1
|
Teschendorff AE, Horvath S. Epigenetic ageing clocks: statistical methods and emerging computational challenges. Nat Rev Genet 2025; 26:350-368. [PMID: 39806006 DOI: 10.1038/s41576-024-00807-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/20/2024] [Indexed: 01/16/2025]
Abstract
Over the past decade, epigenetic clocks have emerged as powerful machine learning tools, not only to estimate chronological and biological age but also to assess the efficacy of anti-ageing, cellular rejuvenation and disease-preventive interventions. However, many computational and statistical challenges remain that limit our understanding, interpretation and application of epigenetic clocks. Here, we review these computational challenges, focusing on interpretation, cell-type heterogeneity and emerging single-cell methods, aiming to provide guidelines for the rigorous construction of interpretable epigenetic clocks at cell-type and single-cell resolution.
Collapse
Affiliation(s)
- 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, Shanghai, China.
| | | |
Collapse
|
2
|
Perez-Correa JF, Stiehl T, Marioni RE, Corley J, Cox SR, Costa IG, Wagner W. Weighted 2D-kernel density estimations provide a new probabilistic measure for epigenetic age. Genome Biol 2025; 26:103. [PMID: 40264182 PMCID: PMC12016065 DOI: 10.1186/s13059-025-03562-1] [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: 06/13/2024] [Accepted: 03/27/2025] [Indexed: 04/24/2025] Open
Abstract
Epigenetic aging signatures provide insights into human aging, but traditional clocks rely on linear regression of DNA methylation levels, assuming linear trajectories. This study explores a non-parametric approach using 2D-kernel density estimation to determine epigenetic age. Our weighted model achieves similar predictive accuracy as conventional clocks and provides a variation score reflecting the inherent variability of age-related epigenetic changes within samples. This score is significantly increased in various diseases and associated with mortality risk in the Lothian Birth Cohort 1921. Thus, weighted 2D-kernel density estimation facilitates accurate epigenetic age predictions and offers an additional variable for biological age estimation.
Collapse
Affiliation(s)
- Juan-Felipe Perez-Correa
- Institute for Stem Cell Biology, RWTH Aachen University Medical School, Aachen, Germany
- Helmholtz Institute for Biomedical Engineering, RWTH Aachen University Medical School, Aachen, Germany
| | - Thomas Stiehl
- Institute for Computational Biomedicine - Disease Modeling, RWTH Aachen University, Aachen, Germany
| | - Riccardo E Marioni
- Centre for Genomic and Experimental Medicine, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, UK
| | - Janie Corley
- Lothian Birth Cohorts, Department of Psychology, University of Edinburgh, Edinburgh, UK
| | - Simon R Cox
- Lothian Birth Cohorts, Department of Psychology, University of Edinburgh, Edinburgh, UK
| | - Ivan G Costa
- Institute for Computational Genomics, RWTH Aachen University Hospital, Aachen, Germany
| | - Wolfgang Wagner
- Institute for Stem Cell Biology, RWTH Aachen University Medical School, Aachen, Germany.
- Helmholtz Institute for Biomedical Engineering, RWTH Aachen University Medical School, Aachen, Germany.
- Center for Integrated Oncology, Aachen Bonn Cologne Düsseldorf (CIO ABCD), Aachen, Germany.
| |
Collapse
|
3
|
Duran I, Tsurumi A. Evaluating transcriptional alterations associated with ageing and developing age prediction models based on the human blood transcriptome. Biogerontology 2025; 26:86. [PMID: 40186010 DOI: 10.1007/s10522-025-10216-z] [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: 02/09/2024] [Accepted: 03/04/2025] [Indexed: 04/07/2025]
Abstract
Ageing-related DNA methylome and proteome changes and machine-learned ageing clock models have been described previously; however, there is a dearth of ageing clock prediction models based on human blood transcript information. Applying various machine learning algorithms is expected to aid in the development of age prediction models. Using blood transcriptome data from healthy subjects ranging in age from 21 to 90 in the 10 K Immunomes repository, we evaluated differentially regulated transcripts, assessed enriched gene ontology, pathway and disease ontology analysis to characterize biological functions associated with the genes associated with age. Furthermore, we constructed and compared age prediction models developed by applying the Least Absolute Shrinkage and Selection Operator (LASSO), Elastic Net (EN), eXtreme Gradient Boosting (XGBoost) and Light Gradient-Boosting Machine (LightGBM) algorithms. Compared to LASSO (7 genes) and EN (9 genes) regularized regression, XGBoost (142 genes) and LightGBM (149 genes) Gradient Boosted Decision Tree methods performed better in this dataset (training set r = 0.836 (LASSO), 0.837 (EN), 1.000 (XGBoost) and 0.995 (LightGBM); test set: r = 0.883 (LASSO), 0.876 (EN), 0.931 (XGBoost) and 0.915 (LightGBM); external validation set: r = 0.535 (LASSO), 0.534 (EN), 0.591 (XGBoost) and 0.645 (LightGBM)). Blood transcriptome-based age prediction models may provide a simple method to monitor biological ageing, and provide additional molecular insight. Future studies to externally validate these models in various diverse large populations and molecular studies to elucidate the underlying mechanisms by which the gene expression levels may be related to ageing phenotypes would be advantageous.
Collapse
Affiliation(s)
- Ivan Duran
- Department of Surgery, Massachusetts General Hospital and Harvard Medical School, 50 Blossom St., Boston, MA, 02114, USA
| | - Amy Tsurumi
- Department of Surgery, Massachusetts General Hospital and Harvard Medical School, 50 Blossom St., Boston, MA, 02114, USA.
- Shriners Hospitals for Children-Boston, 51 Blossom St., Boston, MA, 02114, USA.
| |
Collapse
|
4
|
Herzog CMS, Redl E, Barrett J, Aminzadeh-Gohari S, Weber DD, Tevini J, Lang R, Kofler B, Widschwendter M. Functionally enriched epigenetic clocks reveal tissue-specific discordant aging patterns in individuals with cancer. COMMUNICATIONS MEDICINE 2025; 5:98. [PMID: 40175686 PMCID: PMC11965555 DOI: 10.1038/s43856-025-00739-4] [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: 07/11/2024] [Accepted: 01/08/2025] [Indexed: 04/04/2025] Open
Abstract
BACKGROUND Aging is a key risk factor for many diseases, including cancer, and a better understanding of its underlying molecular mechanisms may help to prevent, delay, or treat age-related pathologies. Epigenetic alterations such as DNA methylation (DNAme) changes are a hallmark of aging and form the basis of so-called epigenetic clocks, yet their functional relevance and directionality in different organs during disease development is often unclear. METHODS Here, we link cell-specific age-related DNAme changes with three key hallmarks of aging and cancer (senescence, promoter methylation in genes associated with stem cell fate, and dysregulated proliferation) to comprehensively dissect their association with current and future cancer development, carcinogen exposure or preventive measures, and mortality using data in different organs from over 12,510 human and 105 mouse samples, benchmarking against existing epigenetic clocks. RESULTS Our findings offer insights into the association of functionally enriched groups of age-related DNAme changes with cancer, identify sites perturbed earliest during carcinogenesis, as well as those distinct between cancer and reprogramming that could inform strategies to prevent teratoma formation upon in vivo reprogramming. Surprisingly, both mouse and human data reveal accelerated aging in breast cancer tissue but decelerated epigenetic aging in some non-cancer surrogate samples from breast cancer patients, in particular cervical samples. CONCLUSIONS This work provides evidence for discordant systemic tissue aging in breast cancer.
Collapse
Affiliation(s)
- Chiara M S Herzog
- European Translational Oncology Prevention and Screening Institute, Universität Innsbruck, Innsbruck, Austria
- Institute for Biomedical Aging Research, Universität Innsbruck, Innsbruck, Austria
| | - Elisa Redl
- European Translational Oncology Prevention and Screening Institute, Universität Innsbruck, Innsbruck, Austria
- Institute for Biomedical Aging Research, Universität Innsbruck, Innsbruck, Austria
| | - James Barrett
- European Translational Oncology Prevention and Screening Institute, Universität Innsbruck, Innsbruck, Austria
- Institute for Biomedical Aging Research, Universität Innsbruck, Innsbruck, Austria
| | - Sepideh Aminzadeh-Gohari
- European Translational Oncology Prevention and Screening Institute, Universität Innsbruck, Innsbruck, Austria
- Institute for Biomedical Aging Research, Universität Innsbruck, Innsbruck, Austria
- Research Program for Receptor Biochemistry and Tumor Metabolism, Department of Pediatrics, University Hospital of the Paracelsus Medical University, Salzburg, Austria
| | - Daniela D Weber
- Research Program for Receptor Biochemistry and Tumor Metabolism, Department of Pediatrics, University Hospital of the Paracelsus Medical University, Salzburg, Austria
| | - Julia Tevini
- Research Program for Receptor Biochemistry and Tumor Metabolism, Department of Pediatrics, University Hospital of the Paracelsus Medical University, Salzburg, Austria
| | - Roland Lang
- Department of Dermatology and Allergology, University Hospital of the Paracelsus Medical University, Salzburg, Austria
| | - Barbara Kofler
- Research Program for Receptor Biochemistry and Tumor Metabolism, Department of Pediatrics, University Hospital of the Paracelsus Medical University, Salzburg, Austria
| | - Martin Widschwendter
- European Translational Oncology Prevention and Screening Institute, Universität Innsbruck, Innsbruck, Austria.
- Institute for Biomedical Aging Research, Universität Innsbruck, Innsbruck, Austria.
- Department of Women's Cancer, UCL EGA Institute for Women's Health, University College London, London, UK.
- Department of Women's and Children's Health, Karolinska Institutet and Karolinska University Hospital, Stockholm, Sweden.
| |
Collapse
|
5
|
Farrell C, Tandon K, Ferrari R, Lapborisuth K, Modi R, Snir S, Pellegrini M. The Multi-State Epigenetic Pacemaker enables the identification of combinations of factors that influence DNA methylation. GeroScience 2025; 47:2439-2454. [PMID: 39549198 PMCID: PMC11979089 DOI: 10.1007/s11357-024-01414-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Accepted: 10/23/2024] [Indexed: 11/18/2024] Open
Abstract
Epigenetic clocks, DNA methylation-based predictive models of chronological age, are often utilized to study aging associated biology. Despite their widespread use, these methods do not account for other factors that also contribute to the variability of DNA methylation data. For example, many CpG sites show strong sex-specific or cell-type-specific patterns that likely impact the predictions of epigenetic age. To overcome these limitations, we developed a multidimensional extension of the Epigenetic Pacemaker, the Multi-state Epigenetic Pacemaker (MSEPM). We show that the MSEPM is capable of accurately modeling multiple methylation-associated factors simultaneously, while also providing site-specific models that describe the per site relationship between methylation and these factors. We utilized the MSEPM with a large aggregate cohort of blood methylation data to construct models of the effects of age-, sex-, and cell-type heterogeneity on DNA methylation. We found that these models capture a large faction of the variability at thousands of DNA methylation sites. Moreover, this approach allows us to identify sites that are primarily affected by aging and no other factors. An analysis of these sites reveals that those that lose methylation over time are enriched for CTCF transcription factor chip peaks, while those that gain methylation over time are associated with bivalent promoters of genes that are not expressed in blood. These observations suggest mechanisms that underlie age-associated methylation changes and suggest that age-associated increases in methylation may not have strong functional consequences on cell states. In conclusion, the MSEPM is capable of accurately modeling multiple methylation-associated factors, and the models produced can illuminate site-specific combinations of factors that affect methylation dynamics.
