYC), which aims to group cancer samples by the molecular alterations they display in specific cell types. We illustrate this concept in the context of DNA methylation data of liver and kidney cancer, deriving in each case novel cancer subtypes and assessing their prognostic relevance against current state-of-the-art prognostic models. RESULTS In both liver and kidney cancer, we reveal improved cell-type-specific prognostic models, not discoverable using standard methods. In the case of kidney cancer, we show how combinatorial indexing of epithelial and immune-cell clusters define improved prognostic models driven by synergy of high mitotic age and altered cytokine signaling. We validate the improved prognostic models in independent datasets and identify underlying cytokine-immune-cell signatures driving poor outcome. CONCLUSIONS In summary, cell-type-specific combinatorial clustering is a valuable strategy to help dissect and improve current prognostic classifications of cancer in terms of the underlying cell-type-specific epigenetic and transcriptomic alterations.
Collapse
Affiliation(s)
- Qi Luo
- CAS Key Laboratory of Computational Biology, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, 320 Yue Yang Road, Shanghai, 200031, China
| | - Andrew E Teschendorff
- CAS Key Laboratory of Computational Biology, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, 320 Yue Yang Road, Shanghai, 200031, China.
| |
Collapse
|
26
|
Hu Y, Haessler J, Lundin JI, Darst BF, Whitsel EA, Grove M, Guan W, Xia R, Szeto M, Raffield LM, Ratliff S, Wang Y, Wang X, Fohner AE, Lynch MT, Patel YM, Lani Park S, Xu H, Mitchell BD, Bis JC, Sotoodehnia N, Brody JA, Psaty BM, Peloso GM, Tsai MY, Rich SS, Rotter JI, Smith JA, Kardia SLR, Reiner AP, Lange L, Fornage M, Pankow JS, Graff M, North KE, Kooperberg C, Peters U. Methylome-wide association analyses of lipids and modifying effects of behavioral factors in diverse race and ethnicity participants. Clin Epigenetics 2025; 17:54. [PMID: 40176173 PMCID: PMC11967142 DOI: 10.1186/s13148-025-01859-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2024] [Accepted: 03/11/2025] [Indexed: 04/04/2025] Open
Abstract
Circulating lipid concentrations are clinically associated with cardiometabolic diseases. The phenotypic variance explained by identified genetic variants remains limited, highlighting the importance of searching for additional factors beyond genetic sequence variants. DNA methylation has been linked to lipid concentrations in previous studies, although most of the studies harbored moderate sample sizes and exhibited underrepresentation of non-European ancestry populations. In addition, knowledge of nongenetic factors on lipid profiles is extremely limited. In the Population Architecture Using Genomics and Epidemiology (PAGE) Study, we performed methylome-wide association analysis on 9,561 participants from diverse race and ethnicity backgrounds for HDL-c, LDL-c, TC, and TG levels, and also tested interactions between smoking or alcohol intake and methylation in their association with lipid levels. We identified novel CpG sites at 16 loci (P < 1.18E-7) with successful replication on 3,215 participants. One additional novel locus was identified in the self-reported White participants (P = 4.66E-8). Although no additional CpG sites were identified in the genome-wide interaction analysis, 13 reported CpG sites showed significant heterogeneous association across smoking or alcohol intake strata. By mapping novel and reported CpG sites to genes, we identified enriched pathways directly linked to lipid metabolism as well as ones spanning various biological functions. These findings provide new insights into the regulation of lipid concentrations.
Collapse
Grants
- N01HC95160 NHLBI NIH HHS
- 75N92021D00001, 75N92021D00002, 75N92021D00003, 75N92021D00004, 75N92021D00005, and S10OD028685 NIH HHS
- 75N92020D00001, HHSN268201500003I, N01-HC-95159, 75N92020D00005, N01-HC-95160, 75N92020D00002, N01-HC-95161, 75N92020D00003, N01-HC-95162, 75N92020D00006, N01-HC-95163, 75N92020D00004, N01-HC-95164, 75N92020D00007, N01-HC-95165, N01-HC-95166, N01-HC-95167, N01-HC-95168, N01-HC-95169, UL1-TR-000040, UL1-TR-001079, UL1-TR-001420, UL1TR001881, DK063491, HL148610, and R01HL105756 NHLBI NIH HHS
- R01 HL087652 NHLBI NIH HHS
- UL1 TR000040 NCATS NIH HHS
- HHSN268201800010I NHLBI NIH HHS
- N01HC85081 NHLBI NIH HHS
- R01 HL103612 NHLBI NIH HHS
- 75N92020D00002 NHLBI NIH HHS
- 75N92021D00002 NHLBI NIH HHS
- HHSN268201500003C NHLBI NIH HHS
- U01HG007397 NHGRI NIH HHS
- HHSN268201800012C NHLBI NIH HHS
- 75N92020D00005 NHLBI NIH HHS
- 75N92021D00005 WHI NIH HHS
- U01HL054457, RC1HL100185, R01HL087660, R01HL119443, R01HL133221 NHLBI NIH HHS
- 75N92022D00001 NIH HHS
- N01HC95163 NHLBI NIH HHS
- U01 HL080295 NHLBI NIH HHS
- UL1 TR001079 NCATS NIH HHS
- DK063491 National Institute of Diabetes and Digestive and Kidney Disease Diabetes Research Center
- HHSN268201800014I NHLBI NIH HHS
- U01CA164973 NCI NIH HHS
- U01 HL130114 NHLBI NIH HHS
- R01 HL087660 NHLBI NIH HHS
- HHSN268200800007C NHLBI NIH HHS
- S10 OD028685 NIH HHS
- 75N92020D00001 NHLBI NIH HHS
- N01HC95169 NHLBI NIH HHS
- N01HC95164 NHLBI NIH HHS
- UL1 TR000124 NCATS NIH HHS
- N01HC55222 NHLBI NIH HHS
- HHSN268201800014C NHLBI NIH HHS
- N01HC95162 NHLBI NIH HHS
- N01HC85086 NHLBI NIH HHS
- 75N92020D00003 NHLBI NIH HHS
- R01 HL119443 NHLBI NIH HHS
- R01 HL105756 NHLBI NIH HHS
- N01HC95168 NHLBI NIH HHS
- K08 HL116640 NHLBI NIH HHS
- 75N92021D00001 NHLBI NIH HHS
- P30 DK063491 NIDDK NIH HHS
- RC1 HL100185 NHLBI NIH HHS
- HHSN268201200036C NHLBI NIH HHS
- HHSN268201800001C NHLBI NIH HHS
- HHSN268201800013I NIMHD NIH HHS
- UL1TR000124 NCATS NIH HHS
- U01 HL054457 NHLBI NIH HHS
- N01HC95165 NHLBI NIH HHS
- N01HC95159 NHLBI NIH HHS
- HHSN268201800012I NHLBI NIH HHS
- 75N92021D00003 WHI NIH HHS
- N01HC95161 NHLBI NIH HHS
- UL1 TR001420 NCATS NIH HHS
- 75N92020D00004 NHLBI NIH HHS
- HHSN268201800011C NHLBI NIH HHS
- 75N92020D00007 NHLBI NIH HHS
- R01AG023629 NIA NIH HHS
- HHSN268201800013I, HHSN268201800014I, HHSN268201800015I, HHSN268201800010I, HHSN268201800011I, and HHSN268201800012I NIMHD NIH HHS
- HHSN268201500003I NHLBI NIH HHS
- R01HL105756, HHSN268201200036C, HHSN268200800007C, HHSN268201800001C, N01HC55222, N01HC85079, N01HC85080, N01HC85081, N01HC85082, N01HC85083, N01HC85086, R01AG023629, 75N92021D00006, U01HL080295, U01HL130114, K08HL116640, R01HL087652, R01HL092111, R01HL103612, R01HL111089, R01HL116747 and R01HL120393 NHLBI NIH HHS
- R01 HL133221 NHLBI NIH HHS
- 75N92021D00006 NHLBI NIH HHS
- R01HG010297 NHGRI NIH HHS
- N01HC85082 NHLBI NIH HHS
- N01HC95167 NHLBI NIH HHS
- N01HC85083 NHLBI NIH HHS
- HHSN268201800015I NHLBI NIH HHS
- 75N92020D00006 NHLBI NIH HHS
- N01HC85079 NHLBI NIH HHS
- N01HC95166 NHLBI NIH HHS
- R01 AG023629 NIA NIH HHS
- UL1 TR001881 NCATS NIH HHS
- HHSN268201800011I NHLBI NIH HHS
- N01HC85080 NHLBI NIH HHS
- R01 HG010297 NHGRI NIH HHS
- U01 CA164973 NCI NIH HHS
- 75N92021D00004 WHI NIH HHS
- R01 HL111089 NHLBI NIH HHS
- R01 HL116747 NHLBI NIH HHS
- R01 HL092111 NHLBI NIH HHS
- National Institutes of Health
- National Human Genome Research Institute
- National Institute on Minority Health and Health Disparities
- National Heart, Lung, and Blood Institute
- National Institute on Aging
- National Center for Advancing Translational Sciences
- National Cancer Institute
Collapse
Affiliation(s)
- Yao Hu
- Division of Public Health Sciences, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Jeff Haessler
- Division of Public Health Sciences, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Jessica I Lundin
- Division of Public Health Sciences, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Burcu F Darst
- Division of Public Health Sciences, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Eric A Whitsel
- Department of Epidemiology, University of North Carolina, Chapel Hill, NC, USA
| | - Megan Grove
- School of Public Health, Human Genetics Center, University of Texas Health Sciences Center at Houston, Houston, TX, USA
| | - Weihua Guan
- Division of Biostatistics and Health Data Science, School of Public Health, University of Minnesota, Minneapolis, MN, USA
| | - Rui Xia
- McGovern Medical School, The Brown Foundation Institute of Molecular Medicine, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Mindy Szeto
- Department of Epidemiology, Colorado School of Public Health, Aurora, CO, USA
| | - Laura M Raffield
- Department of Genetics, University of North Carolina, Chapel Hill, NC, USA
| | - Scott Ratliff
- Department of Epidemiology, School of Public Health, University of Michigan, Ann Arbor, MI, USA
| | - Yuxuan Wang
- Department of Biostatistics, Boston University School of Public Health, Boston, MA, USA
| | - Xuzhi Wang
- Department of Biostatistics, Boston University School of Public Health, Boston, MA, USA
| | - Alison E Fohner
- Cardiovascular Health Research Unit, Department of Medicine, University of Washington, Seattle, WA, USA
| | - Megan T Lynch
- Department of Medicine, University of Maryland School of Medicine, Baltimore, MD, USA
- Center for Immuno-Oncology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Yesha M Patel
- Department of Preventive Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - S Lani Park
- Population Sciences in the Pacific Program, University of Hawaii Cancer Center, Honolulu, HI, USA
| | - Huichun Xu
- Department of Medicine, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Braxton D Mitchell
- Department of Medicine, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Joshua C Bis
- Cardiovascular Health Research Unit, Department of Medicine, University of Washington, Seattle, WA, USA
| | - Nona Sotoodehnia
- Cardiovascular Health Research Unit, Department of Medicine, University of Washington, Seattle, WA, USA
| | - Jennifer A Brody
- Cardiovascular Health Research Unit, Department of Medicine, University of Washington, Seattle, WA, USA
| | - Bruce M Psaty
- Cardiovascular Health Research Unit, Department of Medicine, University of Washington, Seattle, WA, USA
| | - Gina M Peloso
- Department of Biostatistics, Boston University School of Public Health, Boston, MA, USA
| | - Michael Y Tsai
- Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, MN, USA
| | - Stephen S Rich
- Center for Public Health Genomics, University of Virginia, Charlottesville, VA, USA
| | - Jerome I Rotter
- Department of Pediatrics, The Institute for Translational Genomics and Population Sciences, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA, USA
| | - Jennifer A Smith
- Department of Epidemiology, School of Public Health, University of Michigan, Ann Arbor, MI, USA
| | - Sharon L R Kardia
- Department of Epidemiology, School of Public Health, University of Michigan, Ann Arbor, MI, USA
| | - Alex P Reiner
- Department of Epidemiology, University of Washington, Seattle, WA, USA
| | - Leslie Lange
- Department of Epidemiology, Colorado School of Public Health, Aurora, CO, USA
| | - Myriam Fornage
- School of Public Health, Human Genetics Center, University of Texas Health Sciences Center at Houston, Houston, TX, USA
- Human Genome Sequencing Center, Baylor College of Medicine, Houston, TX, USA
| | - James S Pankow
- Division of Epidemiology and Community Health, School of Public Health, University of Minnesota, Minneapolis, MN, USA
| | - Mariaelisa Graff
- Department of Epidemiology, University of North Carolina, Chapel Hill, NC, USA
| | - Kari E North
- Department of Epidemiology, University of North Carolina, Chapel Hill, NC, USA
| | - Charles Kooperberg
- Division of Public Health Sciences, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Ulrike Peters
- Division of Public Health Sciences, Fred Hutchinson Cancer Center, Seattle, WA, USA.
| |
Collapse
|
27
|
Wu Z, Dai Z, Feng Z, Qiu Y, Zhu Z, Xu L. Genome-wide methylation association study in monozygotic twins discordant for curve severity of adolescent idiopathic scoliosis. Spine J 2025; 25:785-796. [PMID: 39515527 DOI: 10.1016/j.spinee.2024.10.015] [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: 06/17/2024] [Revised: 10/03/2024] [Accepted: 10/27/2024] [Indexed: 11/16/2024]
Abstract
BACKGROUND CONTEXT Emerging evidence suggests that abnormal DNA methylation patterns may play a role in the progression of adolescent idiopathic scoliosis (AIS). However, the mechanisms underlying the influence of DNA methylation on curve severity remain largely unknown. PURPOSE To characterize the DNA methylation profiles associated with the curve severity of AIS. STUDY DESIGN Retrospective study with prospectively collected clinical data and blood samples. METHODS A total of 7 AIS monozygotic twin pairs discordant for curve severity were included. Genome-wide methylation profile from blood samples were quantified by Illumina Infinium MethylationEPIC BeadChip (850K chip). Cell type composition of the samples was estimated by RefbaseEWAS method. Differentially methylated CpG sites were identified through comparison between patients with low and high Cobb angle. We also performed a gene-based collapsing analysis using mCSEA by aggregating the CpG sites based on promoter region. Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analysis were performed using clusterProfiler package. RESULTS Genome-wide DNA methylation analysis identified multiple differentially methylated positions across the genome. Gene-based collapsing analysis identified 212 differentially methylated genes (FDR adjusted p<.05), most of which (186/212) were hypermethylated in the group with high Cobb angle. Some of the identified genes were well-documented in AIS literature, such as TBX1, PAX3 and LBX1. Functional enrichment analysis revealed that the differentially methylated genes (DMGs) were involved in pattern specification process, skeletal development, muscle function, neurotransmission and several signaling pathways (cAMP, Wnt and prolactin). CONCLUSIONS The study represents the largest systematic epigenomic analyses of monozygotic twins discordant for curve severity and supports the important role of altered DNA methylation in AIS. CLINICAL SIGNIFICANCE The identified CpG sites provide insight into novel biomarkers predicting curve progression of AIS. Furthermore, the differentially methylated genes and enriched pathways may serve as interventional therapy target for AIS patients.
Collapse
Affiliation(s)
- Zhichong Wu
- Division of Spine Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School; Nanjing, Jiangsu, China
| | - Zhicheng Dai
- Division of Spine Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School; Nanjing, Jiangsu, China
| | - Zhenhua Feng
- Division of Spine Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School; Nanjing, Jiangsu, China
| | - Yong Qiu
- Division of Spine Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School; Nanjing, Jiangsu, China
| | - Zezhang Zhu
- Division of Spine Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School; Nanjing, Jiangsu, China
| | - Leilei Xu
- Division of Spine Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School; Nanjing, Jiangsu, China.
| |
Collapse
|
28
|
Paparazzo E, Aceto MA, Serra Cassano T, Bruno F, Lagrotteria D, Geracitano S, La Russa A, Bauleo A, Falcone E, Lagani V, Passarino G, Montesanto A. Reproducibility and validation of a targeted and flexible epigenetic clock for forensic applications. Forensic Sci Int 2025; 369:112409. [PMID: 39983295 DOI: 10.1016/j.forsciint.2025.112409] [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: 12/20/2024] [Revised: 01/27/2025] [Accepted: 02/11/2025] [Indexed: 02/23/2025]
Abstract
DNA methylation variants have been widely used as biomarkers of ageing and several mathematical models have been developed to estimate the biological age. More recently, DNA technology has triggered efforts toward the simplification of the array-based epigenetic clocks and targeted approaches, based on the assessment of a small number of CpG sites have been developed. Among the markers included in these clocks, ELOVL2, FHL2, KLF14, C1orf132/MIR29B2C, and TRIM59 resulted to be the most strongly validated markers. We tested the reproducibility and validation of a previously developed targeted epigenetic clock purposely optimized for the measurement of chronological age in blood samples. The clock includes DNAm biomarkers strongly correlated with chronological age whose DNA methylation levels were measured by using a multiplex methylation SNaPshot assay. We found that epigenetic age, calculated using the developed clock, was highly correlated with age (r = 0.97) in a total of 201 blood samples covering a full spectrum of human ages. For 74 of these, methylation profiles of the whole genome were obtained through the Infinium Methylation EPIC v2.0 Kit which also allowed to estimate the most frequently used clocks of Horvath. These results show the potential of our efficient and affordable test for simultaneously measuring DNA methylation levels at multiple target CpG sites to assess chronological age. We observed a strong correlation between the prediction models for the analyzed CpG sites measured using the SNaPshot method and those obtained with the Illumina EPIC array, especially with the Horvath2 clock, which was specifically developed for DNA from skin and blood cells.
Collapse
Affiliation(s)
- Ersilia Paparazzo
- Department of Biology, Ecology and Earth Sciences, University of Calabria, Rende 87036, Italy
| | - Mirella Aurora Aceto
- Department of Biology, Ecology and Earth Sciences, University of Calabria, Rende 87036, Italy
| | - Teresa Serra Cassano
- Department of Biology, Ecology and Earth Sciences, University of Calabria, Rende 87036, Italy; University of Florence, Department of Statistic, Computer Science and Application, DiSIA, Viale Morgagni, 59, Florence, FI 50134, Italy
| | - Francesco Bruno
- Department of Human and Social Sciences, Faculty of Social and Communication Sciences, Universitas Mercatorum, Piazza Mattei 10, Rome 00186, Italy
| | - Davide Lagrotteria
- Department of Biology, Ecology and Earth Sciences, University of Calabria, Rende 87036, Italy
| | - Silvana Geracitano
- Department of Biology, Ecology and Earth Sciences, University of Calabria, Rende 87036, Italy
| | - Antonella La Russa
- Nephrology Unit, Department of Health Sciences, Magna Graecia University, Catanzaro 88100, Italy
| | - Alessia Bauleo
- BIOGENET, Medical and Forensic Genetics Laboratory, Cosenza, ASP 87100, Italy
| | - Elena Falcone
- BIOGENET, Medical and Forensic Genetics Laboratory, Cosenza, ASP 87100, Italy
| | - Vincenzo Lagani
- Biological and Environmental Sciences and Engineering Division (BESE), King Abdullah University of Science and Technology KAUST, Thuwal 23952, Saudi Arabia; Institute of Chemical Biology, Ilia State University, Tbilisi 0162, Georgia
| | - Giuseppe Passarino
- Department of Biology, Ecology and Earth Sciences, University of Calabria, Rende 87036, Italy
| | - Alberto Montesanto
- Department of Biology, Ecology and Earth Sciences, University of Calabria, Rende 87036, Italy.
| |
Collapse
|
29
|
Ishii T, Wang T, Shibata K, Nishitani S, Yamanashi T, Wahba NE, Seki T, Thompson KJ, Yamanishi K, Nishiguchi T, Shimura A, Aoyama B, Gorantla N, Phuong NJ, Nguyen HD, Santiago TA, Nishizawa Y, Nagao T, Howard MA, Kawasaki H, Hino K, Ikeda A, Snyder MP, Shinozaki G. Glial Contribution to the Pathogenesis of Post-Operative Delirium Revealed by Multi-omic Analysis of Brain Tissue from Neurosurgery Patients. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.13.643155. [PMID: 40161597 PMCID: PMC11952519 DOI: 10.1101/2025.03.13.643155] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 04/02/2025]
Abstract
Post-operative delirium (POD) is a common complication after surgery especially in elderly patients, characterized by acute disturbances in consciousness and cognition, which negatively impacts long-term outcomes. Effective treatments remain elusive due to the unclear pathophysiology of POD. To address the knowledge gap, we investigated DNA methylation profiles and gene expression changes in brain cells from POD and non-POD patients who underwent brain resection surgery for medication refractory epilepsy. DNA methylation analysis revealed alteration in epigenetic status of immune and inflammation-related genes. Single-nucleus RNA sequencing (snRNAseq) identified POD-specific glial cell alterations, particularly in microglia, where neuroinflammation was strongly enhanced, consistent with epigenetic findings. Astrocytes exhibited changes in synapse-related functions and migration. Furthermore, downstream analysis indicated similarities between POD-associated glial cell states and pathologies such as encephalitis and dementia. Overall, this study-the first multi-omics analysis of brain tissue from POD patients-provides direct evidence of glial cell contributions to POD pathogenesis, and highlights potential therapeutic targets.