Collapse
Affiliation(s)
- Colin Farrell
- Dept. of Molecular, Cell and Developmental Biology, University of California, Los Angeles, 90095, CA, USA.
| | - Keshiv Tandon
- Dept. of Molecular, Cell and Developmental Biology, University of California, Los Angeles, 90095, CA, USA
| | - Roberto Ferrari
- Dept. of Chemistry, Life Sciences and Environmental Sustainability, Laboratory of Molecular Cell Biology of the Epigenome (MCBE), University of Parma, Parma, Italy
| | - Kalsuda Lapborisuth
- Dept. of Molecular, Cell and Developmental Biology, University of California, Los Angeles, 90095, CA, USA
| | - Rahil Modi
- Dept. of Molecular, Cell and Developmental Biology, University of California, Los Angeles, 90095, CA, USA
| | - Sagi Snir
- Dept. of Evolutionary Biology, University of Haifa, Haifa, Israel
| | - Matteo Pellegrini
- Dept. of Molecular, Cell and Developmental Biology, University of California, Los Angeles, 90095, CA, USA.
| |
Collapse
|
6
|
Kusters CDJ, Horvath S. Quantification of Epigenetic Aging in Public Health. Annu Rev Public Health 2025; 46:91-110. [PMID: 39681336 DOI: 10.1146/annurev-publhealth-060222-015657] [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] [Indexed: 12/18/2024]
Abstract
Estimators of biological age hold promise for use in preventive medicine, for early detection of chronic conditions, and for monitoring the effectiveness of interventions aimed at improving population health. Among the promising biomarkers in this field are DNA methylation-based biomarkers, commonly referred to as epigenetic clocks. This review provides a survey of these clocks, with an emphasis on second-generation clocks that predict human morbidity and mortality. It explores the validity of epigenetic clocks when considering factors such as race, sex differences, lifestyle, and environmental influences. Furthermore, the review addresses the current challenges and limitations in this research area.
Collapse
Affiliation(s)
- Cynthia D J Kusters
- Department of Epidemiology, Fielding School of Public Health, University of California, Los Angeles, California, USA;
| | - Steve Horvath
- Altos Labs, Cambridge, United Kingdom;
- Department of Biostatistics, Fielding School of Public Health, University of California, Los Angeles, California, USA
| |
Collapse
|
7
|
Kuznetsov NV, Statsenko Y, Ljubisavljevic M. An Update on Neuroaging on Earth and in Spaceflight. Int J Mol Sci 2025; 26:1738. [PMID: 40004201 PMCID: PMC11855577 DOI: 10.3390/ijms26041738] [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: 01/04/2025] [Revised: 02/06/2025] [Accepted: 02/08/2025] [Indexed: 02/27/2025] Open
Abstract
Over 400 articles on the pathophysiology of brain aging, neuroaging, and neurodegeneration were reviewed, with a focus on epigenetic mechanisms and numerous non-coding RNAs. In particular, this review the accent is on microRNAs, the discovery of whose pivotal role in gene regulation was recognized by the 2024 Nobel Prize in Physiology or Medicine. Aging is not a gradual process that can be easily modeled and described. Instead, multiple temporal processes occur during aging, and they can lead to mosaic changes that are not uniform in pace. The rate of change depends on a combination of external and internal factors and can be boosted in accelerated aging. The rate can decrease in decelerated aging due to individual structural and functional reserves created by cognitive, physical training, or pharmacological interventions. Neuroaging can be caused by genetic changes, epigenetic modifications, oxidative stress, inflammation, lifestyle, and environmental factors, which are especially noticeable in space environments where adaptive changes can trigger aging-like processes. Numerous candidate molecular biomarkers specific to neuroaging need to be validated to develop diagnostics and countermeasures.
Collapse
Affiliation(s)
- Nik V. Kuznetsov
- ASPIRE Precision Medicine Research Institute Abu Dhabi, United Arab Emirates University, Al Ain P.O. Box 15551, United Arab Emirates; (Y.S.); (M.L.)
| | - Yauhen Statsenko
- ASPIRE Precision Medicine Research Institute Abu Dhabi, United Arab Emirates University, Al Ain P.O. Box 15551, United Arab Emirates; (Y.S.); (M.L.)
- Department of Radiology, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain P.O. Box 15551, United Arab Emirates
| | - Milos Ljubisavljevic
- ASPIRE Precision Medicine Research Institute Abu Dhabi, United Arab Emirates University, Al Ain P.O. Box 15551, United Arab Emirates; (Y.S.); (M.L.)
- Department of Physiology, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain P.O. Box 15551, United Arab Emirates
| |
Collapse
|
8
|
Li ZP, Du Z, Huang DS, Teschendorff AE. Interpretable deep learning of single-cell and epigenetic data reveals novel molecular insights in aging. Sci Rep 2025; 15:5048. [PMID: 39934290 PMCID: PMC11814351 DOI: 10.1038/s41598-025-89646-1] [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: 11/15/2024] [Accepted: 02/06/2025] [Indexed: 02/13/2025] Open
Abstract
Deep learning (DL) and explainable artificial intelligence (XAI) have emerged as powerful machine-learning tools to identify complex predictive data patterns in a spatial or temporal domain. Here, we consider the application of DL and XAI to large omic datasets, in order to study biological aging at the molecular level. We develop an advanced multi-view graph-level representation learning (MGRL) framework that integrates prior biological network information, to build molecular aging clocks at cell-type resolution, which we subsequently interpret using XAI. We apply this framework to one of the largest single-cell transcriptomic datasets encompassing over a million immune cells from 981 donors, revealing a ribosomal gene subnetwork, whose expression correlates with age independently of cell-type. Application of the same DL-XAI framework to DNA methylation data of sorted monocytes reveals an epigenetically deregulated inflammatory response pathway whose activity increases with age. We show that the ribosomal module and inflammatory pathways would not have been discovered had we used more standard machine-learning methods. In summary, the computational deep learning framework presented here illustrates how deep learning when combined with explainable AI tools, can reveal novel biological insights into the complex process of aging.
Collapse
Grants
- 2021ZD0200403 STI 2030-Major Projects
- 2021ZD0200403 STI 2030-Major Projects
- 32170652, 31970632, 62333018, 62372255 National Natural Science Foundation of China
- 32170652, 31970632, 62333018, 62372255 National Natural Science Foundation of China
- 32170652, 31970632, 62333018, 62372255 National Natural Science Foundation of China
- 2023Z219, 2023Z226, 2024Z112 Key Research and Development Program of Ningbo City
- STI 2030—Major Projects
Collapse
Affiliation(s)
- Zhi-Peng Li
- Ningbo Institute of Digital Twin, Eastern Institute of Technology, Ningbo, 315201, Zhejiang, China
- School of life sciences, University of Science and Technology of China, Hefei, 230026, Anhui, China
| | - Zhaozhen Du
- 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
| | - De-Shuang Huang
- Ningbo Institute of Digital Twin, Eastern Institute of Technology, Ningbo, 315201, Zhejiang, 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
|
9
|
Tennant N, Pavuluri A, O'Connor-Giles K, Singh G, Larschan E, Singh R. TimeFlies: an snRNA-seq aging clock for the fruit fly head sheds light on sex-biased aging. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2024.11.25.625273. [PMID: 39896546 PMCID: PMC11785003 DOI: 10.1101/2024.11.25.625273] [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: 02/04/2025]
Abstract
Although multiple high-performing epigenetic aging clocks exist, few are based directly on gene expression. Such transcriptomic aging clocks allow us to extract age-associated genes directly. However, most existing transcriptomic clocks model a subset of genes and are limited in their ability to predict novel biomarkers. With the growing popularity of single-cell sequencing, there is a need for robust single-cell transcriptomic aging clocks. Moreover, clocks have yet to be applied to investigate the elusive phenomenon of sex differences in aging. We introduce TimeFlies, a pan-cell-type scRNA-seq aging clock for the Drosophila melanogaster head. TimeFlies uses deep learning to classify the donor age of cells based on genome-wide gene expression profiles. Using explainability methods, we identified key marker genes contributing to the classification, with lncRNAs showing up as highly enriched among predicted biomarkers. The top biomarker gene across cell types is lncRNA:roX1, a regulator of X chromosome dosage compensation, a pathway previously identified as a top biomarker of aging in the mouse brain. We validated this finding experimentally, showing a decrease in survival probability in the absence of roX1 in vivo. Furthermore, we trained sex-specific TimeFlies clocks and noted significant differences in model predictions and explanations between male and female clocks, suggesting that different pathways drive aging in males and females.
Collapse
Affiliation(s)
- Nikolai Tennant
- Data Science Institute, Brown University, Providence, RI, USA
| | - Ananya Pavuluri
- Center for Computational Molecular Biology, Brown University, Providence, RI, USA
| | - Kate O'Connor-Giles
- Department of Neuroscience, Brown University, Providence, RI, USA
- Carney Institute for Brain Science, Brown University, Providence, RI, USA
| | - Gunjan Singh
- Department of Molecular Biology, Cell Biology, and Biochemistry, Brown University, Providence, RI, USA
- Department of Neuroscience, Brown University, Providence, RI, USA
| | - Erica Larschan
- Center for Computational Molecular Biology, Brown University, Providence, RI, USA
- Department of Molecular Biology, Cell Biology, and Biochemistry, Brown University, Providence, RI, USA
| | - Ritambhara Singh
- Data Science Institute, Brown University, Providence, RI, USA
- Center for Computational Molecular Biology, Brown University, Providence, RI, USA
- Department of Computer Science, Brown University, Providence, RI, USA
| |
Collapse
|
10
|
Xu K, Hernández B, Arpawong TE, Camuzeaux S, Chekmeneva E, Crimmins EM, Elliott P, Fiorito G, Jiménez B, Kenny RA, McCrory C, McLoughlin S, Pinto R, Sands C, Vineis P, Lau CHE, Robinson O. Assessing Metabolic Ageing via DNA Methylation Surrogate Markers: A Multicohort Study in Britain, Ireland and the USA. Aging Cell 2025:e14484. [PMID: 39829316 DOI: 10.1111/acel.14484] [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: 05/28/2024] [Revised: 12/20/2024] [Accepted: 12/26/2024] [Indexed: 01/22/2025] Open
Abstract
Metabolomics and epigenomics have been used to develop 'ageing clocks' that assess biological age and identify 'accelerated ageing'. While metabolites are subject to short-term variation, DNA methylation (DNAm) may capture longer-term metabolic changes. We aimed to develop a hybrid DNAm-metabolic clock using DNAm as metabolite surrogates ('DNAm-metabolites') for age prediction. Within the UK Airwave cohort (n = 820), we developed DNAm metabolites by regressing 594 metabolites on DNAm and selected 177 DNAm metabolites and 193 metabolites to construct 'DNAm-metabolic' and 'metabolic' clocks. We evaluated clocks in their age prediction and association with noncommunicable disease risk factors. We additionally validated the DNAm-metabolic clock for the prediction of age and health outcomes in The Irish Longitudinal Study of Ageing (TILDA, n = 488) and the Health and Retirement Study (HRS, n = 4018). Around 70% of DNAm metabolites showed significant metabolite correlations (Pearson's r: > 0.30, p < 10-4) in the Airwave test set and overall stronger age associations than metabolites. The DNAm-metabolic clock was enriched for metabolic traits and was associated (p < 0.05) with male sex, heavy drinking, anxiety, depression and trauma. In TILDA and HRS, the DNAm-metabolic clock predicted age (r = 0.73 and 0.69), disability and gait speed (p < 0.05). In HRS, it additionally predicted time to death, diabetes, cardiovascular disease, frailty and grip strength. DNAm metabolite surrogates may facilitate metabolic studies using only DNAm data. Clocks built from DNAm metabolites provided a novel approach to assess metabolic ageing, potentially enabling early detection of metabolic-related diseases for personalised medicine.
Collapse
Affiliation(s)
- Kexin Xu
- MRC Centre for Environment and Health, Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, London, UK
- MRC WIMM Centre of Computational Biology, Radcliffe Department of Medicine, Medical Sciences Division, University of Oxford, Oxford, UK
| | - Belinda Hernández
- The Irish Longitudinal Study on Ageing (TILDA), Department of Medical Gerontology, School of Medicine, Trinity College Dublin, Dublin, Ireland
| | - Thalida Em Arpawong
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, California, USA
| | - Stephane Camuzeaux
- National Phenome Centre and Imperial Clinical Phenotyping Centre, Section of Bioanalytical Chemistry, Department of Metabolism, Digestion and Reproduction, IRDB Building, Imperial College London, London, UK
| | - Elena Chekmeneva
- National Phenome Centre and Imperial Clinical Phenotyping Centre, Section of Bioanalytical Chemistry, Department of Metabolism, Digestion and Reproduction, IRDB Building, Imperial College London, London, UK
| | - Eileen M Crimmins
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, California, USA
| | - Paul Elliott
- MRC Centre for Environment and Health, Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, London, UK
- NIHR Health Protection Research Unit in Chemical and Radiation Threats and Hazards, London, UK
- UK Dementia Research Institute at Imperial College London, London, UK
| | - Giovani Fiorito
- The Irish Longitudinal Study on Ageing (TILDA), Department of Medical Gerontology, School of Medicine, Trinity College Dublin, Dublin, Ireland
- Clinical Bioinformatics Unit, IRCCS Istituto Giannina Gaslini, Genoa, Italy
| | - Beatriz Jiménez
- National Phenome Centre and Imperial Clinical Phenotyping Centre, Section of Bioanalytical Chemistry, Department of Metabolism, Digestion and Reproduction, IRDB Building, Imperial College London, London, UK
| | - Rose Anne Kenny
- The Irish Longitudinal Study on Ageing (TILDA), Department of Medical Gerontology, School of Medicine, Trinity College Dublin, Dublin, Ireland
| | - Cathal McCrory
- The Irish Longitudinal Study on Ageing (TILDA), Department of Medical Gerontology, School of Medicine, Trinity College Dublin, Dublin, Ireland
| | - Sinead McLoughlin
- The Irish Longitudinal Study on Ageing (TILDA), Department of Medical Gerontology, School of Medicine, Trinity College Dublin, Dublin, Ireland
| | - Rui Pinto
- MRC Centre for Environment and Health, Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, London, UK
- National Phenome Centre and Imperial Clinical Phenotyping Centre, Section of Bioanalytical Chemistry, Department of Metabolism, Digestion and Reproduction, IRDB Building, Imperial College London, London, UK
- UK Dementia Research Institute at Imperial College London, London, UK
| | - Caroline Sands
- National Phenome Centre and Imperial Clinical Phenotyping Centre, Section of Bioanalytical Chemistry, Department of Metabolism, Digestion and Reproduction, IRDB Building, Imperial College London, London, UK
| | - Paolo Vineis
- MRC Centre for Environment and Health, Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, London, UK
| | - Chung-Ho E Lau
- MRC Centre for Environment and Health, Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, London, UK
| | - Oliver Robinson
- MRC Centre for Environment and Health, Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, London, UK
- Ageing Epidemiology (AGE) Research Unit, School of Public Health, Imperial College London, London, UK
| |
Collapse
|
11
|
Wilczok D. Deep learning and generative artificial intelligence in aging research and healthy longevity medicine. Aging (Albany NY) 2025; 17:251-275. [PMID: 39836094 PMCID: PMC11810058 DOI: 10.18632/aging.206190] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2024] [Accepted: 01/08/2025] [Indexed: 01/22/2025]
Abstract
With the global population aging at an unprecedented rate, there is a need to extend healthy productive life span. This review examines how Deep Learning (DL) and Generative Artificial Intelligence (GenAI) are used in biomarker discovery, deep aging clock development, geroprotector identification and generation of dual-purpose therapeutics targeting aging and disease. The paper explores the emergence of multimodal, multitasking research systems highlighting promising future directions for GenAI in human and animal aging research, as well as clinical application in healthy longevity medicine.
Collapse
Affiliation(s)
- Dominika Wilczok
- Duke University, Durham, NC 27708, USA
- Duke Kunshan University, Kunshan, Jiangsu 215316, China
| |
Collapse
|
12
|
de Lima Camillo LP, Asif MH, Horvath S, Larschan E, Singh R. Histone mark age of human tissues and cell types. SCIENCE ADVANCES 2025; 11:eadk9373. [PMID: 39742485 DOI: 10.1126/sciadv.adk9373] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/19/2023] [Accepted: 11/25/2024] [Indexed: 01/03/2025]
Abstract
Aging is a complex and multifaceted process involving many epigenetic alterations. One key area of interest in aging research is the role of histone modifications, which can dynamically regulate gene expression. Here, we conducted a pan-tissue analysis of the dynamics of seven key histone modifications during human aging. Our histone-specific age prediction models showed surprisingly accurate performance, proving resilient to experimental and artificial noise. Simulation experiments for comparison with DNA methylation age predictors revealed competitive performance. Moreover, gene set enrichment analysis uncovered several critical developmental pathways for age prediction. Different from DNA methylation age predictors, genes known to be involved in aging biology are among the most important ones for the models. Last, we developed a pan-tissue pan-histone age predictor, suggesting that age-related epigenetic information is degenerated across the epigenome. This research highlights the power of histone marks as input for creating robust age predictors and opens avenues for understanding the role of epigenetic changes during aging.
Collapse
Affiliation(s)
- Lucas Paulo de Lima Camillo
- School of Biological Sciences, University of Cambridge, Cambridge, UK
- School of Clinical Medicine, University of Cambridge, Cambridge, UK
| | | | | | - Erica Larschan
- Center for Computational Molecular Biology, Brown University, Providence, RI, USA
- Department of Molecular Biology, Cell Biology and Biochemistry, Brown University, Providence, RI, USA
| | - Ritambhara Singh
- Department of Computer Science, Brown University, Providence, RI, USA
- Center for Computational Molecular Biology, Brown University, Providence, RI, USA
| |
Collapse
|
13
|
Richardson M, Brandt C, Jain N, Li JL, Demanelis K, Jasmine F, Kibriya MG, Tong L, Pierce BL. Characterization of DNA methylation clock algorithms applied to diverse tissue types. Aging (Albany NY) 2025; 17:67-96. [PMID: 39754638 PMCID: PMC11810061 DOI: 10.18632/aging.206182] [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: 12/05/2023] [Accepted: 12/12/2024] [Indexed: 01/06/2025]
Abstract
BACKGROUND DNA methylation (DNAm) data from human samples has been leveraged to develop "epigenetic clock" algorithms that predict age and other aging-related phenotypes. Some DNAm clocks were trained using DNAm obtained from blood cells, while other clocks were trained using data from diverse tissue/cell types. To assess how DNAm clocks perform across non-blood tissue types, we applied DNAm algorithms to DNAm data generated from 9 different human tissue types. METHODS We generated array-based DNAm measurements for 973 samples from deceased tissue donors from the GTEx (Genotype Tissue Expression) project representing nine distinct tissue types: lung, colon, prostate, ovary, breast, kidney, testis, skeletal muscle, and whole blood. For all samples, we generated DNAm clock estimates for 8 epigenetic clocks and characterized these tissue-specific clock estimates in terms of their distributions, correlations with chronological age, correlations of clock estimates between tissue types, and association with participant characteristics. RESULTS For each clock, the mean DNAm age estimate varied substantially across tissue types, and the mean values for the different clocks varied substantially within tissue types. For most clocks, the correlation with chronological age varied across tissue types, with blood often showing the strongest correlation. Each clock showed strong correlation across tissues, with some evidence of some residual correlation after adjusting for chronological age. In lung tissue, smoking generally had a positive association with epigenetic age. CONCLUSIONS This work demonstrates how differences in epigenetic aging among tissue types leads to clear differences in DNAm clock characteristics across tissue types. Tissue or cell-type specific epigenetic clocks are needed to optimize predictive performance of DNAm clocks in non-blood tissues and cell types.
Collapse
Affiliation(s)
- Mark Richardson
- Department of Public Health Sciences, University of Chicago, Chicago, IL 60615, USA
| | - Courtney Brandt
- Department of Public Health Sciences, University of Chicago, Chicago, IL 60615, USA
| | - Niyati Jain
- Department of Public Health Sciences, University of Chicago, Chicago, IL 60615, USA
| | - James L. Li
- Department of Public Health Sciences, University of Chicago, Chicago, IL 60615, USA
| | - Kathryn Demanelis
- Department of Medicine, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Farzana Jasmine
- Department of Public Health Sciences, University of Chicago, Chicago, IL 60615, USA
| | - Muhammad G. Kibriya
- Department of Public Health Sciences, University of Chicago, Chicago, IL 60615, USA
| | - Lin Tong
- Department of Public Health Sciences, University of Chicago, Chicago, IL 60615, USA
| | - Brandon L. Pierce
- Department of Public Health Sciences, University of Chicago, Chicago, IL 60615, USA
- Department of Human Genetics, University of Chicago, Chicago, IL 60615, USA
- Comprehensive Cancer Center, University of Chicago, Chicago, IL 60615, USA
| |
Collapse
|
14
|
Levy JJ, Diallo AB, Saldias Montivero MK, Gabbita S, Salas LA, Christensen BC. Insights to aging prediction with AI based epigenetic clocks. Epigenomics 2025; 17:49-57. [PMID: 39584810 DOI: 10.1080/17501911.2024.2432854] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2024] [Accepted: 11/15/2024] [Indexed: 11/26/2024] Open
Abstract
Over the past century, human lifespan has increased remarkably, yet the inevitability of aging persists. The disparity between biological age, which reflects pathological deterioration and disease, and chronological age, indicative of normal aging, has driven prior research focused on identifying mechanisms that could inform interventions to reverse excessive age-related deterioration and reduce morbidity and mortality. DNA methylation has emerged as an important predictor of age, leading to the development of epigenetic clocks that quantify the extent of pathological deterioration beyond what is typically expected for a given age. Machine learning technologies offer promising avenues to enhance our understanding of the biological mechanisms governing aging by further elucidating the gap between biological and chronological ages. This perspective article examines current algorithmic approaches to epigenetic clocks, explores the use of machine learning for age estimation from DNA methylation, and discusses how refining the interpretation of ML methods and tailoring their inferences for specific patient populations and cell types can amplify the utility of these technologies in age prediction. By harnessing insights from machine learning, we are well-positioned to effectively adapt, customize and personalize interventions aimed at aging.