Collapse
Affiliation(s)
- Takaya Ishii
- Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Palo Alto, California, USA
- Regenerative & Cellular Medicine Kobe Center, Sumitomo Pharma Co., Ltd., Osaka, Osaka, Japan
- Current affiliation is RACTHERA Co., Ltd., Kobe, Hyogo, Japan
| | - Tao Wang
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA
| | - Kazuki Shibata
- Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Palo Alto, California, USA
- Drug Research Division, Sumitomo Pharma Co., Ltd., Osaka, Osaka, Japan
| | - Shota Nishitani
- Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Palo Alto, California, USA
| | - Takehiko Yamanashi
- Faculty of Medicine, Department of Neuropsychiatry, Tottori University, Yonago, Tottori, Japan
| | - Nadia E. Wahba
- Department of Psychiatry, Oregon Health and Science University, School of Medicine, Portland, Oregon, USA
| | - Tomoteru Seki
- Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Palo Alto, California, USA
- Department of Psychiatry, Tokyo Medical University, Shinjuku-ku, Tokyo, Japan
| | | | - Kyosuke Yamanishi
- Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Palo Alto, California, USA
- Department of Neuropsychiatry, School of Medicine, Hyogo Medical University, Nishinomiya, Hyogo, Japan
| | - Tsuyoshi Nishiguchi
- Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Palo Alto, California, USA
- Faculty of Medicine, Department of Neuropsychiatry, Tottori University, Yonago, Tottori, Japan
| | - Akiyoshi Shimura
- Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Palo Alto, California, USA
- Department of Psychiatry, Tokyo Medical University, Shinjuku-ku, Tokyo, Japan
| | - Bun Aoyama
- Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Palo Alto, California, USA
- Department of Anesthesiology and Intensive Care Medicine, Kochi Medical School, Kochi, Kochi, Japan
| | - Nipun Gorantla
- Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Palo Alto, California, USA
| | - Nathan J. Phuong
- Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Palo Alto, California, USA
| | - Hieu D. Nguyen
- Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Palo Alto, California, USA
| | - Therese A. Santiago
- Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Palo Alto, California, USA
| | - Yoshitaka Nishizawa
- Department of Psychiatry, Osaka Medical and Pharmaceutical University School of Medicine, Osaka, Japan
| | - Takaaki Nagao
- Department of Neurosurgery (Sakura), Toho University School of Medicine Faculty of Medicine, Sakura, Chiba, Japan
| | - Mathew A Howard
- Department of Neurosurgery, University of Iowa Carver College of Medicine, Iowa City, IA, USA
| | - Hiroto Kawasaki
- Department of Neurosurgery, University of Iowa Carver College of Medicine, Iowa City, IA, USA
| | - Kyosuke Hino
- Regenerative & Cellular Medicine Kobe Center, Sumitomo Pharma Co., Ltd., Osaka, Osaka, Japan
- Current affiliation is RACTHERA Co., Ltd., Kobe, Hyogo, Japan
| | - Atsushi Ikeda
- Regenerative & Cellular Medicine Kobe Center, Sumitomo Pharma Co., Ltd., Osaka, Osaka, Japan
- Current affiliation is RACTHERA Co., Ltd., Kobe, Hyogo, Japan
| | - Michael P. Snyder
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA
| | - Gen Shinozaki
- Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Palo Alto, California, USA
- Department of Psychiatry, University of Iowa Carver College of Medicine, Iowa City, Iowa, USA
| |
Collapse
|
30
|
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
|
31
|
Yang M, Zheng C, Miao Y, Yin C, Tang L, Zhang C, Yu P, Han Q, Ma Y, Li S, Jiang G, Li W, Xia P. BTLA promoter hypomethylation correlates with enhanced immune cell infiltration, favorable prognosis, and immunotherapy response in melanoma. J Immunother Cancer 2025; 13:e009841. [PMID: 40081944 PMCID: PMC11907004 DOI: 10.1136/jitc-2024-009841] [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: 06/11/2024] [Accepted: 02/17/2025] [Indexed: 03/16/2025] Open
Abstract
BACKGROUND Immune checkpoint blockade (ICB)-based immunotherapy has significantly improved survival in advanced melanoma. However, many patients exhibit resistance to these therapies. This study examines the impact of BTLA promoter methylation on its expression, immune cell infiltration, and clinical outcomes, evaluating its potential as a prognostic and predictive biomarker for immunotherapy response. METHODS We analyzed methylation and gene expression data from public datasets (The Cancer Genome Atlas (TCGA), Gene Expression Omnibus (GEO)) and an in-house cohort of melanoma patients treated with ICB therapy at the First Affiliated Hospital of Zhengzhou University. We developed a quantitative methylation-specific PCR (qMSP) assay to measure methylation levels of the cg24157392 and cg03995631 CpG sites, and a targeted bisulfite sequencing assay was used to validate the accuracy of qMSP. We measured BTLA protein expression using multiplex immunofluorescence and immunohistochemical staining methods. Pearson correlation, survival analysis, and immune cell infiltration estimation were conducted to explore the associations between BTLA promoter methylation, mRNA and protein expression, clinical outcomes, and immune characteristics. RESULTS Hypomethylation at CpG sites cg24157392 and cg03995631 in the BTLA promoter were significantly associated with higher BTLA mRNA and protein expression. In the TCGA dataset, low methylation at these sites predicted longer overall survival and was validated in an independent cohort of 50 stage III/IV melanoma patients, with an area under the curve of 0.94 for predicting 5-year survival. Furthermore, BTLA promoter hypomethylation correlated with higher infiltration of immune cells, such as CD8+T cells, CD4+T cells, B cells, and macrophages. Additionally, low methylation at cg24157392 and cg03995631, as quantified by the qMSP assay, was significantly associated with better progression-free survival in patients treated with immune checkpoint inhibitors. These findings were further validated using GEO datasets. CONCLUSIONS BTLA promoter hypomethylation serves as a significant biomarker for favorable prognosis and enhanced response to ICB therapy in melanoma. The developed qMSP assays for cg24157392 and cg03995631 accurately quantified methylation levels and demonstrated their potential for clinical application in patient stratification and personalized immunotherapy.
Collapse
Affiliation(s)
- Minglei Yang
- Department of Pathology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Chenxi Zheng
- The Department of Dermatology, Henan Second Provincial People's Hospital, Zhengzhou, China
| | - Yu Miao
- Henan Academy of Sciences, Zhengzhou, Henan, China
| | - Cuicui Yin
- Department of Pathology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Longfei Tang
- Department of Pathology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Chongli Zhang
- Department of Pathology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Pu Yu
- Department of Oncology, First Affiliated Hospital of Zhengzhou University, Zhengzhou, Hennan, China
| | - Qingfang Han
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Yihui Ma
- First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Shenglei Li
- Department of Pathology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Guozhong Jiang
- Department of Pathology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Wencai Li
- Department of Pathology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Peiyi Xia
- Department of Pathology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| |
Collapse
|
32
|
León I, Góngora D, Rodrigo MJ, Herrero-Roldán S, López Rodríguez M, Mitchell C, Fisher J, Iturria-Medina Y. Maternal epigenetic index links early neglect to later neglectful care and other psychopathological, cognitive, and bonding effects. Clin Epigenetics 2025; 17:46. [PMID: 40057810 PMCID: PMC11890505 DOI: 10.1186/s13148-025-01839-7] [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: 09/28/2024] [Accepted: 02/09/2025] [Indexed: 05/13/2025] Open
Abstract
BACKGROUND Past experiences of maltreatment and life adversity induce DNA methylation changes in adults, but less is known about their impact on mothers' maladaptive neglectful parenting and its negative effects. We performed an epigenome-wide association study to investigate the role of DNA methylation levels in mothers with neglectful care, who were exposed to childhood maltreatment and neglect, and their current negative effects. Saliva DNA methylation was determined with the Illumina Human Methylation EPIC BeadChip v1. The individual epigenome was the input to a machine learning algorithm for trajectory inference, which assigned a specific state to each mother in the progression from healthy controls to the extreme neglect condition. A compound epigenetic maternal neglect score (EMN) was derived from 138 mothers (n = 51 in the neglectful group; n = 87 in the control non-neglectful group) having young children. Differential methylation between groups was utilized to derive the EMNs adjusted for education level, age, experimental variables, and blood cell types in saliva samples. RESULTS Structural equation modeling: X2 (29) = 37.81; p = 0.127; RMSEA = 0.048, confirmed that EMNs link their early experience of physical neglect to current reports of psychopathological symptoms, lower cognitive status, and observed poor mother-child emotional availability. A third of the genes annotated to the CpGs that affect EMNs are related to cognitive impairment and neurodegenerative and psychopathological disorders. CONCLUSIONS EMNs are a novel index to assess the contribution of DNA methylations as a neglected girl to later neglectful caregiving behavior and other negative effects. The evidence provided expands the possibilities for earlier interventions on the neglect condition to prevent and ameliorate the direct or indirect epigenetic impact of maternal adversities on mother-child care, helping to break the cycle of maltreatment.
Collapse
Affiliation(s)
- Inmaculada León
- Instituto Universitario de Neurociencia, Universidad de La Laguna, Campus de Guajara, 38201, San Cristóbal de La Laguna, Spain
- Facultad de Psicología, Universidad de La Laguna, San Cristóbal de La Laguna, Spain
| | - Daylín Góngora
- Department of Microeconomics and Public Economics, Maastricht University School of Business and Economics, Maastricht University - Center of Neuroeconomics, Maastricht, The Netherlands
- Faculty of Psychology and Neuroscience, Maastricht University, Maastricht, The Netherlands
| | - María José Rodrigo
- Instituto Universitario de Neurociencia, Universidad de La Laguna, Campus de Guajara, 38201, San Cristóbal de La Laguna, Spain
- Facultad de Psicología, Universidad de La Laguna, San Cristóbal de La Laguna, Spain
| | - Silvia Herrero-Roldán
- Instituto Universitario de Neurociencia, Universidad de La Laguna, Campus de Guajara, 38201, San Cristóbal de La Laguna, Spain.
- Facultad de Ciencias Sociales Aplicadas y de La Comunicación, UNIE Universidad, Madrid, Spain.
| | - Maykel López Rodríguez
- Department of Pathology and Laboratory Medicine at the David Geffen School of Medicine, UCLA, Los Angeles, USA
| | - Colter Mitchell
- Institute for Social Research, University of Michigan, Ann Arbor, MI, USA
| | - Jonah Fisher
- Institute for Social Research, University of Michigan, Ann Arbor, MI, USA
| | - Yasser Iturria-Medina
- Neurology and Neurosurgery Department, Montreal Neurological Institute, Montreal, Canada
- McConnell Brain Imaging Centre, Montreal Neurological Institute, Montreal, Canada
- Ludmer Centre for Neuroinformatics and Mental Health, Montreal, Canada
| |
Collapse
|
33
|
Antonacci C, Abballe L, Patrizi S, Pedace L, Barresi S, Giovannoni I, Tancredi C, Vinciarelli F, Megaro G, Carai A, Rossi S, Locatelli F, Mastronuzzi A, Miele E. DNA methylation profiling from cerebrospinal fluid as a diagnostic tool for pineoblastoma. Acta Neuropathol Commun 2025; 13:52. [PMID: 40057797 PMCID: PMC11889783 DOI: 10.1186/s40478-025-01960-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: 01/13/2025] [Accepted: 02/15/2025] [Indexed: 05/13/2025] Open
Abstract
Pineoblastoma is a rare and aggressive malignancy that often affects pediatric populations. Accurate diagnosis is challenging due to histological overlap with other central nervous system tumors and limited molecular data. DNA methylation profiling and analysis of circulating tumor DNA (derived from both cell dissemination as well as cell-free- cfDNA) in cerebrospinal fluid (CSF) are emerging tools for precise tumor classification, in the field of pediatric central nervous system tumors. Here, we report a challenging case of a 17-year-old refugee girl with a previous diagnosis of a primitive neuroectodermal tumor. Formalin-fixed, paraffin-embedded tissue was not available for histopathological re-evaluation. However, the methylation profiling of low amount of CSF-derived DNA classified the tumor as "pineoblastoma, subtype miRNA processing altered 1, subclass A," enabling patient management. The diagnosis was later confirmed through tissue-based DNA methylation analysis of a secondary lesion, demonstrating that the epigenetic signature faithfully reflected tumor features. This case report highlights the potential of CSF-based DNA methylation profiling as a minimally invasive yet accurate diagnostic tool for pediatric CNS tumors. The concordance between CSF and tissue profiling supports the integration of liquid biopsy into diagnostic workflows, allowing for earlier diagnosis and personalized treatment strategies. However, more studies are needed to demonstrate the reliability of our approach in other CNS malignancies.
Collapse
Affiliation(s)
- Celeste Antonacci
- Onco-Hematology, Cell Therapy, Gene Therapies and Hemopoietic Transplant, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Luana Abballe
- Onco-Hematology, Cell Therapy, Gene Therapies and Hemopoietic Transplant, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy.
| | - Sara Patrizi
- Onco-Hematology, Cell Therapy, Gene Therapies and Hemopoietic Transplant, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Lucia Pedace
- Onco-Hematology, Cell Therapy, Gene Therapies and Hemopoietic Transplant, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Sabina Barresi
- Pathology Unit, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | | | - Chantal Tancredi
- Pathology Unit, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Flavia Vinciarelli
- Onco-Hematology, Cell Therapy, Gene Therapies and Hemopoietic Transplant, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Giacomina Megaro
- Onco-Hematology, Cell Therapy, Gene Therapies and Hemopoietic Transplant, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Andrea Carai
- Neurosurgery Unit, Department of Neuroscience and Neurorehabilitation, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Sabrina Rossi
- Pathology Unit, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Franco Locatelli
- Onco-Hematology, Cell Therapy, Gene Therapies and Hemopoietic Transplant, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
- Department of Life Sciences and Public Health, Catholic University of the Sacred Heart, Rome, Italy
| | - Angela Mastronuzzi
- Onco-Hematology, Cell Therapy, Gene Therapies and Hemopoietic Transplant, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Evelina Miele
- Onco-Hematology, Cell Therapy, Gene Therapies and Hemopoietic Transplant, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy.
| |
Collapse
|
34
|
Lopez-Pleguezuelos C, Aguado-Barrera ME, Carballo-Castro A, Peleteiro P, Calvo-Crespo P, Taboada-Valladares B, Lobato-Busto R, Fuentes-Ríos O, Galego-Carro J, Coedo-Costa C, Gómez-Caamaño A, Vega A. Epigenome-wide analysis reveals potential biomarkers for radiation-induced toxicity risk in prostate cancer. Clin Epigenetics 2025; 17:43. [PMID: 40050897 PMCID: PMC11887099 DOI: 10.1186/s13148-025-01846-8] [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: 08/02/2024] [Accepted: 02/17/2025] [Indexed: 03/09/2025] Open
Abstract
BACKGROUND Prostate cancer is the second most common cancer globally, with radiation therapy (RT) being a key treatment for clinically localized and locally advanced cases. Given high survival rates, addressing long-term side effects of RT is crucial for preserving quality-of-life. Radiogenomics, the study of genetic variations affecting response to radiation, has primarily focussed on genomic biomarkers, while DNA methylation studies offer insights into RT responses. Although most research has centred on tumours, no epigenome-wide association studies have explored peripheral blood biomarkers of RT-induced toxicities in prostate cancer patients. Identifying such biomarkers could reveal molecular mechanisms underlying RT response and enable personalized treatment. METHODS We analysed 105 prostate cancer patients (52 cases and 53 controls). Cases developed grade ≥ 2 genitourinary and/or gastrointestinal late toxicity after 12 months of starting RT, whereas controls did not. An epigenome-wide association study of post-RT toxicities was performed using the Illumina MethylationEPIC BeadChip, adjusting for age and cell type composition. We constructed two methylation risk scores-one using differentially methylated positions (MRSsites) and another using differentially methylated regions (MRSregions)-as well as a Support Vector Machine-based methylation signature (SVMsites). We evaluated RT effects on biological age and stochastic epigenetic mutations within established radiation response pathways. Gene Ontology and pathway enrichment analyses were also performed. RESULTS Pre-RT methylation analysis identified 56 differentially methylated positions (adjusted p-value ≤ 0.05), and 6 differentially methylated regions (p-value ≤ 0.05) associated with the genes NTM, ACAP1, IL1RL2, VOOP1, AKR1E2, and an intergenic region on chromosome 13 related to Short/Long Interspersed Nuclear Elements. Both Methylation Risk Scores (MRSsites AUC = 0.87; MRSregions AUC = 0.89) and the 8-CpG Support Vector Machine signature (SVMsites AUC = 0.98) exhibited strong discriminatory accuracy in classifying patients in the discovery cohort. Gene ontology analysis revealed significant enrichment (adjusted p-value ≤ 0.05) of genes involved in DNA repair, inflammatory response, tissue repair, and oxidative stress response pathways. CONCLUSIONS Epigenetic biomarkers show potential for predicting severe long-term adverse effects of RT in prostate cancer patients. The identified methylation patterns provide valuable insights into toxicity mechanisms and may aid personalized treatment strategies. However, validation in independent cohorts is essential to confirm their predictive value and clinical applicability.
Collapse
Affiliation(s)
- Carlos Lopez-Pleguezuelos
- Genetics in Cancer and Rare Diseases Group, Instituto de Investigación Sanitaria de Santiago de Compostela, Santiago de Compostela, Spain
- Fundación Pública Galega de Medicina Xenómica, Hospital Clínico Universitario de Santiago de Compostela, Servizo Galego de Saúde (SERGAS), Edificio de Consultas, Planta Menos 2, Choupana S/N, 15706, Santiago de Compostela, Spain
| | - Miguel E Aguado-Barrera
- Genetics in Cancer and Rare Diseases Group, Instituto de Investigación Sanitaria de Santiago de Compostela, Santiago de Compostela, Spain
- Fundación Pública Galega de Medicina Xenómica, Hospital Clínico Universitario de Santiago de Compostela, Servizo Galego de Saúde (SERGAS), Edificio de Consultas, Planta Menos 2, Choupana S/N, 15706, Santiago de Compostela, Spain
- Grupo de Medicina Xenómica, Universidade de Santiago de Compostela, Santiago de Compostela, Spain
| | - Ana Carballo-Castro
- Department of Radiation Oncology, Hospital Clínico Universitario de Santiago de Compostela, Servizo Galego de Saúde (SERGAS), Santiago de Compostela, Spain
| | - Paula Peleteiro
- Genetics in Cancer and Rare Diseases Group, Instituto de Investigación Sanitaria de Santiago de Compostela, Santiago de Compostela, Spain
- Department of Radiation Oncology, Hospital Clínico Universitario de Santiago de Compostela, Servizo Galego de Saúde (SERGAS), Santiago de Compostela, Spain
| | - Patricia Calvo-Crespo
- Genetics in Cancer and Rare Diseases Group, Instituto de Investigación Sanitaria de Santiago de Compostela, Santiago de Compostela, Spain
- Department of Radiation Oncology, Hospital Clínico Universitario de Santiago de Compostela, Servizo Galego de Saúde (SERGAS), Santiago de Compostela, Spain
| | - Begoña Taboada-Valladares
- Genetics in Cancer and Rare Diseases Group, Instituto de Investigación Sanitaria de Santiago de Compostela, Santiago de Compostela, Spain
- Department of Radiation Oncology, Hospital Clínico Universitario de Santiago de Compostela, Servizo Galego de Saúde (SERGAS), Santiago de Compostela, Spain
| | - Ramón Lobato-Busto
- Department of Medical Physics, Hospital Clínico Universitario de Santiago de Compostela, Servizo Galego de Saúde (SERGAS), Santiago de Compostela, Spain
| | - Olivia Fuentes-Ríos
- Genetics in Cancer and Rare Diseases Group, Instituto de Investigación Sanitaria de Santiago de Compostela, Santiago de Compostela, Spain
- Fundación Pública Galega de Medicina Xenómica, Hospital Clínico Universitario de Santiago de Compostela, Servizo Galego de Saúde (SERGAS), Edificio de Consultas, Planta Menos 2, Choupana S/N, 15706, Santiago de Compostela, Spain
| | - Javier Galego-Carro
- Genetics in Cancer and Rare Diseases Group, Instituto de Investigación Sanitaria de Santiago de Compostela, Santiago de Compostela, Spain
- Fundación Pública Galega de Medicina Xenómica, Hospital Clínico Universitario de Santiago de Compostela, Servizo Galego de Saúde (SERGAS), Edificio de Consultas, Planta Menos 2, Choupana S/N, 15706, Santiago de Compostela, Spain
- Grupo de Medicina Xenómica, Universidade de Santiago de Compostela, Santiago de Compostela, Spain
| | - Carla Coedo-Costa
- Genetics in Cancer and Rare Diseases Group, Instituto de Investigación Sanitaria de Santiago de Compostela, Santiago de Compostela, Spain
- Fundación Pública Galega de Medicina Xenómica, Hospital Clínico Universitario de Santiago de Compostela, Servizo Galego de Saúde (SERGAS), Edificio de Consultas, Planta Menos 2, Choupana S/N, 15706, Santiago de Compostela, Spain
| | - Antonio Gómez-Caamaño
- Genetics in Cancer and Rare Diseases Group, Instituto de Investigación Sanitaria de Santiago de Compostela, Santiago de Compostela, Spain
- Department of Radiation Oncology, Hospital Clínico Universitario de Santiago de Compostela, Servizo Galego de Saúde (SERGAS), Santiago de Compostela, Spain
| | - Ana Vega
- Genetics in Cancer and Rare Diseases Group, Instituto de Investigación Sanitaria de Santiago de Compostela, Santiago de Compostela, Spain.
- Fundación Pública Galega de Medicina Xenómica, Hospital Clínico Universitario de Santiago de Compostela, Servizo Galego de Saúde (SERGAS), Edificio de Consultas, Planta Menos 2, Choupana S/N, 15706, Santiago de Compostela, Spain.
- Grupo de Medicina Xenómica, Universidade de Santiago de Compostela, Santiago de Compostela, Spain.
- Biomedical Network on Rare Diseases (CIBERER), Madrid, Spain.
| |
Collapse
|
35
|
Li S, Kuan PF. A systematic evaluation of cell-type-specific differential methylation analysis in bulk tissue. Brief Bioinform 2025; 26:bbaf170. [PMID: 40237763 PMCID: PMC12001786 DOI: 10.1093/bib/bbaf170] [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: 12/01/2024] [Revised: 03/01/2025] [Accepted: 03/24/2025] [Indexed: 04/18/2025] Open
Abstract
We conducted a systematic assessment of computational models-CellDMC, TCA, HIRE, TOAST, and CeDAR-for detecting cell-type-specific differential methylation CpGs in bulk methylation data profiled using the Illumina DNA Methylation BeadArrays. This assessment was performed through simulations and case studies involving two epigenome-wide association studies (EWAS) on rheumatoid arthritis and major depressive disorder. Our evaluation provided insights into the strengths and limitations of each model. The results revealed that the models varied in performance across different metrics, sample sizes, and computational efficiency. Additionally, we proposed integrating the results from these models using the minimum p-value ($minpv$) and average p-value ($avepv$) approaches. Our findings demonstrated that these aggregation methods significantly improved performance in identifying cell-type-specific differential methylation CpGs.
Collapse
Affiliation(s)
- Shuo Li
- Department of Applied Mathematics and Statistics, Stony Brook University, Nicolls Road, 11794, New York, USA
| | - Pei Fen Kuan
- Department of Applied Mathematics and Statistics, Stony Brook University, Nicolls Road, 11794, New York, USA
| |
Collapse
|
36
|
Lukacsovich D, Zambare W, Wu C, Huang H, Zhang W, Kim MJ, Alvarez J, Bercz A, Paty PB, Romesser PB, Wang L, Smith JJ, Chen XS. Integrating Tumor and Organoid DNA Methylation Profiles Reveals Robust Predictors of Chemotherapy Response in Rectal Cancer. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2025:2025.02.28.25322951. [PMID: 40093220 PMCID: PMC11908278 DOI: 10.1101/2025.02.28.25322951] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 03/19/2025]
Abstract
Rectal cancer patients display heterogeneous responses to neoadjuvant treatment-including the intensive total neoadjuvant therapy (TNT)-and reliable biomarkers are lacking to guide which tumors will benefit most from these regimens. Here, we profiled DNA methylation in tumor tissue and matched patient-derived organoids (PDOs) from 18 rectal cancer cases (50 total samples), leveraging the Illumina MethylationEPIC array and quality control filters that retained 771,964 CpG sites. Analyses used linear models (for tissue-only or PDO-only) and a joint linear mixed-effects approach (accounting for patient-level random effects) to identify significant CpGs associated with log-transformed FOLFOX IC50. We found that PDOs faithfully recapitulate patient-tumor methylation patterns (Spearman's correlation >0.95 among replicate organoids), and the joint model uncovered 745 CpGs tied to FOLFOX sensitivity, many of which were missed in tissue-only analyses. Differentially methylated regions reinforced that broader epigenetic blocks near TSS or enhancer regions may modulate chemo-resistance, while pathway enrichment pinpointed focal adhesion, ECM-receptor interaction, calcium signaling, and folate metabolism as key processes. A methylation risk score derived from these CpGs significantly predicted progression-free survival in an independent colorectal cancer cohort (p=0.019), outperforming single-sample-based signatures. These findings suggest that combining methylation profiles from both tumors and PDOs can expose robust epigenetic drivers of therapy response, aiding the development of clinically actionable biomarkers for rectal cancer TNT.