Collapse
Affiliation(s)
- Joshua J Levy
- Department of Pathology and Laboratory Medicine, Cedars Sinai Medical Center, Los Angeles, CA, USA
- Department of Computational Biomedicine, Cedars Sinai Medical Center, Los Angeles, CA, USA
- Emerging Diagnostic and Investigative Technologies, Department of Pathology and Laboratory Medicine, Dartmouth Health, Lebanon, NH, USA
- Department of Dermatology, Dartmouth Health, Lebanon, NH, USA
- Department of Epidemiology, Dartmouth College Geisel School of Medicine, Hanover, NH, USA
| | - Alos B Diallo
- Program in Quantitative Biomedical Sciences, Dartmouth College Geisel School of Medicine, Hanover, NH, USA
| | | | - Sameer Gabbita
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, USA
| | - Lucas A Salas
- Department of Epidemiology, Dartmouth College Geisel School of Medicine, Hanover, NH, USA
- Integrative Neuroscience at Dartmouth, Guarini School of Graduate and Advanced Studies at Dartmouth College, Hanover, NH, USA
| | - Brock C Christensen
- Department of Epidemiology, Dartmouth College Geisel School of Medicine, Hanover, NH, USA
- Molecular and Cellular Biology Program, Guarini School of Graduate and Advanced Studies, Hanover, NH, USA
| |
Collapse
|
15
|
Kiselev IS, Baulina NM, Favorova OO. Epigenetic Clock: DNA Methylation as a Marker of Biological Age and Age-Associated Diseases. BIOCHEMISTRY. BIOKHIMIIA 2025; 90:S356-S372. [PMID: 40164166 DOI: 10.1134/s0006297924602843] [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: 06/25/2024] [Revised: 07/11/2024] [Accepted: 07/20/2024] [Indexed: 04/02/2025]
Abstract
Age is one of the key criteria of human health used in practical medicine to predict the risk of common chronic diseases. However, biological age, which reflects the state of an individual organism, functional capabilities, social well-being, and risk of premature death from various causes, often does not coincide with chronological age. To determine biological age of a particular individuals and the rate of their aging, specific panels of DNA methylation markers called "epigenetic clock" (EC) were proposed. This review summarizes the data about the main types of ECs developed to date and their key characteristics. We described the results of works studying individual aging rates in common age-associated diseases and outlined main directions, development of which could expand application of ECs in fundamental and practical medicine. There is no doubt that revealing complex mechanisms underlying interaction between the rate of epigenetic aging and the risk of age-associated diseases could play a key role for prediction and early diagnosis, as well as for the development of preventive measures that could delay onset of the disease.
Collapse
Affiliation(s)
- Ivan S Kiselev
- Chazov National Medical Research Center of Cardiology, Ministry of Health of the Russian Federation, Moscow, 121552, Russia.
- Pirogov Russian National Research Medical University, Ministry of Health of the Russian Federation, Moscow, 117513, Russia
| | - Natalia M Baulina
- Chazov National Medical Research Center of Cardiology, Ministry of Health of the Russian Federation, Moscow, 121552, Russia
- Pirogov Russian National Research Medical University, Ministry of Health of the Russian Federation, Moscow, 117513, Russia
| | - Olga O Favorova
- Chazov National Medical Research Center of Cardiology, Ministry of Health of the Russian Federation, Moscow, 121552, Russia
- Pirogov Russian National Research Medical University, Ministry of Health of the Russian Federation, Moscow, 117513, Russia
| |
Collapse
|
16
|
Passemiers A, Folco P, Raimondi D, Birolo G, Moreau Y, Fariselli P. A quantitative benchmark of neural network feature selection methods for detecting nonlinear signals. Sci Rep 2024; 14:31180. [PMID: 39732866 DOI: 10.1038/s41598-024-82583-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2024] [Accepted: 12/06/2024] [Indexed: 12/30/2024] Open
Abstract
Classification and regression problems can be challenging when the relevant input features are diluted in noisy datasets, in particular when the sample size is limited. Traditional Feature Selection (FS) methods address this issue by relying on some assumptions such as the linear or additive relationship between features. Recently, a proliferation of Deep Learning (DL) models has emerged to tackle both FS and prediction at the same time, allowing non-linear modeling of the selected features. In this study, we systematically assess the performance of DL-based feature selection methods on synthetic datasets of varying complexity, and benchmark their efficacy in uncovering non-linear relationships between features. We also use the same settings to benchmark the reliability of gradient-based feature attribution techniques for Neural Networks (NNs), such as Saliency Maps (SM). A quantitative evaluation of the reliability of these approaches is currently missing. Our analysis indicates that even simple synthetic datasets can significantly challenge most of the DL-based FS and SM methods, while Random Forests, TreeShap, mRMR and LassoNet are the best performing FS methods. Our conclusion is that when quantifying the relevance of a few non linearly-entangled predictive features diluted in a large number of irrelevant noisy variables, DL-based FS and SM interpretation methods are still far from being reliable.
Collapse
Affiliation(s)
| | - Pietro Folco
- Department of Medical Sciences, University of Torino, Torino, Italy
| | - Daniele Raimondi
- ESAT-STADIUS, KU Leuven, Leuven, Belgium.
- Institut de Génétique Moléculaire de Montpellier, Université de Montpellier, Montpellier, France.
| | - Giovanni Birolo
- Department of Medical Sciences, University of Torino, Torino, Italy
| | | | - Piero Fariselli
- Department of Medical Sciences, University of Torino, Torino, Italy.
| |
Collapse
|
17
|
Liang R, Tang Q, Chen J, Zhu L. Epigenetic Clocks: Beyond Biological Age, Using the Past to Predict the Present and Future. Aging Dis 2024:AD.2024.1495. [PMID: 39751861 DOI: 10.14336/ad.2024.1495] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2024] [Accepted: 12/13/2024] [Indexed: 01/04/2025] Open
Abstract
Predicting health trajectories and accurately measuring aging processes across the human lifespan remain profound scientific challenges. Assessing the effectiveness and impact of interventions targeting aging is even more elusive, largely due to the intricate, multidimensional nature of aging-a process that defies simple quantification. Traditional biomarkers offer only partial perspectives, capturing limited aspects of the aging landscape. Yet, over the past decade, groundbreaking advancements have emerged. Epigenetic clocks, derived from DNA methylation patterns, have established themselves as powerful aging biomarkers, capable of estimating biological age and assessing aging rates across diverse tissues with remarkable precision. These clocks provide predictive insights into mortality and age-related disease risks, effectively distinguishing biological age from chronological age and illuminating enduring questions in gerontology. Despite significant progress in epigenetic clock development, substantial challenges remain, underscoring the need for continued investigation to fully unlock their potential in the science of aging.
Collapse
Affiliation(s)
- Runyu Liang
- Heilongjiang University of Chinese Medicine, Harbin, China
| | - Qiang Tang
- Second Affiliated Hospital of Heilongjiang University of Chinese Medicine, Harbin, China
| | - Jia Chen
- Heilongjiang University of Chinese Medicine, Harbin, China
| | - Luwen Zhu
- Second Affiliated Hospital of Heilongjiang University of Chinese Medicine, Harbin, China
| |
Collapse
|
18
|
Chervova O, Panteleeva K, Chernysheva E, Widayati TA, Baronik ŽF, Hrbková N, Schneider JL, Bobak M, Beck S, Voloshin V. Breaking new ground on human health and well-being with epigenetic clocks: A systematic review and meta-analysis of epigenetic age acceleration associations. Ageing Res Rev 2024; 102:102552. [PMID: 39423872 DOI: 10.1016/j.arr.2024.102552] [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: 07/10/2024] [Revised: 09/13/2024] [Accepted: 10/15/2024] [Indexed: 10/21/2024]
Abstract
Epigenetic clocks provide an accurate molecular readout of epigenetic age and epigenetic age acceleration (EAA) derived from DNA methylation data have shown promise as biomarkers of ageing. This systematic review synthesised research on associations between EAA measures and various physiological, cognitive, social, and environmental factors. A comprehensive search strategy identified 299 publications reporting 1050 unique EAA-factor associations based on 53 methylation clocks. Random-effects meta-analyses pooled results across studies for selected EAA-factor pairs. Significant pooled associations emerged, providing insights into relationships between specific factors and accelerated epigenetic ageing. We developed a novel four-level classification system to categorise this diverse range of factors and enable a structured synthesis. To aid further research planning in this rapidly evolving field, TEAPEE (Tracker of EAA Associations with Phenotype & Environmental Exposure) - an interactive, searchable web table detailing all EAA-factor associations - was developed, cataloguing the epigenetic clocks, associated factors, classification categories, and direct links to the original studies. This resource will empower future investigations into the multifaceted determinants of epigenetic ageing, contributing to a deeper understanding of the epigenome's sensitivity to various life experiences and exposures.