Collapse
Affiliation(s)
- David Lukacsovich
- 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
- 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
- Department of Radiation Oncology, Colorectal and Anal Cancer Service, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA
- Colorectal Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA
| | - Wini Zambare
- Colorectal Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA
| | - Chao Wu
- Colorectal Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA
| | - Hanchen Huang
- 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
- 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
- Department of Radiation Oncology, Colorectal and Anal Cancer Service, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA
- Colorectal Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA
| | - Wei Zhang
- Department of Public Health Sciences, University of Miami Miller School of Medicine, Miami, FL, 33136, USA
| | - Min Jung Kim
- Colorectal Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA
| | - Janet Alvarez
- Colorectal Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA
| | - Aron Bercz
- Colorectal Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA
| | - Philip B Paty
- Colorectal Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA
| | - Paul B Romesser
- Department of Radiation Oncology, Colorectal and Anal Cancer Service, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA
| | - Lily Wang
- 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
- 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
| | - J Joshua Smith
- Colorectal Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA
| | - X Steven Chen
- 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
| |
Collapse
|
37
|
Ambrosone CB, Yao S, Long MD, Liu C, Chen J, Davis W, Zirpoli G, Payne-Ondracek R, Khoury T, Gong Z, Hu Q, Szewczyk S, Omilian AR, Bandera EV, Liu S, Kushi L, Higgins MJ, Palmer JR. Associations of DNA methylation in breast tumour subtypes with parity and breastfeeding in a cohort of 1459 Black women: implications for public health. BMJ ONCOLOGY 2025; 4:e000675. [PMID: 40519219 PMCID: PMC12164313 DOI: 10.1136/bmjonc-2024-000675] [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: 11/22/2024] [Accepted: 02/17/2025] [Indexed: 06/18/2025]
Abstract
Objective Having children reduces risk of breast cancer overall, but parity without breastfeeding, more prevalent among black women, increases risk of poor-prognosis oestrogen receptor negative (ER-) breast cancer. We investigated if relationships between parity, breastfeeding and ER subtypes result from epigenetic programming, potentially steering breast progenitor cells to a basal-like phenotype. Methods and analysis The Illumina MethylationEPIC platform was used to assess genome-wide methylation in formalin-fixed, paraffin-embedded tumours from 1459 Black women with breast cancer. Methylation was evaluated in relation to parity, breastfeeding and breast cancer subtypes in a case-only analysis, with methylation-gene expression pairs tested in a subset of cases. We then performed functional enrichment analysis for probes significantly associated with parity and breastfeeding. Results Among women who did not breastfeed (n=634), there were 500 significant (p<1e-5) differentially methylated loci (DML) by parity, compared with only five DMLs among women who had breastfed their children (n=568). One of the top DML genes was FOXA1, pivotal in governing the luminal lineage of progenitor cells, with a statistically significant interaction (p=0.04) for number of births and breastfeeding. Associations were strongest for ER- disease. Conclusion In this large study of Black women with breast cancer, we elucidated biological pathways for the observed associations between parity without breastfeeding and breast cancer subtypes, revealing distinct molecular alterations in breast DNA, particularly for ER- tumours. Black women in the USA tend to have more children and are less likely to breastfeed; their breast cancer risk may be reduced by societal systems that promote and support breastfeeding.
Collapse
Affiliation(s)
- Christine B Ambrosone
- Cancer Prevention and Control, Roswell Park Cancer Institute, Buffalo, New York, USA
| | - Song Yao
- Cancer Prevention and Control, Roswell Park Cancer Institute, Buffalo, New York, USA
| | - Mark D Long
- Biostatistics and Bioinformatics, Roswell Park Cancer Institute, Buffalo, New York, USA
| | - Chunyu Liu
- Biostatistics, Boston University, Boston, Massachusetts, USA
| | - Jianhong Chen
- Cancer Prevention and Control, Roswell Park Cancer Institute, Buffalo, New York, USA
| | - Warren Davis
- Cancer Prevention and Control, Roswell Park Cancer Institute, Buffalo, New York, USA
| | - Gary Zirpoli
- Slone Epidemiology, Boston University, Boston, Massachusetts, USA
| | | | - Thaer Khoury
- Pathology, Roswell Park Cancer Institute, Buffalo, New York, USA
| | - Zhihong Gong
- Cancer Prevention and Control, Roswell Park Cancer Institute, Buffalo, New York, USA
| | - Qiang Hu
- Biostatistics and Bioinformatics, Roswell Park Cancer Institute, Buffalo, New York, USA
| | - Sirinapa Szewczyk
- Molecular and Cellular Biology, Roswell Park Cancer Institute, Buffalo, New York, USA
| | - Angela R Omilian
- Cancer Prevention and Control, Roswell Park Cancer Institute, Buffalo, New York, USA
| | - Elisa V Bandera
- Cancer Epidemiology and Health Outcomes, Rutgers The State University of New Jersey, New Brunswick, New Jersey, USA
| | - Song Liu
- Biostatistics and Bioinformatics, Roswell Park Cancer Institute, Buffalo, New York, USA
| | - Lawrence Kushi
- Division of Research, Kaiser Permanente, Oakland, California, USA
| | - Michael J Higgins
- Molecular and Cellular Biology, Roswell Park Cancer Institute, Buffalo, New York, USA
| | - Julie R Palmer
- Slone Epidemiology Center, Boston University, Boston, Massachusetts, USA
| |
Collapse
|
38
|
Everson TM, Sehgal N, Campbell K, Barr DB, Panuwet P, Yakimavets V, Chen K, Perez C, Shankar K, Eick SM, Pearson KJ, Andres A. Placental PFAS concentrations are associated with perturbations of placental DNA methylation. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2025; 368:125737. [PMID: 39862910 DOI: 10.1016/j.envpol.2025.125737] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/05/2024] [Revised: 01/12/2025] [Accepted: 01/21/2025] [Indexed: 01/27/2025]
Abstract
The placenta is crucial for fetal development, is affected by PFAS toxicity, and evidence is accumulating that gestational PFAS perturb the epigenetic activity of the placenta. Gestational PFAS exposure can adversely affect offspring, yet individual and cumulative impacts of PFAS on the placental epigenome remain underexplored. Here, we conducted an epigenome-wide association study (EWAS) to examine the relationships between placental PFAS levels and DNA methylation in a cohort of mother-infant dyads in Arkansas (N = 151). We measured 17 PFAS in human placental tissues and quantified placental DNA methylation levels via the Illumina EPIC Microarray. We tested for differential DNA methylation with individual PFAS, and with mixtures of multiple PFAS. Our results demonstrated that numerous epigenetic loci were perturbed by PFAS, with PFHxS exhibiting the most abundant effects. Mixture analyses suggested cumulative effects of PFOA and PFOS, while PFHxS may act more independently. We additionally explored whether sex-specific effects may be present and concluded that future large studies should explicitly test for sex-specific effects. The genes that are annotated to our PFAS-associated epigenetic loci are primarily involved in growth processes and cardiometabolic health, while some genes are involved in neurodevelopment. These findings shed light on how prenatal PFAS exposures affect birth outcomes and children's health, emphasizing the importance of understanding PFAS mechanisms in the in-utero environment.
Collapse
Affiliation(s)
- Todd M Everson
- Gangarosa Department of Environmental Health, Emory University Rollins School of Public Health, Atlanta, GA, USA; Department of Epidemiology, Emory University Rollins School of Public Health, Atlanta, GA, USA.
| | - Neha Sehgal
- Gangarosa Department of Environmental Health, Emory University Rollins School of Public Health, Atlanta, GA, USA
| | - Kyle Campbell
- Gangarosa Department of Environmental Health, Emory University Rollins School of Public Health, Atlanta, GA, USA
| | - Dana Boyd Barr
- Gangarosa Department of Environmental Health, Emory University Rollins School of Public Health, Atlanta, GA, USA
| | - Parinya Panuwet
- Gangarosa Department of Environmental Health, Emory University Rollins School of Public Health, Atlanta, GA, USA
| | - Volha Yakimavets
- Gangarosa Department of Environmental Health, Emory University Rollins School of Public Health, Atlanta, GA, USA
| | - Kelsey Chen
- Gangarosa Department of Environmental Health, Emory University Rollins School of Public Health, Atlanta, GA, USA
| | - Cynthia Perez
- Gangarosa Department of Environmental Health, Emory University Rollins School of Public Health, Atlanta, GA, USA
| | - Kartik Shankar
- USDA Agricultural Research Service, Responsive Agricultural Food Systems Research Unit, College Station, TX, USA
| | - Stephanie M Eick
- Gangarosa Department of Environmental Health, Emory University Rollins School of Public Health, Atlanta, GA, USA; Department of Epidemiology, Emory University Rollins School of Public Health, Atlanta, GA, USA
| | - Kevin J Pearson
- Department of Pharmacology & Nutritional Sciences, University of Kentucky College of Medicine, USA
| | - Aline Andres
- Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, AR, USA; Arkansas Children's Nutrition Center, Little Rock, AR, USA
| |
Collapse
|
39
|
Yang J, Chen S, Liu Y, Wang P, Zhao J, Yi J, Wei J, Wang R. Identification of a novel hypermethylation marker, ZSCAN18, and construction of a diagnostic model in cervical cancer. Clin Transl Oncol 2025:10.1007/s12094-025-03864-7. [PMID: 39969762 DOI: 10.1007/s12094-025-03864-7] [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: 12/17/2024] [Accepted: 01/28/2025] [Indexed: 02/20/2025]
Abstract
PURPOSE Cervical cancer (CC), a common female malignancy, has been linked to alterations in DNA methylation. This study employed an integrated "dry-wet lab" strategy combining bioinformatics, machine learning, and experimental validation to identify novel methylation biomarkers for CC. METHODS Methylome and transcriptome data from the TCGA and GEO cohorts (n=349 discovery, n=414 validation) were analyzed to identify differentially methylated CpGs. The top candidates were validated by pyrosequencing, methylation-specific PCR, and quantitative assays. Diagnostic models were developed, and functional studies were performed for the target markers. RESULTS Eighteen differentially methylated CpGs were identified, with five top candidates (three in the ZSCAN18 promoter) showing diagnostic potential. ZSCAN18 promoter methylation levels and positivity rates were significantly greater in CC tissues than in normal tissues (p<0.05), reaching 77.8% (21/27) in ThinPrep cytology test (TCT) samples. The ridge regression diagnostic model achieved an AUC of 0.9421 in the validation cohort. Similarly, ZSCAN18 overexpression suppressed CC cell proliferation (p<0.05). CONCLUSIONS This study established a rapid, effective and systematic systemic research strategy to screen novel methylation markers for CC. ZSCAN18 promoter methylation correlates with cervical lesion severity, and the diagnostic model enhances the diagnostic ability. These findings highlight the dual role of ZSCAN18 as a diagnostic marker and potential therapeutic target.