Collapse
Affiliation(s)
- Olga Chervova
- UCL Research Department of Epidemiology & Public Health, University College London, London, United Kingdom; UCL Cancer Institute, University College London, London, United Kingdom.
| | - Kseniia Panteleeva
- University of Cambridge, School of Clinical Medicine, Cambridge, United Kingdom
| | - Elizabeth Chernysheva
- University of Otago, Department of Pathology and Biomedical Science, Christchurch, New Zealand
| | | | | | - Natálie Hrbková
- UCL Cancer Institute, University College London, London, United Kingdom
| | | | - Martin Bobak
- UCL Research Department of Epidemiology & Public Health, University College London, London, United Kingdom
| | - Stephan Beck
- UCL Cancer Institute, University College London, London, United Kingdom
| | - Vitaly Voloshin
- School of Biological and Behavioural Sciences, Queen Mary University of London, London, United Kingdom
| |
Collapse
|
19
|
Pruszkowska-Przybylska P, Noroozi R, Rudnicka J, Pisarek A, Wronka I, Kobus M, Wysocka B, Ossowski A, Spólnicka M, Wiktorska J, Iljin A, Pośpiech E, Branicki W, Sitek A. Potential Predictor of Epigenetic Age Acceleration in Men: 2D:4D Finger Pattern. Am J Hum Biol 2024; 36:e24151. [PMID: 39243113 DOI: 10.1002/ajhb.24151] [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: 05/20/2024] [Revised: 08/23/2024] [Accepted: 08/24/2024] [Indexed: 09/09/2024] Open
Abstract
OBJECTIVES Second to fourth digit ratio is widely known indicator of prenatal sex hormones proportion. Higher prenatal androgenization results in longer fourth finger and lower 2D:4D index. The aim of this study was to determine whether the 2D:4D digit ratio is associated with DNA methylation (DNAm) age dependently on sex. MATERIAL AND METHODS The study included 182 adults (106 females and 76 males) with a mean age of 51.5 ± 13 years. The investigation consisted of three main parts: a survey, anthropometric dimensions measurements (fingers length) and methylome analysis using collected blood samples. Genome-wide methylation was analyzed using EPIC microarray technology. Epigenetic age and epigenetic age acceleration were calculated using several widely applied algorithms. RESULTS Males with the female left hand pattern had more accelerated epigenetic age than those with the male pattern as calculated with PhenoAge and DNAmTL clocks. CONCLUSIONS Finger female pattern 2D:4D above or equal to 1 in males is associated with epigenetic age acceleration, indicating that prenatal exposure to estrogens in males may be related to aging process in the later ontogenesis.
Collapse
Affiliation(s)
| | - Rezvan Noroozi
- Doctoral School of Exact and Natural Sciences, Jagiellonian University, Krakow, Poland
- Małopolska Centre of Biotechnology, Jagiellonian University, Kraków, Poland
| | - Joanna Rudnicka
- Doctoral School of Exact and Natural Sciences, Jagiellonian University, Krakow, Poland
- Małopolska Centre of Biotechnology, Jagiellonian University, Kraków, Poland
| | - Aleksandra Pisarek
- Laboratory of Anthropology, Institute of Zoology and Biomedical Research, Kraków, Poland
| | - Iwona Wronka
- Laboratory of Anthropology, Institute of Zoology and Biomedical Research, Kraków, Poland
| | - Magdalena Kobus
- Institute of Biological Sciences, Faculty of Biology and Environmental Sciences, Cardinal Stefan Wyszynski University in Warsaw, Warsaw, Poland
| | - Bożena Wysocka
- Central Forensic Laboratory of the Police, Warsaw, Poland
| | - Andrzej Ossowski
- Department of Forensic Genetics, Pomeranian Medical University in Szczecin, Szczecin, Poland
| | | | | | - Aleksandra Iljin
- Department of Plastic, Reconstructive and Aesthetic Surgery, Medical University of Lodz 90-153, Lodz, Poland
| | - Ewelina Pośpiech
- Department of Forensic Genetics, Pomeranian Medical University in Szczecin, Szczecin, Poland
| | - Wojciech Branicki
- Laboratory of Anthropology, Institute of Zoology and Biomedical Research, Kraków, Poland
| | - Aneta Sitek
- Department of Anthropology, Faculty of Biology and Environmental Protection, University of Łódź, Łódź, Poland
| |
Collapse
|
20
|
Zakar-Polyák E, Csordas A, Pálovics R, Kerepesi C. Profiling the transcriptomic age of single-cells in humans. Commun Biol 2024; 7:1397. [PMID: 39462118 PMCID: PMC11513945 DOI: 10.1038/s42003-024-07094-5] [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: 01/16/2024] [Accepted: 10/17/2024] [Indexed: 10/28/2024] Open
Abstract
Although aging clocks predicting the age of individual organisms have been extensively studied, the age of individual cells remained largely unexplored. Most recently single-cell omics clocks were developed for the mouse, however, extensive profiling the age of human cells is still lacking. To fill this gap, here we use available scRNA-seq data of 1,058,909 blood cells of 508 healthy, human donors (between 19 and 75 years), for developing single-cell transcriptomic clocks and predicting the age of human blood cells. By the application of the proposed cell-type-specific single-cell clocks, our main observations are that (i) transcriptomic age is associated with cellular senescence; (ii) the transcriptomic age of classical monocytes as well as naive B and T cells is decreased in moderate COVID-19 followed by an increase for some cell types in severe COVID-19; and (iii) the human embryo cells transcriptomically rejuvenated at the morulae and blastocyst stages. In summary, here we demonstrate that single-cell transcriptomic clocks are useful tools to investigate aging and rejuvenation at the single-cell level.
Collapse
Affiliation(s)
- Enikő Zakar-Polyák
- Institute for Computer Science and Control (SZTAKI), Hungarian Research Network (HUN-REN), Budapest, Hungary.
- Doctoral School of Informatics, Eötvös Loránd University, Budapest, Hungary.
| | - Attila Csordas
- AgeCurve Limited, Cambridge, UK
- Doctoral School of Clinical Medicine, University of Szeged, Szeged, Hungary
| | - Róbert Pálovics
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA
- Wu Tsai Neurosciences Institute, Stanford University, Stanford, CA, USA
| | - Csaba Kerepesi
- Institute for Computer Science and Control (SZTAKI), Hungarian Research Network (HUN-REN), Budapest, Hungary.
| |
Collapse
|
21
|
Dabrowski JK, Yang EJ, Crofts SJC, Hillary RF, Simpson DJ, McCartney DL, Marioni RE, Kirschner K, Latorre-Crespo E, Chandra T. Probabilistic inference of epigenetic age acceleration from cellular dynamics. NATURE AGING 2024; 4:1493-1507. [PMID: 39313745 PMCID: PMC11485233 DOI: 10.1038/s43587-024-00700-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/19/2023] [Accepted: 07/31/2024] [Indexed: 09/25/2024]
Abstract
The emergence of epigenetic predictors was a pivotal moment in geroscience, propelling the measurement and concept of biological aging into a quantitative era; however, while current epigenetic clocks show strong predictive power, they are data-driven in nature and are not based on the underlying biological mechanisms driving methylation dynamics. We show that predictions of these clocks are susceptible to several confounding non-age-related phenomena that make interpretation of these estimates and associations difficult. To address these limitations, we developed a probabilistic model describing methylation transitions at the cellular level. Our approach reveals two measurable components, acceleration and bias, which directly reflect perturbations of the underlying cellular dynamics. Acceleration is the proportional increase in the speed of methylation transitions across CpG sites, whereas bias corresponds to global changes in methylation levels. Using data from 15,900 participants from the Generation Scotland study, we develop a robust inference framework and show that these are two distinct processes confounding current epigenetic predictors. Our results show improved associations of acceleration and bias with physiological traits known to impact healthy aging, such as smoking and alcohol consumption, respectively. Furthermore, a genome-wide association study of epigenetic age acceleration identified seven genomic loci.
Collapse
Affiliation(s)
- Jan K Dabrowski
- School of Informatics, University of Edinburgh, Edinburgh, UK
- MRC Human Genetics Unit, University of Edinburgh, Edinburgh, UK
| | - Emma J Yang
- MRC Human Genetics Unit, University of Edinburgh, Edinburgh, UK
| | - Samuel J C Crofts
- MRC Human Genetics Unit, University of Edinburgh, Edinburgh, UK
- Institute of Ecology and Evolution, School of Biological Sciences, University of Edinburgh, Edinburgh, UK
| | - Robert F Hillary
- Centre for Genomic and Experimental Medicine, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, UK
| | | | - Daniel L McCartney
- Centre for Genomic and Experimental Medicine, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, UK
| | - Riccardo E Marioni
- Centre for Genomic and Experimental Medicine, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, UK
| | - Kristina Kirschner
- Cancer Research UK Scotland Institute, Glasgow, UK
- School of Cancer Sciences, University of Glasgow, Glasgow, UK
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, MN, USA
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN, USA
- Department of Hematology, Mayo Clinic, Rochester, MN, USA
| | - Eric Latorre-Crespo
- MRC Human Genetics Unit, University of Edinburgh, Edinburgh, UK.
- School of Cancer Sciences, University of Glasgow, Glasgow, UK.
| | - Tamir Chandra
- MRC Human Genetics Unit, University of Edinburgh, Edinburgh, UK.
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, MN, USA.
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN, USA.
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN, USA.
| |
Collapse
|
22
|
Li Z, Katz S, Saccenti E, Fardo DW, Claes P, Martins dos Santos VAP, Van Steen K, Roshchupkin GV. Novel multi-omics deconfounding variational autoencoders can obtain meaningful disease subtyping. Brief Bioinform 2024; 25:bbae512. [PMID: 39413796 PMCID: PMC11483139 DOI: 10.1093/bib/bbae512] [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: 03/12/2024] [Revised: 09/20/2024] [Accepted: 09/25/2024] [Indexed: 10/18/2024] Open
Abstract
Unsupervised learning, particularly clustering, plays a pivotal role in disease subtyping and patient stratification, especially with the abundance of large-scale multi-omics data. Deep learning models, such as variational autoencoders (VAEs), can enhance clustering algorithms by leveraging inter-individual heterogeneity. However, the impact of confounders-external factors unrelated to the condition, e.g. batch effect or age-on clustering is often overlooked, introducing bias and spurious biological conclusions. In this work, we introduce four novel VAE-based deconfounding frameworks tailored for clustering multi-omics data. These frameworks effectively mitigate confounding effects while preserving genuine biological patterns. The deconfounding strategies employed include (i) removal of latent features correlated with confounders, (ii) a conditional VAE, (iii) adversarial training, and (iv) adding a regularization term to the loss function. Using real-life multi-omics data from The Cancer Genome Atlas, we simulated various confounding effects (linear, nonlinear, categorical, mixed) and assessed model performance across 50 repetitions based on reconstruction error, clustering stability, and deconfounding efficacy. Our results demonstrate that our novel models, particularly the conditional multi-omics VAE (cXVAE), successfully handle simulated confounding effects and recover biologically driven clustering structures. cXVAE accurately identifies patient labels and unveils meaningful pathological associations among cancer types, validating deconfounded representations. Furthermore, our study suggests that some of the proposed strategies, such as adversarial training, prove insufficient in confounder removal. In summary, our study contributes by proposing innovative frameworks for simultaneous multi-omics data integration, dimensionality reduction, and deconfounding in clustering. Benchmarking on open-access data offers guidance to end-users, facilitating meaningful patient stratification for optimized precision medicine.
Collapse
Affiliation(s)
- Zuqi Li
- BIO3 - Laboratory for Systems Medicine, Department of Human Genetics, KU Leuven, Herestraat 49, 3000 Leuven, Belgium
- Medical Imaging Research Center, University Hospitals Leuven, Herestraat 49, 3000 Leuven, Belgium
- BIO3 - Laboratory for Systems Genetics, GIGA Molecular & Computational Biology, University of Liège, Avenue de l'Hôpital 11, 4000 Liège, Belgium
| | - Sonja Katz
- Department of Radiology and Nuclear Medicine, Erasmus MC, Dr. Molewaterplein 40, 3015 GD Rotterdam, Netherlands
- Laboratory of Systems and Synthetic Biology, Wageningen University & Research, PO Box 8033, 6700 EJ Wageningen, Netherlands
- LifeGlimmer GmbH, Markelstraße 38, 12163 Berlin, Germany
| | - Edoardo Saccenti
- Laboratory of Systems and Synthetic Biology, Wageningen University & Research, PO Box 8033, 6700 EJ Wageningen, Netherlands
| | - David W Fardo
- Department of Biostatistics, University of Kentucky, 111 Washington Avenue, Lexington, KY 40536, United States
- Sanders-Brown Center on Aging, University of Kentucky, 789 S Limestone, Lexington, KY 40536, United States
| | - Peter Claes
- Medical Imaging Research Center, University Hospitals Leuven, Herestraat 49, 3000 Leuven, Belgium
- Department of Human Genetics, KU Leuven, Herestraat 49, 3000 Leuven, Belgium
- Department of Electrical Engineering, ESAT-PSI, KU Leuven, Kasteelpark Arenberg 10, 3001 Leuven, Belgium
| | - Vitor A P Martins dos Santos
- LifeGlimmer GmbH, Markelstraße 38, 12163 Berlin, Germany
- Laboratory of Bioprocess Engineering, WageningenUniversity & Research, PO Box 16, 6700 AA Wageningen, the Netherlands
| | - Kristel Van Steen
- BIO3 - Laboratory for Systems Medicine, Department of Human Genetics, KU Leuven, Herestraat 49, 3000 Leuven, Belgium
- BIO3 - Laboratory for Systems Genetics, GIGA Molecular & Computational Biology, University of Liège, Avenue de l'Hôpital 11, 4000 Liège, Belgium
| | - Gennady V Roshchupkin
- Medical Imaging Research Center, University Hospitals Leuven, Herestraat 49, 3000 Leuven, Belgium
- Department of Epidemiology, Erasmus MC, Dr. Molewaterplein 40, 3015 GD Rotterdam, Netherlands
| |
Collapse
|
23
|
Warner B, Ratner E, Datta A, Lendasse A. A systematic review of phenotypic and epigenetic clocks used for aging and mortality quantification in humans. Aging (Albany NY) 2024; 16:12414-12427. [PMID: 39215995 PMCID: PMC11424583 DOI: 10.18632/aging.206098] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2024] [Accepted: 07/15/2024] [Indexed: 09/04/2024]
Abstract
Aging is the leading driver of disease in humans and has profound impacts on mortality. Biological clocks are used to measure the aging process in the hopes of identifying possible interventions. Biological clocks may be categorized as phenotypic or epigenetic, where phenotypic clocks use easily measurable clinical biomarkers and epigenetic clocks use cellular methylation data. In recent years, methylation clocks have attained phenomenal performance when predicting chronological age and have been linked to various age-related diseases. Additionally, phenotypic clocks have been proven to be able to predict mortality better than chronological age, providing intracellular insights into the aging process. This review aimed to systematically survey all proposed epigenetic and phenotypic clocks to date, excluding mitotic clocks (i.e., cancer risk clocks) and those that were modeled using non-human samples. We reported the predictive performance of 33 clocks and outlined the statistical or machine learning techniques used. We also reported the most influential clinical measurements used in the included phenotypic clocks. Our findings provide a systematic reporting of the last decade of biological clock research and indicate possible avenues for future research.
Collapse
Affiliation(s)
| | | | | | - Amaury Lendasse
- Department of IST, University of Houston, Houston, TX 77004, USA
- Department of Engineering Management and Systems Engineering, Missouri University of Science and Technology, Rolla, MO 65409, USA
| |
Collapse
|
24
|
Jain N, Li JL, Tong L, Jasmine F, Kibriya MG, Demanelis K, Oliva M, Chen LS, Pierce BL. DNA methylation correlates of chronological age in diverse human tissue types. Epigenetics Chromatin 2024; 17:25. [PMID: 39118140 PMCID: PMC11308253 DOI: 10.1186/s13072-024-00546-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2024] [Accepted: 06/15/2024] [Indexed: 08/10/2024] Open
Abstract
BACKGROUND While the association of chronological age with DNA methylation (DNAm) in whole blood has been extensively studied, the tissue-specificity of age-related DNAm changes remains an active area of research. Studies investigating the association of age with DNAm in tissues such as brain, skin, immune cells, fat, and liver have identified tissue-specific and non-specific effects, thus, motivating additional studies of diverse human tissue and cell types. RESULTS Here, we performed an epigenome-wide association study, leveraging DNAm data (Illumina EPIC array) from 961 tissue samples representing 9 tissue types (breast, lung, colon, ovary, prostate, skeletal muscle, testis, whole blood, and kidney) from the Genotype-Tissue Expression (GTEx) project. We identified age-associated CpG sites (false discovery rate < 0.05) in 8 tissues (all except skeletal muscle, n = 47). This included 162,002 unique hypermethylated and 90,626 hypomethylated CpG sites across all tissue types, with 130,137 (80%) hypermethylated CpGs and 74,703 (82%) hypomethylated CpG sites observed in a single tissue type. While the majority of age-associated CpG sites appeared tissue-specific, the patterns of enrichment among genomic features, such as chromatin states and CpG islands, were similar across most tissues, suggesting common mechanisms underlying cellular aging. Consistent with previous findings, we observed that hypermethylated CpG sites are enriched in regions with repressed polycomb signatures and CpG islands, while hypomethylated CpG sites preferentially occurred in non-CpG islands and enhancers. To gain insights into the functional effects of age-related DNAm changes, we assessed the correlation between DNAm and local gene expression changes to identify age-related expression quantitative trait methylation (age-eQTMs). We identified several age-eQTMs present in multiple tissue-types, including in the CDKN2A, HENMT1, and VCWE regions. CONCLUSION Overall, our findings will aid future efforts to develop biomarkers of aging and understand mechanisms of aging in diverse human tissue types.
Collapse
Affiliation(s)
- Niyati Jain
- Department of Public Health Sciences, University of Chicago, Chicago, IL, 60637, USA
- Committee on Genetics, Genomics and Systems Biology, University of Chicago, Chicago, IL, 60637, USA
| | - James L Li
- Department of Public Health Sciences, University of Chicago, Chicago, IL, 60637, USA
- Interdisciplinary Scientist Training Program, University of Chicago, Chicago, IL, 60637, USA
| | - Lin Tong
- Department of Public Health Sciences, University of Chicago, Chicago, IL, 60637, USA
| | - Farzana Jasmine
- Institute for Population and Precision Health (IPPH), Biological Sciences Division, University of Chicago, Chicago, IL, 60637, USA
| | - Muhammad G Kibriya
- Department of Public Health Sciences, University of Chicago, Chicago, IL, 60637, USA
| | - Kathryn Demanelis
- Department of Medicine, University of Pittsburgh, Pittsburgh, PA, 15261, USA
- UPMC Hillman Cancer Center, Pittsburgh, PA, 15232, USA
| | - Meritxell Oliva
- Department of Public Health Sciences, University of Chicago, Chicago, IL, 60637, USA
- Genomics Research Center, AbbVie, North Chicago, IL, 60064, USA
| | - Lin S Chen
- Department of Public Health Sciences, University of Chicago, Chicago, IL, 60637, USA
| | - Brandon L Pierce
- Department of Public Health Sciences, University of Chicago, Chicago, IL, 60637, USA.
- Department of Human Genetics, University of Chicago, Chicago, IL, 60637, USA.
- Comprehensive Cancer Center, University of Chicago, Chicago, IL, 60637, USA.
| |
Collapse
|
25
|
Kwiatkowska KM, Anticoli S, Salvioli S, Calzari L, Gentilini D, Albano C, Di Prinzio RR, Zaffina S, Carsetti R, Ruggieri A, Garagnani P. B Cells Isolated from Individuals Who Do Not Respond to the HBV Vaccine Are Characterized by Higher DNA Methylation-Estimated Aging Compared to Responders. Vaccines (Basel) 2024; 12:880. [PMID: 39204006 PMCID: PMC11360008 DOI: 10.3390/vaccines12080880] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2024] [Revised: 07/25/2024] [Accepted: 07/27/2024] [Indexed: 09/03/2024] Open
Abstract
Healthcare workers (HCWs) are a high-risk group for hepatitis B virus (HBV) infection. Notably, about 5-10% of the general population does not respond to the HBV vaccination. In this study, we aimed to investigate DNA methylation (DNAm) in order to estimate the biological age of B cells from HCW of both sexes, either responder (R) or non-responder (NR), to HBV vaccination. We used genome-wide DNA methylation data to calculate a set of biomarkers in B cells collected from 41 Rs and 30 NRs between 22 and 62 years old. Unresponsiveness to HBV vaccination was associated with accelerated epigenetic aging (DNAmAge, AltumAge, DunedinPoAm) and was accompanied by epigenetic drift. Female non-responders had higher estimates of telomere length and lower CRP inflammation risk score when compared to responders. Overall, epigenetic differences between responders and non-responders were more evident in females than males. In this study we demonstrated that several methylation DNAm-based clocks and biomarkers are associated with an increased risk of non-response to HBV vaccination, particularly in females. Based on these results, we propose that accelerated epigenetic age could contribute to vaccine unresponsiveness. These insights may help improve the evaluation of the effectiveness of vaccination strategies, especially among HCWs and vulnerable patients.
Collapse
Affiliation(s)
| | - Simona Anticoli
- Istituto Superiore di Sanità, Center for Gender Specific Medicine, 00161 Rome, Italy; (S.A.); (A.R.)
| | - Stefano Salvioli
- Department of Medical and Surgical Sciences (DIMEC), University of Bologna, 40126 Bologna, Italy;
- IRCCS Azienda Ospedaliero, Universitaria di Bologna, 40138 Bologna, Italy
| | - Luciano Calzari
- Bioinformatics and Statistical Genomics Unit, Istituto Auxologico Italiano IRCCS, 20095 Cusano Milanino, Italy; (L.C.)
| | - Davide Gentilini
- Bioinformatics and Statistical Genomics Unit, Istituto Auxologico Italiano IRCCS, 20095 Cusano Milanino, Italy; (L.C.)