Collapse
Affiliation(s)
- Jinhao Yang
- Department of Laboratory Medicine, School of Medical Technology, Tianjin Medical University, Tianjin, 300203, China
| | - Shuang Chen
- Department of Laboratory Medicine, School of Medical Technology, Tianjin Medical University, Tianjin, 300203, China
| | - Yuqing Liu
- Department of Laboratory Medicine, School of Medical Technology, Tianjin Medical University, Tianjin, 300203, China
| | - Ping Wang
- Department of Laboratory Medicine, School of Medical Technology, Tianjin Medical University, Tianjin, 300203, China
| | - Jing Zhao
- Department of Gynecology and Obstetrics, Tianjin Medical University General Hospital, Tianjin, 300041, China
| | - Jianying Yi
- Department of Clinical Laboratory, Tianjin First Central Hospital, School of Medicine, Nankai University, Tianjin, 300192, China
| | - Jin Wei
- Tianjin Key Laboratory of Extracorporeal Life Support for Critical Diseases,Tianjin Third Central Hospital, Tianjin, 300170, China
| | - Rong Wang
- Department of Laboratory Medicine, School of Medical Technology, Tianjin Medical University, Tianjin, 300203, China.
| |
Collapse
|
40
|
Li Y, Goodrich JM, Peterson KE, Song PXK, Luo L. Uncertainty quantification in epigenetic clocks via conformalized quantile regression. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2025:2024.09.06.24313192. [PMID: 39281769 PMCID: PMC11398601 DOI: 10.1101/2024.09.06.24313192] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 09/18/2024]
Abstract
DNA methylation (DNAm) is a chemical modification of DNA that can be influenced by various factors, including age, the environment, and lifestyle. An epigenetic clock is a predictive tool that measures biological age based on DNAm levels. It can provide insights into an individual's biological age, which may differ from their chronological age. This difference, known as the epigenetic age acceleration, may reflect health status and the risk for age-related diseases. Moreover, epigenetic clocks are used in studies of aging to assess the effectiveness of anti-aging interventions and to understand the underlying mechanisms of aging and disease. Various epigenetic clocks have been developed using samples from different populations, tissues, and cell types, typically by training high-dimensional linear regression models with an elastic net penalty. While these models can predict mean biological age based on DNAm with high precision, there is a lack of uncertainty quantification which is important for interpreting the precision of age estimations and for clinical decision-making. To understand the distribution of a biological age clock beyond its mean, we propose a general pipeline for training epigenetic clocks, based on an integration of high-dimensional quantile regression and conformal prediction, to effectively reveal population heterogeneity and construct prediction intervals. Our approach produces adaptive prediction intervals not only achieving nominal coverage but also accounting for the inherent variability across individuals. By using the data collected from 728 blood samples in 11 DNAm datasets from children, we find that our quantile regression-based prediction intervals are narrower than those derived from conventional mean regression-based epigenetic clocks. This observation demonstrates an improved statistical efficiency over the existing pipeline for training epigenetic clocks. In addition, the resulting intervals have a synchronized varying pattern to age acceleration, effectively revealing cellular evolutionary heterogeneity in age patterns in different developmental stages during individual childhoods and adolescent cohort. Our findings suggest that conformalized high-dimensional quantile regression can produce valid prediction intervals and uncover underlying population heterogeneity. Although our methodology focuses on the distribution of measures of biological aging in children, it is applicable to a broader range of age groups to improve understanding of epigenetic age beyond the mean. This inference-based toolbox could provide valuable insights for future applications of epigenetic interventions for age-related diseases.
Collapse
Affiliation(s)
- Yanping Li
- School of Statistics and Data Science, Nankai University, China
| | - Jaclyn M. Goodrich
- Department of Environmental Health Sciences, University of Michigan, Ann Arbor, USA
| | - Karen E Peterson
- Department of Nutritional Sciences, University of Michigan, Ann Arbor, USA
| | - Peter X-K Song
- Department of Biostatistics, University of Michigan, Ann Arbor, USA
| | - Lan Luo
- Department of Biostatistics and Epidemiology, Rutgers University, USA
| |
Collapse
|
41
|
Kelly C, Kiltschewskij DJ, Leong AJW, Haw TJ, Croft AJ, Balachandran L, Chen D, Bond DR, Lee HJ, Cairns MJ, Sverdlov AL, Ngo DTM. Identifying common pathways for doxorubicin and carfilzomib-induced cardiotoxicities: transcriptomic and epigenetic profiling. Sci Rep 2025; 15:4395. [PMID: 39910168 PMCID: PMC11799237 DOI: 10.1038/s41598-025-87442-5] [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: 03/13/2024] [Accepted: 01/20/2025] [Indexed: 02/07/2025] Open
Abstract
Cancer therapy-related cardiovascular toxicity (CTR-CVT) is now recognised as one of the leading causes of long-term morbidity and mortality in cancer patients. To date, potential overlapping cardiotoxicity mechanism(s) across different chemotherapeutic classes have not been elucidated. Doxorubicin, an anthracycline, and Carfilzomib, a proteasome inhibitor, are both known to cause heart failure in some patients. Given this common cardiotoxic effect of these chemotherapies, we aimed to investigate differential and common mechanism(s) associated with Doxorubicin and Carfilzomib-induced cardiac dysfunction. Primary human cardiomyocyte-like cells (HCM-ls) were treated with 1 µM of either Doxorubicin or Carfilzomib for 72 h. Both Doxorubicin and Carfilzomib induced a significant reduction in HCM cell viability and cell damage. DNA methylation analysis performed using MethylationEPIC array showed distinct and common changes induced by Doxorubicin and Carfilzomib (10,270 or approximately 12.9% of the DMPs for either treatment overlapped). RNA-seq analyses identified 5,643 differentially expressed genes (DEGs) that were commonly dysregulated for both treatments. Pathway analysis revealed that the PI3K-Akt signalling pathway was the most significantly enriched pathway with common DEGs, shared between Doxorubicin and Carfilzomib. We identified that there are shared cardiotoxicity mechanisms for Doxorubicin and Carfilzomib pathways that can be potential therapeutic targets for treatments across 2 classes of anti-cancer agents.
Collapse
Affiliation(s)
- Conagh Kelly
- School of Biomedical Sciences and Pharmacy, The University of Newcastle, Callaghan, NSW, 2305, Australia
- Heart and Stroke Research Program, Hunter Medical Research Institute, New Lambton, NSW, Australia
- Newcastle Centre of Excellence in Cardio-Oncology, Hunter Medical Research Institute, Hunter New England Local Health District, University of Newcastle and Calvary Mater Newcastle, Newcastle, NSW, Australia
| | - Dylan J Kiltschewskij
- School of Biomedical Sciences and Pharmacy, The University of Newcastle, Callaghan, NSW, 2305, Australia
- Precision Medicine Research Program, Hunter Medical Research Institute, New Lambton, NSW, Australia
| | - Angeline J W Leong
- Heart and Stroke Research Program, Hunter Medical Research Institute, New Lambton, NSW, Australia
- Newcastle Centre of Excellence in Cardio-Oncology, Hunter Medical Research Institute, Hunter New England Local Health District, University of Newcastle and Calvary Mater Newcastle, Newcastle, NSW, Australia
- School of Medicine and Public Health, University of Newcastle, Callaghan, NSW, 2305, Australia
| | - Tatt Jhong Haw
- Heart and Stroke Research Program, Hunter Medical Research Institute, New Lambton, NSW, Australia
- Newcastle Centre of Excellence in Cardio-Oncology, Hunter Medical Research Institute, Hunter New England Local Health District, University of Newcastle and Calvary Mater Newcastle, Newcastle, NSW, Australia
- School of Medicine and Public Health, University of Newcastle, Callaghan, NSW, 2305, Australia
| | - Amanda J Croft
- Heart and Stroke Research Program, Hunter Medical Research Institute, New Lambton, NSW, Australia
- Newcastle Centre of Excellence in Cardio-Oncology, Hunter Medical Research Institute, Hunter New England Local Health District, University of Newcastle and Calvary Mater Newcastle, Newcastle, NSW, Australia
- School of Medicine and Public Health, University of Newcastle, Callaghan, NSW, 2305, Australia
| | - Lohis Balachandran
- Heart and Stroke Research Program, Hunter Medical Research Institute, New Lambton, NSW, Australia
- Newcastle Centre of Excellence in Cardio-Oncology, Hunter Medical Research Institute, Hunter New England Local Health District, University of Newcastle and Calvary Mater Newcastle, Newcastle, NSW, Australia
- School of Medicine and Public Health, University of Newcastle, Callaghan, NSW, 2305, Australia
| | - Dongqing Chen
- School of Biomedical Sciences and Pharmacy, The University of Newcastle, Callaghan, NSW, 2305, Australia
- Heart and Stroke Research Program, Hunter Medical Research Institute, New Lambton, NSW, Australia
- Newcastle Centre of Excellence in Cardio-Oncology, Hunter Medical Research Institute, Hunter New England Local Health District, University of Newcastle and Calvary Mater Newcastle, Newcastle, NSW, Australia
| | - Danielle R Bond
- School of Biomedical Sciences and Pharmacy, The University of Newcastle, Callaghan, NSW, 2305, Australia
- Cancer Research Program, Hunter Medical Research Institute, New Lambton, NSW, Australia
| | - Heather J Lee
- School of Biomedical Sciences and Pharmacy, The University of Newcastle, Callaghan, NSW, 2305, Australia
- Cancer Research Program, Hunter Medical Research Institute, New Lambton, NSW, Australia
| | - Murray J Cairns
- School of Biomedical Sciences and Pharmacy, The University of Newcastle, Callaghan, NSW, 2305, Australia
- Precision Medicine Research Program, Hunter Medical Research Institute, New Lambton, NSW, Australia
| | - Aaron L Sverdlov
- Heart and Stroke Research Program, Hunter Medical Research Institute, New Lambton, NSW, Australia.
- Newcastle Centre of Excellence in Cardio-Oncology, Hunter Medical Research Institute, Hunter New England Local Health District, University of Newcastle and Calvary Mater Newcastle, Newcastle, NSW, Australia.
- School of Medicine and Public Health, University of Newcastle, Callaghan, NSW, 2305, Australia.
- Cardiovascular Department, John Hunter Hospital, New Lambton Heights, NSW, Australia.
| | - Doan T M Ngo
- School of Biomedical Sciences and Pharmacy, The University of Newcastle, Callaghan, NSW, 2305, Australia.
- Heart and Stroke Research Program, Hunter Medical Research Institute, New Lambton, NSW, Australia.
- Newcastle Centre of Excellence in Cardio-Oncology, Hunter Medical Research Institute, Hunter New England Local Health District, University of Newcastle and Calvary Mater Newcastle, Newcastle, NSW, Australia.
| |
Collapse
|
42
|
Hurwitz LM, Bailey-Whyte M, Daneshvar MA, Vocke CD, Custer J, Ryan BM, Ambs S, Pinto PA, Rossi EL. Methylation-based immune deconvolution in prostate cancer patients before and after radical prostatectomy. Cancer Causes Control 2025; 36:101-106. [PMID: 39390262 DOI: 10.1007/s10552-024-01924-x] [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: 08/28/2024] [Accepted: 09/27/2024] [Indexed: 10/12/2024]
Abstract
PURPOSE Surgery, an established short-term immunosuppressive event, may spur dissemination of circulating tumor cells and promote the growth of micrometastases. Whether surgical treatment for prostate cancer (i.e., radical prostatectomy) leads to long-term immune changes is unknown. METHODS We characterized intra-individual changes in circulating immune cell subsets across a six-month period using serial blood samples from prostate cancer patients pre- and post-radical prostatectomy (n = 11), and from a comparison group managed with active surveillance (n = 8). Immune cell subsets for each patient at each time point were deconvoluted using genome-wide methylation data. RESULTS There were no statistically significant intra-individual changes in immune cell proportions from pre- to six months post-radical prostatectomy. There were also no intra-individual changes in immune cell proportions in the active surveillance group, and no differences between treatment groups in immune cell changes over time. CONCLUSION We observed no meaningful changes in circulating immune cell subsets six months after radical prostatectomy, suggesting that surgery-induced immune changes may not be long-lasting.
Collapse
Affiliation(s)
- Lauren M Hurwitz
- Occupational and Environmental Epidemiology Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Rockville, MD, USA
| | - Maeve Bailey-Whyte
- Laboratory of Human Carcinogenesis, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA.