- Department of Brain and Behavioral Sciences, Università di Pavia, 27100 Pavia, Italy
| | - Christian Albano
- Immunology Research Area, B Cell Unit, Ospedale Pediatrico Bambino Gesù IRCCS, 00146 Rome, Italy (R.C.)
| | - Reparata Rosa Di Prinzio
- Occupational Medicine/Health Technology Assessment and Safety Research Unit, Clinical-Technological Innovations Research Area, Ospedale Pediatrico Bambino Gesù IRCCS, 00146 Rome, Italy (S.Z.)
| | - Salvatore Zaffina
- Occupational Medicine/Health Technology Assessment and Safety Research Unit, Clinical-Technological Innovations Research Area, Ospedale Pediatrico Bambino Gesù IRCCS, 00146 Rome, Italy (S.Z.)
| | - Rita Carsetti
- Immunology Research Area, B Cell Unit, Ospedale Pediatrico Bambino Gesù IRCCS, 00146 Rome, Italy (R.C.)
| | - Anna Ruggieri
- Istituto Superiore di Sanità, Center for Gender Specific Medicine, 00161 Rome, Italy; (S.A.); (A.R.)
| | - Paolo Garagnani
- Department of Medical and Surgical Sciences (DIMEC), University of Bologna, 40126 Bologna, Italy;
- IRCCS Azienda Ospedaliero, Universitaria di Bologna, 40138 Bologna, Italy
| |
Collapse
|
26
|
Rosko AE, Elsaid MI, Woyach J, Islam N, Lepola N, Urrutia J, Christian LM, Presley C, Mims A, Burd CE. Determining the relationship of p16 INK4a and additional molecular markers of aging with clinical frailty in hematologic malignancy. J Cancer Surviv 2024; 18:1168-1178. [PMID: 38678524 PMCID: PMC11324703 DOI: 10.1007/s11764-024-01591-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2023] [Accepted: 04/04/2024] [Indexed: 05/01/2024]
Abstract
PURPOSE Older adults with hematologic malignancies (HM) have unique challenges due to age and fitness. The primary aim of this pilot study was to benchmark the ability of multiple biomarkers of aging (p16, epigenetic clocks, T cell gene expression profiles, and T cell receptor excision circles (TREC) to identify frailty as measured by a clinical impairment index (I2) in patients with HM. METHODS 70 patients newly diagnosed with HM had peripheral blood T lymphocytes (PBTL) analyzed for p16INK4a expression using the OSU_Senescence Nanostring CodeSet. PBTL epigenetic age was measured using 7 epigenetic clocks, and TREC were quantified by qRT-PCR. A composite clinical impairment index (I2) was generated by combining values from 11 geriatric metrics (Independent Activities of Daily Living (iADL), physical health score, Short Physical Performance Battery (SPPB), Body Mass Index (BMI), Eastern Cooperative Oncology Group (ECOG) performance status, self-reported KPS, Blessed Orientation Memory Concentration (BOMC), polypharmacy, Mental Health Inventory (MHI)-17, Medical Outcomes Study (MOS) subscales). Clinical frailty was defined as a score of 7 or greater on the I2. RESULTS Age-adjusted p16INK4a was similar in newly diagnosed patients and healthy controls (p > 0.1). PBTL p16INK4a levels correlated positively with the Hannum [r = 0.35, 95% CI (0.09-0.75); p adj. = 0.04] and PhenoAge [r = 0.37, 95% CI (0.11-0.59); p adj. = 0.04] epigenetic clocks. The discrimination ability of the I2 model was calculated using the area under the receiver operating characteristic curve (AUC). After adjusting for chronologic age and disease group, baseline p16INK4a [AUC = 0.76, 95% CI (0.56-0.98); p = 0.01], Hannum [AUC = 0.70, 95% CI (0.54-0.85); p = 0.01], PhenoAge [AUC = 0.71, 95% CI (0.55-0.86); p = 0.01], and DunedinPACE [AUC = 0.73, 95% CI (0.57-0.88); p = < 0.01] measures showed the greatest potential to identify clinical frailty using the I2. CONCLUSIONS Our pilot data suggest that multiple blood-based aging biomarkers have potential to identify frailty in older adults with HM. IMPLICATIONS FOR CANCER SURVIVORS We developed the I2 index to quantify impairments across geriatric domains and discovered that PBTL p16, Hannum, PhenoAge, and DunedinPACE are promising indicators of frailty in HM.
Collapse
Affiliation(s)
- Ashley E Rosko
- Division of Hematology, The Ohio State University, Columbus, OH, USA.
- James Comprehensive Cancer Center, 300 West 10th Ave, Columbus, Ohio, 43210, United States.
| | - Mohamed I Elsaid
- Department of Biomedical Informatics, The Ohio State University, Columbus, OH, USA
- Division of Medical Oncology, The Ohio State University, Columbus, OH, USA
| | - Jennifer Woyach
- Division of Hematology, The Ohio State University, Columbus, OH, USA
| | - Nowshin Islam
- Division of Hematology, The Ohio State University, Columbus, OH, USA
| | - Noah Lepola
- Departments of Molecular Genetics, Cancer Biology and Genetics, The Ohio State University, Columbus, OH, USA
| | - Jazmin Urrutia
- Departments of Molecular Genetics, Cancer Biology and Genetics, The Ohio State University, Columbus, OH, USA
| | - Lisa M Christian
- Department of Psychiatry and Behavioral Health, Institute for Behavioral Medicine Research, The Ohio State University, Columbus, OH, USA
| | - Carolyn Presley
- Division of Medical Oncology, The Ohio State University, Columbus, OH, USA
| | - Alice Mims
- Division of Hematology, The Ohio State University, Columbus, OH, USA
| | - Christin E Burd
- Departments of Molecular Genetics, Cancer Biology and Genetics, The Ohio State University, Columbus, OH, USA
| |
Collapse
|
27
|
van Hilten A, Katz S, Saccenti E, Niessen WJ, Roshchupkin GV. Designing interpretable deep learning applications for functional genomics: a quantitative analysis. Brief Bioinform 2024; 25:bbae449. [PMID: 39293804 PMCID: PMC11410376 DOI: 10.1093/bib/bbae449] [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: 04/05/2024] [Revised: 08/07/2024] [Accepted: 08/28/2024] [Indexed: 09/20/2024] Open
Abstract
Deep learning applications have had a profound impact on many scientific fields, including functional genomics. Deep learning models can learn complex interactions between and within omics data; however, interpreting and explaining these models can be challenging. Interpretability is essential not only to help progress our understanding of the biological mechanisms underlying traits and diseases but also for establishing trust in these model's efficacy for healthcare applications. Recognizing this importance, recent years have seen the development of numerous diverse interpretability strategies, making it increasingly difficult to navigate the field. In this review, we present a quantitative analysis of the challenges arising when designing interpretable deep learning solutions in functional genomics. We explore design choices related to the characteristics of genomics data, the neural network architectures applied, and strategies for interpretation. By quantifying the current state of the field with a predefined set of criteria, we find the most frequent solutions, highlight exceptional examples, and identify unexplored opportunities for developing interpretable deep learning models in genomics.
Collapse
Affiliation(s)
- Arno van Hilten
- Department of Radiology and Nuclear Medicine, Erasmus MC, 3015 GD Rotterdam, The Netherlands
| | - Sonja Katz
- Department of Radiology and Nuclear Medicine, Erasmus MC, 3015 GD Rotterdam, The Netherlands
- Laboratory of Systems and Synthetic Biology, Wageningen University & Research, 6700 HB Wageningen WE, The Netherlands
| | - Edoardo Saccenti
- Laboratory of Systems and Synthetic Biology, Wageningen University & Research, 6700 HB Wageningen WE, The Netherlands
| | - Wiro J Niessen
- Department of Imaging Physics, Delft University of Technology, 2628 CD Delft, The Netherlands
| | - Gennady V Roshchupkin
- Department of Radiology and Nuclear Medicine, Erasmus MC, 3015 GD Rotterdam, The Netherlands
- Department of Epidemiology, Erasmus MC, 3015 GD Rotterdam, The Netherlands
| |
Collapse
|
28
|
Martínez-Magaña JJ, Hurtado-Soriano J, Rivero-Segura NA, Montalvo-Ortiz JL, Garcia-delaTorre P, Becerril-Rojas K, Gomez-Verjan JC. Towards a Novel Frontier in the Use of Epigenetic Clocks in Epidemiology. Arch Med Res 2024; 55:103033. [PMID: 38955096 DOI: 10.1016/j.arcmed.2024.103033] [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: 01/10/2024] [Revised: 05/10/2024] [Accepted: 06/17/2024] [Indexed: 07/04/2024]
Abstract
Health problems associated with aging are a major public health concern for the future. Aging is a complex process with wide intervariability among individuals. Therefore, there is a need for innovative public health strategies that target factors associated with aging and the development of tools to assess the effectiveness of these strategies accurately. Novel approaches to measure biological age, such as epigenetic clocks, have become relevant. These clocks use non-sequential variable information from the genome and employ mathematical algorithms to estimate biological age based on DNA methylation levels. Therefore, in the present study, we comprehensively review the current status of the epigenetic clocks and their associations across the human phenome. We emphasize the potential utility of these tools in an epidemiological context, particularly in evaluating the impact of public health interventions focused on promoting healthy aging. Our review describes associations between epigenetic clocks and multiple traits across the life and health span. Additionally, we highlighted the evolution of studies beyond mere associations to establish causal mechanisms between epigenetic age and disease. We explored the application of epigenetic clocks to measure the efficacy of interventions focusing on rejuvenation.
Collapse
Affiliation(s)
- José Jaime Martínez-Magaña
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA; U.S. Department of Veterans Affairs National Center for Post-Traumatic Stress Disorder, Clinical Neuroscience Division, West Haven, CT, USA; VA 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 Veterans Affairs National Center for Post-Traumatic Stress Disorder, Clinical Neuroscience Division, West Haven, CT, USA; VA Connecticut Healthcare System, West Haven, CT, USA
| | - Paola Garcia-delaTorre
- Unidad de Investigación Epidemiológica y en Servicios de Salud, Área de Envejecimiento, Centro Médico Nacional, Siglo XXI, Instituto Mexicano del Seguro Social, Mexico City, Mexico
| | | | | |
Collapse
|
29
|
Roig-Genoves JV, García-Giménez JL, Mena-Molla S. A miRNA-based epigenetic molecular clock for biological skin-age prediction. Arch Dermatol Res 2024; 316:326. [PMID: 38822910 PMCID: PMC11144124 DOI: 10.1007/s00403-024-03129-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Revised: 04/27/2024] [Accepted: 05/02/2024] [Indexed: 06/03/2024]
Abstract
Skin aging is one of the visible characteristics of the aging process in humans. In recent years, different biological clocks have been generated based on protein or epigenetic markers, but few have focused on biological age in the skin. Arrest the aging process or even being able to restore an organism from an older to a younger stage is one of the main challenges in the last 20 years in biomedical research. We have implemented several machine learning models, including regression and classification algorithms, in order to create an epigenetic molecular clock based on miRNA expression profiles of healthy subjects to predict biological age-related to skin. Our best models are capable of classifying skin samples according to age groups (18-28; 29-39; 40-50; 51-60 or 61-83 years old) with an accuracy of 80% or predict age with a mean absolute error of 10.89 years using the expression levels of 1856 unique miRNAs. Our results suggest that this kind of epigenetic clocks arises as a promising tool with several applications in the pharmaco-cosmetic industry.
Collapse
Affiliation(s)
| | - José Luis García-Giménez
- Consortium Center for Biomedical Network Research on Rare Diseases (CIBERER), Institute of Health Carlos III, Valencia, 46010, Spain
- INCLIVA Health Research Institute, INCLIVA, Valencia, 46010, Spain
- EpiDisease S.L (Spin-off from the CIBER-ISCIII), Parc Científic de la Universitat de Valencia, Paterna, 46980, Spain
- Department of Physiology, Faculty of Pharmacy, University of Valencia, Burjassot, 46100, Spain
| | - Salvador Mena-Molla
- INCLIVA Health Research Institute, INCLIVA, Valencia, 46010, Spain.