- School of Medicine, University of Limerick, Limerick, V94XD21, Ireland.
| | - Michael A Daneshvar
- Urologic Oncology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Cathy D Vocke
- Urologic Oncology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Julian Custer
- Urologic Oncology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Bríd M Ryan
- Laboratory of Human Carcinogenesis, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Stefan Ambs
- Laboratory of Human Carcinogenesis, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Peter A Pinto
- Urologic Oncology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Emily L Rossi
- Laboratory of Human Carcinogenesis, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
43
|
Zhao M, Cai M, Lei F, Yuan X, Liu Q, Fang Y, Zhu B. AI-driven feature selection and epigenetic pattern analysis: A screening strategy of CpGs validated by pyrosequencing for body fluid identification. Forensic Sci Int 2025; 367:112339. [PMID: 39729807 DOI: 10.1016/j.forsciint.2024.112339] [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: 08/29/2024] [Revised: 12/01/2024] [Accepted: 12/06/2024] [Indexed: 12/29/2024]
Abstract
Identification of body fluid stain at crime scene is one of the important tasks of forensic evidence analysis. Currently, body fluid-specific CpGs detected by DNA methylation microarray screening, have been widely studied for forensic body fluid identification. However, some CpGs have limited ability to distinguish certain body fluid types. The ongoing need is to discover novel methylation markers and fully validate them to enhance their evidentiary strength in complex forensic scenarios. This research gathered forensic-related DNA methylation microarrays data from the Gene Expression Omnibus (GEO) database. A novel screening strategy for marker selection was developed, combining feature selection algorithms (elastic net, information gain ratio, feature importance based on Random Forest, and mutual information coefficient) with epigenetic pattern analysis, to identify CpG markers for body fluid identification. The selected CpGs were validated through pyrosequencing on peripheral blood, saliva, semen, vaginal secretions, and menstrual blood samples, and machine learning classification models were constructed based on the sequencing results. Pyrosequencing results revealed 14 CpGs with high specificity in five types of body fluid samples. A machine learning classification model, developed based on the pyrosequencing results, could effectively distinguish five types of body fluid samples, achieving 100 % accuracy on the test set. Utilizing six CpG markers, it was also feasible to attain ideal efficacy in identifying body fluid stains. Our research proposes a systematic and scientific strategy for screening body fluid-specific CpGs, contributing new insights and methods to forensic body fluid identification.
Collapse
Affiliation(s)
- Ming Zhao
- Guangzhou Key Laboratory of Forensic Multi-Omics for Precision Identification, School of Forensic Medicine, Southern Medical University, Guangzhou 510515, China
| | - Meiming Cai
- Guangzhou Key Laboratory of Forensic Multi-Omics for Precision Identification, School of Forensic Medicine, Southern Medical University, Guangzhou 510515, China
| | - Fanzhang Lei
- Guangzhou Key Laboratory of Forensic Multi-Omics for Precision Identification, School of Forensic Medicine, Southern Medical University, Guangzhou 510515, China
| | - Xi Yuan
- Guangzhou Key Laboratory of Forensic Multi-Omics for Precision Identification, School of Forensic Medicine, Southern Medical University, Guangzhou 510515, China
| | - Qinglin Liu
- Guangzhou Key Laboratory of Forensic Multi-Omics for Precision Identification, School of Forensic Medicine, Southern Medical University, Guangzhou 510515, China
| | - Yating Fang
- School of Basic Medical Science, Anhui Medical University, Hefei 230031, China.
| | - Bofeng Zhu
- Guangzhou Key Laboratory of Forensic Multi-Omics for Precision Identification, School of Forensic Medicine, Southern Medical University, Guangzhou 510515, China.
| |
Collapse
|
44
|
Carver K, Clark C, Zhong Y, Yang G, Mishra M, Alarcon C, Perera M. MeQTL Mapping in African American Hepatocytes Reveals Shared Genetic Regulators of DNA Methylation and Gene Expression. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.23.634506. [PMID: 39896509 PMCID: PMC11785176 DOI: 10.1101/2025.01.23.634506] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/04/2025]
Abstract
Methylation quantitative trait loci (meQTL) mapping can provide insight into the genetic architecture underlying the epigenome by identifying single-nucleotide polymorphisms (SNPs) associated with differential methylation at methylation sites (CpGs) across the genome. Given that the epigenetic architecture underlying differences in gene expression can vary across racial populations, performing epigenomic studies in African Americans is crucial for understanding the interplay between genetic variation, DNA methylation, and gene expression in this understudied group. By performing cis-meQTL mapping in African American hepatocytes, we identified 410,186 cis-meQTLs associated with methylation at 24,425 CpGs in the liver. Through colocalization analysis, we found that 18,206 of these meQTLs are also colocalized with known liver eQTLs. Additionally, we found that using African American eQTL data results in an increased ability to detect additional colocalized variants that exhibit strong differences in allele frequency between people of European and African ancestry. Furthermore, the presence of smaller linkage disequilibrium blocks in African Americans allows us to identify narrower genomic regions of potentially causal variants compared to when data from Europeans is used. Importantly, these colocalized SNPs are significantly enriched for genetic associations with lipid and inflammatory traits in the GWAS catalog, suggesting that DNA methylation may contribute to the etiologies of these diseases. Furthermore, while it is generally presumed that the genetic regulation of DNA methylation is shared between blood and liver, we found that only 5.4% of African American liver meQTLs colocalize with blood meQTLs. Overall, our results reveal that studying African American populations results in the identification of additional genetic and epigenetic factors that may regulate gene expression in the liver, thereby expanding our understanding of gene regulation in African Americans.
Collapse
Affiliation(s)
- Kathryn Carver
- Department of Pharmacology, Center for Pharmacogenetics, Feinberg School of Medicine, Northwestern University, Chicago, IL, 60611
| | - Carolina Clark
- Department of Pharmacology, Center for Pharmacogenetics, Feinberg School of Medicine, Northwestern University, Chicago, IL, 60611
| | - Yizhen Zhong
- Department of Pharmacology, Center for Pharmacogenetics, Feinberg School of Medicine, Northwestern University, Chicago, IL, 60611
| | - Guang Yang
- Department of Pharmacology, Center for Pharmacogenetics, Feinberg School of Medicine, Northwestern University, Chicago, IL, 60611
- Center for Applied Bioinformatics, St. Jude Children's Research Hospital, Memphis, TN
| | - Mrinal Mishra
- Department of Pharmacology, Center for Pharmacogenetics, Feinberg School of Medicine, Northwestern University, Chicago, IL, 60611
| | - Cristina Alarcon
- Department of Pharmacology, Center for Pharmacogenetics, Feinberg School of Medicine, Northwestern University, Chicago, IL, 60611
| | - Minoli Perera
- Department of Pharmacology, Center for Pharmacogenetics, Feinberg School of Medicine, Northwestern University, Chicago, IL, 60611
| |
Collapse
|
45
|
Mareckova K, Mendes-Silva AP, Jáni M, Pacinkova A, Piler P, Gonçalves VF, Nikolova YS. Mitochondrial DNA variants and their impact on epigenetic and biological aging in young adulthood. Transl Psychiatry 2025; 15:16. [PMID: 39837837 PMCID: PMC11751369 DOI: 10.1038/s41398-025-03235-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Revised: 12/16/2024] [Accepted: 01/14/2025] [Indexed: 01/23/2025] Open
Abstract
The pace of biological aging varies between people independently of chronological age and mitochondria dysfunction is a key hallmark of biological aging. We hypothesized that higher functional impact (FI) score of mitochondrial DNA (mtDNA) variants might contribute to premature aging and tested the relationships between a novel FI score of mtDNA variants and epigenetic and biological aging in young adulthood. A total of 81 participants from the European Longitudinal Study of Pregnancy and Childhood (ELSPAC) prenatal birth cohort had good quality genetic data as well as blood-based markers to estimate biological aging in the late 20. A subset of these participants (n = 69) also had epigenetic data to estimate epigenetic aging in the early 20s using Horvath's epigenetic clock. The novel FI score was calculated based on 7 potentially pathogenic mtDNA variants. Greater FI score of mtDNA variants was associated with older epigenetic age in the early 20s and older biological age in the late 20s. These medium to large effects were independent of sex, current BMI, cigarette smoking, cannabis, and alcohol use. These findings suggest that elevated FI score of mtDNA variants might contribute to premature aging in young adulthood.
Collapse
Affiliation(s)
- Klara Mareckova
- Brain and Mind Research, Central European Institute of Technology, Masaryk University (CEITEC), Brno, Czech Republic.
- 1st Department of Neurology, St Anne's University Hospital and Faculty of Medicine, Masaryk University, Brno, Czech Republic.
| | - Ana Paula Mendes-Silva
- Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, ON, Canada
- Tanenbaum Centre for Pharmacogenetics, Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, ON, Canada
- Department of Psychiatry, University of Saskatchewan, Saskatoon, SK, Canada
| | - Martin Jáni
- Brain and Mind Research, Central European Institute of Technology, Masaryk University (CEITEC), Brno, Czech Republic
| | - Anna Pacinkova
- Brain and Mind Research, Central European Institute of Technology, Masaryk University (CEITEC), Brno, Czech Republic
- Faculty of Informatics, Masaryk University, Brno, Czechia
| | - Pavel Piler
- RECETOX Faculty of Science, Masaryk Univeristy, Brno, Czech Republic
| | - Vanessa F Gonçalves
- Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, ON, Canada
- Tanenbaum Centre for Pharmacogenetics, Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, ON, Canada
- Department of Psychiatry, University of Toronto, Toronto, ON, Canada
| | - Yuliya S Nikolova
- Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, ON, Canada.
- Department of Psychiatry, University of Toronto, Toronto, ON, Canada.
| |
Collapse
|
46
|
Cheishvili D, Do Carmo S, Caraci F, Grasso M, Cuello AC, Szyf M. EpiAge: a next-generation sequencing-based ELOVL2 epigenetic clock for biological age assessment in saliva and blood across health and disease. Aging (Albany NY) 2025; 17:131-160. [PMID: 39853302 PMCID: PMC11810066 DOI: 10.18632/aging.206188] [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: 05/08/2024] [Accepted: 01/06/2025] [Indexed: 01/26/2025]
Abstract
This study introduces EpiAgePublic, a new method to estimate biological age using only three specific sites on the gene ELOVL2, known for its connection to aging. Unlike traditional methods that require complex and extensive data, our model uses a simpler approach that is well-suited for next-generation sequencing technology, which is a more advanced method of analyzing DNA methylation. This new model overcomes some of the common challenges found in older methods, such as errors due to sample quality and processing variations. We tested EpiAgePublic with a large and varied group of over 4,600 people to ensure its accuracy. It performed on par with, and sometimes better than, more complicated models that use much more data for age estimation. We examined its effectiveness in understanding how factors like HIV infection and stress affect aging, confirming its usefulness in real-world clinical settings. Our results prove that our simple yet effective model, EpiAgePublic, can capture the subtle signs of aging with high accuracy. We also used this model in a study involving patients with Alzheimer's Disease, demonstrating the practical benefits of next-generation sequencing in making precise age-related assessments. This study lays the groundwork for future research on aging mechanisms and assessing how different interventions might impact the aging process using this clock.
Collapse
Affiliation(s)
- David Cheishvili
- EpiMedTech Global, Singapore 409051, Singapore
- HKG Epitherapeutics Ltd., Hong Kong SAR, China
- Gerald Bronfman Department of Oncology, McGill University, Montreal H4A 3T2, Canada
| | - Sonia Do Carmo
- Department of Pharmacology & Therapeutics, McGill University, Montreal H3G 1Y6, Canada
| | - Filippo Caraci
- Department of Drug and Health Sciences, University of Catania, Catania 95125, Italy
- Neuropharmacology and Translational Neurosciences Research Unit, Oasi Research Institute-IRCCS, Troina 94018, Italy
| | - Margherita Grasso
- Neuropharmacology and Translational Neurosciences Research Unit, Oasi Research Institute-IRCCS, Troina 94018, Italy
| | - A Claudio Cuello
- Department of Pharmacology & Therapeutics, McGill University, Montreal H3G 1Y6, Canada
- Visiting Professor, Department of Pharmacology, Oxford University, Oxford OX13QT, UK
| | - Moshe Szyf
- EpiMedTech Global, Singapore 409051, Singapore
- HKG Epitherapeutics Ltd., Hong Kong SAR, China
| |
Collapse
|
47
|
Tang X, Guo R, Mo Z, Fu W, Qian X. Causality-driven candidate identification for reliable DNA methylation biomarker discovery. Nat Commun 2025; 16:680. [PMID: 39814752 PMCID: PMC11735613 DOI: 10.1038/s41467-025-56054-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: 06/28/2024] [Accepted: 01/06/2025] [Indexed: 01/18/2025] Open
Abstract
Despite vast data support in DNA methylation (DNAm) biomarker discovery to facilitate health-care research, this field faces huge resource barriers due to preliminary unreliable candidates and the consequent compensations using expensive experiments. The underlying challenges lie in the confounding factors, especially measurement noise and individual characteristics. To achieve reliable identification of a candidate pool for DNAm biomarker discovery, we propose a Causality-driven Deep Regularization framework to reinforce correlations that are suggestive of causality with disease. It integrates causal thinking, deep learning, and biological priors to handle non-causal confounding factors, through a contrastive scheme and a spatial-relation regularization that reduces interferences from individual characteristics and noises, respectively. The comprehensive reliability of the proposed method was verified by simulations and applications involving various human diseases, sample origins, and sequencing technologies, highlighting its universal biomedical significance. Overall, this study offers a causal-deep-learning-based perspective with a compatible tool to identify reliable DNAm biomarker candidates, promoting resource-efficient biomarker discovery.
Collapse
Affiliation(s)
- Xinlu Tang
- The Medical Image and Health Informatics Lab, the School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China
| | - Rui Guo
- The Medical Image and Health Informatics Lab, the School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China
| | - Zhanfeng Mo
- College of Computing and Data Science, Nanyang Technological University, Singapore, Singapore
| | - Wenli Fu
- The Medical Image and Health Informatics Lab, the School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China
| | - Xiaohua Qian
- The Medical Image and Health Informatics Lab, the School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China.
| |
Collapse
|
48
|
Laha S, Das S, Banerjee U, Ganguly T, Senapati S, Chatterjee G, Chatterjee R. Genome-wide RNA-seq, DNA methylation and small RNA-seq analysis unraveled complex gene regulatory networks in psoriasis pathogenesis. Gene 2025; 933:148903. [PMID: 39233195 DOI: 10.1016/j.gene.2024.148903] [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: 01/24/2024] [Revised: 08/12/2024] [Accepted: 08/28/2024] [Indexed: 09/06/2024]
Abstract
Psoriasis is a complex inflammatory skin disease characterized by reversible albeit relapsing red scaly plaques in the skin of a patient. In addition to the genetic predisposition, involvement of epigenetic and non-coding RNAs have also been liked with the disease. Nevertheless, any comprehensive study involving transcriptomic, small-RNA and DNA methylation at the genomic level from same patients is lacking. To investigate the complex regulation of molecular pathways in psoriasis, we carried out multi-omics integrative analysis of RNA-sequencing, small RNA-sequencing and DNA methylation profiling from the psoriatic and adjacent normal skin tissues. Our multi-omics analysis identified the genes and biological processes regulated either independently or in combination by DNA methylation and microRNAs. We identified miRNAs that specifically regulated keratinocyte hyper-proliferation, and cell cycle progression and checkpoint signaling in psoriasis. On contrary, DNA methylation was found to be more predominant in regulating immune and inflammatory responses, another causative factor in psoriasis pathogenesis. Many characteristic pathways in psoriasis e.g., Th17 cell differentiation and JAK-STAT signaling, were found to be regulated by both miRNAs and DNA methylation. We carried out functional characterization of a downregulated miRNA hsa-let-7c-5p, predicted to target upregulated genes in psoriasis involved in cell cycle processes, Th17 cell differentiation and JAK-STAT signaling pathways. Overexpression of hsa-let-7c-5p in keratinocytes caused the downregulation of its target genes, resulting in reduced cell proliferation and migration rates, demonstrating potential of miRNAs in regulating psoriasis pathogenesis. In conclusion, our findings identified distinct and shared gene-networks regulated by DNA methylation and miRNAs of a complex disease with reversible phenotype.
Collapse
Affiliation(s)
- Sayantan Laha
- Human Genetics Unit, Indian Statistical Institute, 203 B. T. Road, Kolkata, West Bengal 700108, India
| | - Shantanab Das
- Human Genetics Unit, Indian Statistical Institute, 203 B. T. Road, Kolkata, West Bengal 700108, India
| | - Urbee Banerjee
- Human Genetics Unit, Indian Statistical Institute, 203 B. T. Road, Kolkata, West Bengal 700108, India
| | - Torsa Ganguly
- Human Genetics Unit, Indian Statistical Institute, 203 B. T. Road, Kolkata, West Bengal 700108, India
| | - Swapan Senapati
- Consultant Dermatologist, Uttarpara, Hooghly, West Bengal 712258, India
| | - Gobinda Chatterjee
- Department of Dermatology, IPGMER/SSKM Hospital, Kolkata, West Bengal, India
| | - Raghunath Chatterjee
- Human Genetics Unit, Indian Statistical Institute, 203 B. T. Road, Kolkata, West Bengal 700108, India.
| |
Collapse
|
49
|
Lim H, El-Serag HB, Luster M, Grove ML, Byun J, Jung Y, Han Y, Boerwinkle E, Amos CI, Thrift AP. DNA Methylation Profile in Buffy Coat Identifies Methylation Differences Between Cirrhosis with and Without Hepatocellular Carcinoma. Cancers (Basel) 2025; 17:266. [PMID: 39858049 PMCID: PMC11763440 DOI: 10.3390/cancers17020266] [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: 11/28/2024] [Revised: 01/10/2025] [Accepted: 01/13/2025] [Indexed: 01/27/2025] Open
Abstract
BACKGROUND/OBJECTIVES Cirrhosis is the precursor to most cases of hepatocellular carcinoma (HCC). Understanding the mechanisms leading to the transition from cirrhosis to HCC and identifying key biomarkers is crucial to developing effective screening strategies and reducing HCC-related mortality. DNA methylation is associated with gene inactivation and plays an important role in physiological and pathological processes; however, its role in cirrhosis progression to HCC is unknown. METHODS We performed genome-wide DNA methylation profiling using Illumina Infinium MethylationEPI BeadChip in pre-diagnostic samples from 22 cirrhosis patients who subsequently developed HCC and 22 cirrhosis patients who remained HCC-free during an average 4-year follow-up. In a secondary analysis, we examined a subset of patients without hepatitis C virus (HCV) infection. RESULTS We identified three differentially methylated positions (DMPs) located in ADAM12 (cg13674437) and PSD3 (cg06758847 and cg24595678) that show a strong association with HCC risk (lower median vs. higher median hazards ratio (HR): HR cg13674437 = 0.34, 95% CI = 0.14-0.83; HR cg06758847 = 4.89, 95% CI = 1.79-13.33; HR cg24595678 = 11.19, 95% CI = 3.27-38.35). After excluding all HCV-active patients from our analysis, the HR for the DMPs remained significant. CONCLUSIONS In conclusion, the findings in this study support the theory that buffy coat-derived DNA methylation markers could be used to identify biomarkers among cirrhosis patients at high risk for HCC before clinical symptoms appear. A further study with a large prospective cohort is required to validate these findings.
Collapse
Affiliation(s)
- Hyeyeun Lim
- Section of Epidemiology and Population Sciences, Department of Medicine, Baylor College of Medicine, Houston, TX 77030, USA;
| | - Hashem B. El-Serag
- Section of Gastroenterology and Hepatology, Department of Medicine, Baylor College of Medicine, Houston, TX 77030, USA; (H.B.E.-S.); (M.L.)
| | - Michelle Luster
- Section of Gastroenterology and Hepatology, Department of Medicine, Baylor College of Medicine, Houston, TX 77030, USA; (H.B.E.-S.); (M.L.)
| | - Megan L. Grove
- Human Genetics Center, Department of Epidemiology, School of Public Health, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA; (M.L.G.); (E.B.)
| | - Jinyoung Byun
- Institute for Clinical and Translational Research, Baylor College of Medicine, Houston, TX 77030, USA; (J.B.); (Y.H.)
| | - Yuri Jung
- Ridgewood High School, Ridgewood, NJ 07450, USA;
| | - Younghun Han
- Institute for Clinical and Translational Research, Baylor College of Medicine, Houston, TX 77030, USA; (J.B.); (Y.H.)
| | - Eric Boerwinkle
- Human Genetics Center, Department of Epidemiology, School of Public Health, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA; (M.L.G.); (E.B.)
| | - Christopher I. Amos
- Section of Epidemiology and Population Sciences, Department of Medicine, Baylor College of Medicine, Houston, TX 77030, USA;
- Institute for Clinical and Translational Research, Baylor College of Medicine, Houston, TX 77030, USA; (J.B.); (Y.H.)
- Dan L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX 77054, USA
| | - Aaron P. Thrift
- Section of Epidemiology and Population Sciences, Department of Medicine, Baylor College of Medicine, Houston, TX 77030, USA;
- Dan L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX 77054, USA
| |
Collapse
|
50
|
Bozack A, Khodasevich D, Nwanaji-Enwerem JC, Gladish N, Shen H, Daredia S, Gamble M, Needham BL, Rehkopf DH, Cardenas A. One-carbon metabolism-related compounds are associated with epigenetic aging biomarkers: Results from National Health and Nutrition Examination Survey (NHANES) 1999-2002. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2025:2025.01.06.25320074. [PMID: 39830269 PMCID: PMC11741460 DOI: 10.1101/2025.01.06.25320074] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/22/2025]
Abstract
Background One-carbon metabolism (OCM), a biochemical pathway dependent on micronutrients including folate and vitamin B12, plays an essential role in aging-related physiological processes. DNA methylation-based aging biomarkers may be influenced by OCM. Objective This study investigated associations of OCM-related biomarkers with epigenetic aging biomarkers in the National Health and Nutrition Examination Survey (NHANES). Methods Blood DNA methylation was measured in adults aged ≥50 years in the 1999-2000 and 2001-2002 cycles of NHANES. The following epigenetic aging biomarkers were included: Horvath1, Horvath2, Hannum, PhenoAge, GrimAge2, DunedinPoAm, and DNA methylation telomere length (DNAmTL). We tested for associations of serum folate, red blood cell (RBC) folate, vitamin B12, homocysteine, and methylmalonic acid concentrations with epigenetic age deviation (EAD) among 2,346 participants with epigenetic and nutritional status biomarkers using survey weighted general linear regression models adjusting for sociodemographics, BMI, and behavioral factors. Results A doubling of serum folate concentration was associated with -0.82 years (95% confidence interval (CI) = -1.40, -0.23) lower GrimAge EAD, -0.13 SDs (-0.22, -0.03) lower DunedinPoAm, and 0.02 kb (0.00, 0.04) greater DNAmTL EAD. Associations were attenuated after adjusting for smoking status and alcohol intake, folate antagonists. Conversely, a doubling in homocysteine concentration was associated with 1.05 years (0.06, 2.04) greater PhenoAge EAD, 1.93 years (1.16, 2.71) greater GrimAge2 EAD, and 0.26 SDs (0.10, 0.41) greater DunedinPoAm. Associations with GrimAge2 EAD and DunedinPoAm were robust to alcohol and smoking adjustment. Conclusions In a nationally representative sample of U.S. adults, greater folate, a carbon donor, was associated with lower EAD, and greater homocysteine, an indicator of OCM deficiencies, was associated with greater EAD; however, some associations were influenced by smoking status. Future research should focus on high-risk populations. Randomized controlled trials with long-term follow-up are also needed to established causality and investigate the clinical relevance of changes in EAD.
Collapse
|