- EpiDisease S.L (Spin-off from the CIBER-ISCIII), Parc Científic de la Universitat de Valencia, Paterna, 46980, Spain.
- Department of Physiology, Faculty of Pharmacy, University of Valencia, Burjassot, 46100, Spain.
| |
Collapse
|
30
|
Zhao J, Li H, Qu J, Zong X, Liu Y, Kuang Z, Wang H. A multi-organization epigenetic age prediction based on a channel attention perceptron networks. Front Genet 2024; 15:1393856. [PMID: 38725481 PMCID: PMC11080615 DOI: 10.3389/fgene.2024.1393856] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2024] [Accepted: 04/09/2024] [Indexed: 05/12/2024] Open
Abstract
DNA methylation indicates the individual's aging, so-called Epigenetic clocks, which will improve the research and diagnosis of aging diseases by investigating the correlation between methylation loci and human aging. Although this discovery has inspired many researchers to develop traditional computational methods to quantify the correlation and predict the chronological age, the performance bottleneck delayed access to the practical application. Since artificial intelligence technology brought great opportunities in research, we proposed a perceptron model integrating a channel attention mechanism named PerSEClock. The model was trained on 24,516 CpG loci that can utilize the samples from all types of methylation identification platforms and tested on 15 independent datasets against seven methylation-based age prediction methods. PerSEClock demonstrated the ability to assign varying weights to different CpG loci. This feature allows the model to enhance the weight of age-related loci while reducing the weight of irrelevant loci. The method is free to use for academics at www.dnamclock.com/#/original.
Collapse
Affiliation(s)
- Jian Zhao
- School of Computer Science and Technology, Changchun University, Changchun, China
| | - Haixia Li
- School of Computer Science and Technology, Changchun University, Changchun, China
| | - Jing Qu
- School of Computer Science and Technology, Jilin University, Changchun, China
- School of Information Science and Technology, Institute of Computational Biology, Northeast Normal University, Changchun, China
| | - Xizeng Zong
- Clinical Research Centre, Guangzhou First People’s Hospital, School of Medicine, South China University of Technology, Guangzhou, Guangdong, China
- School of Computer Science and Engineering, Changchun University of Technology, Changchun, China
| | - Yuchen Liu
- Department of Medicine, Boston University School of Medicine, Boston, MA, United States
| | - Zhejun Kuang
- School of Computer Science and Technology, Changchun University, Changchun, China
| | - Han Wang
- School of Information Science and Technology, Institute of Computational Biology, Northeast Normal University, Changchun, China
| |
Collapse
|
31
|
Meng D, Zhang S, Huang Y, Mao K, Han JDJ. Application of AI in biological age prediction. Curr Opin Struct Biol 2024; 85:102777. [PMID: 38310737 DOI: 10.1016/j.sbi.2024.102777] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2023] [Revised: 12/12/2023] [Accepted: 01/15/2024] [Indexed: 02/06/2024]
Abstract
The development of anti-aging interventions requires quantitative measurement of biological age. Machine learning models, known as "aging clocks," are built by leveraging diverse aging biomarkers that vary across lifespan to predict biological age. In addition to traditional aging clocks harnessing epigenetic signatures derived from bulk samples, emerging technologies allow the biological age estimating at single-cell level to dissect cellular diversity in aging tissues. Moreover, imaging-based aging clocks are increasingly employed with the advantage of non-invasive measurement, making it suitable for large-scale human cohort studies. To fully capture the features in the ever-growing multi-modal and high-dimensional aging-related data and uncover disease associations, deep-learning based approaches, which are effective to learn complex and non-linear relationships without relying on pre-defined features, are increasingly applied. The use of big data and AI-based aging clocks has achieved high accuracy, interpretability and generalizability, guiding clinical applications to delay age-related diseases and extend healthy lifespans.
Collapse
Affiliation(s)
- Dawei Meng
- Peking-Tsinghua Center for Life Sciences, Academy for Advanced Interdisciplinary Studies, Center for Quantitative Biology (CQB), Peking University, Beijing 100871, China
| | - Shiqiang Zhang
- Peking-Tsinghua Center for Life Sciences, Academy for Advanced Interdisciplinary Studies, Center for Quantitative Biology (CQB), Peking University, Beijing 100871, China
| | - Yuanfang Huang
- Peking-Tsinghua Center for Life Sciences, Academy for Advanced Interdisciplinary Studies, Center for Quantitative Biology (CQB), Peking University, Beijing 100871, China
| | - Kehang Mao
- Peking-Tsinghua Center for Life Sciences, Academy for Advanced Interdisciplinary Studies, Center for Quantitative Biology (CQB), Peking University, Beijing 100871, China
| | - Jing-Dong J Han
- Peking-Tsinghua Center for Life Sciences, Academy for Advanced Interdisciplinary Studies, Center for Quantitative Biology (CQB), Peking University, Beijing 100871, China.
| |
Collapse
|
32
|
de Lima Camillo LP. pyaging: a Python-based compendium of GPU-optimized aging clocks. Bioinformatics 2024; 40:btae200. [PMID: 38603598 PMCID: PMC11058068 DOI: 10.1093/bioinformatics/btae200] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2023] [Revised: 03/28/2024] [Accepted: 04/09/2024] [Indexed: 04/13/2024] Open
Abstract
MOTIVATION Aging is intricately linked to diseases and mortality. It is reflected in molecular changes across various tissues which can be leveraged for the development of biomarkers of aging using machine learning models, known as aging clocks. Despite advancements in the field, a significant challenge remains: the lack of robust, Python-based software tools for integrating and comparing these diverse models. This gap highlights the need for comprehensive solutions that can handle the complexity and variety of data in aging research. RESULTS To address this gap, I introduce pyaging, a comprehensive open-source Python package designed to facilitate aging research. pyaging harmonizes dozens of aging clocks, covering a range of molecular data types such as DNA methylation, transcriptomics, histone mark ChIP-Seq, and ATAC-Seq. The package is not limited to traditional model types; it features a diverse array, from linear and principal component models to neural networks and automatic relevance determination models. Thanks to a PyTorch-based backend that enables GPU acceleration, pyaging is capable of rapid inference, even when dealing with large datasets and complex models. In addition, the package's support for multi-species analysis extends its utility across various organisms, including humans, various mammals, and Caenorhabditis elegans. AVAILABILITY AND IMPLEMENTATION pyaging is accessible on GitHub, at https://github.com/rsinghlab/pyaging, and the distribution is available on PyPi, at https://pypi.org/project/pyaging/. The software is also archived on Zenodo, at https://zenodo.org/doi/10.5281/zenodo.10335011.
Collapse
|
33
|
Georgievskaya A, Tlyachev T, Kiselev K, Hillebrand G, Chekanov K, Danko D, Golodyaev A, Majmudar G. Predicting human chronological age via AI analysis of dorsal hand versus facial images: A study in a cohort of Indian females. Exp Dermatol 2024; 33:e15045. [PMID: 38509744 DOI: 10.1111/exd.15045] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Revised: 02/11/2024] [Accepted: 02/18/2024] [Indexed: 03/22/2024]
Abstract
Predicting a person's chronological age (CA) from visible skin features using artificial intelligence (AI) is now commonplace. Often, convolutional neural network (CNN) models are built using images of the face as biometric data. However, hands hold telltale signs of a person's age. To determine the utility of using only hand images in predicting CA, we developed two deep CNNs based on 1) dorsal hand images (H) and 2) frontal face images (F). Subjects (n = 1454) were Indian women, 20-80 years, across three geographic cohorts (Mumbai, New Delhi and Bangalore) and having a broad variation in skin tones. Images were randomised: 70% of F and 70% of H were used to train CNNs. The remaining 30% of F and H were retained for validation. CNN validation showed mean absolute error for predicting CA using F and H of 4.1 and 4.7 years, respectively. In both cases correlations of predicted and actual age were statistically significant (r(F) = 0.93, r(H) = 0.90). The CNNs for F and H were validated for dark and light skin tones. Finally, by blurring or accentuating visible features on specific regions of the hand and face, we identified those features that contributed to the CNN models. For the face, areas of the inner eye corner and around the mouth were most important for age prediction. For the hands, knuckle texture was a key driver for age prediction. Collectively, for AI estimates of CA, CNNs based solely on hand images are a viable alternative and comparable to CNNs based on facial images.
Collapse
Affiliation(s)
| | | | | | - Greg Hillebrand
- James L. Winkle College of Pharmacy, University of Cincinnati, Cincinnati, Ohio, USA
| | | | | | | | - Gopa Majmudar
- James L. Winkle College of Pharmacy, University of Cincinnati, Cincinnati, Ohio, USA
| |
Collapse
|
34
|
Prosz A, Pipek O, Börcsök J, Palla G, Szallasi Z, Spisak S, Csabai I. Biologically informed deep learning for explainable epigenetic clocks. Sci Rep 2024; 14:1306. [PMID: 38225268 PMCID: PMC10789766 DOI: 10.1038/s41598-023-50495-5] [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: 01/17/2023] [Accepted: 12/20/2023] [Indexed: 01/17/2024] Open
Abstract
Ageing is often characterised by progressive accumulation of damage, and it is one of the most important risk factors for chronic disease development. Epigenetic mechanisms including DNA methylation could functionally contribute to organismal aging, however the key functions and biological processes may govern ageing are still not understood. Although age predictors called epigenetic clocks can accurately estimate the biological age of an individual based on cellular DNA methylation, their models have limited ability to explain the prediction algorithm behind and underlying key biological processes controlling ageing. Here we present XAI-AGE, a biologically informed, explainable deep neural network model for accurate biological age prediction across multiple tissue types. We show that XAI-AGE outperforms the first-generation age predictors and achieves similar results to deep learning-based models, while opening up the possibility to infer biologically meaningful insights of the activity of pathways and other abstract biological processes directly from the model.
Collapse
Affiliation(s)
- Aurel Prosz
- Danish Cancer Institute, Copenhagen, Denmark
| | - Orsolya Pipek
- Department of Physics of Complex Systems, ELTE Eötvös Loránd University, Budapest, Hungary
| | - Judit Börcsök
- Danish Cancer Institute, Copenhagen, Denmark
- Biotech Research & Innovation Centre (BRIC), University of Copenhagen, Copenhagen, Denmark
| | - Gergely Palla
- Department of Biological Physics, ELTE Eötvös Loránd University, Budapest, Hungary
- Health Services Management Training Centre, Semmelweis University, Budapest, Hungary
| | | | - Sandor Spisak
- Institute of Enzymology, HUN-REN Research Centre for Natural Sciences, Budapest, Hungary.
| | - István Csabai
- Department of Physics of Complex Systems, ELTE Eötvös Loránd University, Budapest, Hungary
| |
Collapse
|
35
|
Abudahab S, Slattum PW, Price ET, McClay JL. Epigenetic regulation of drug metabolism in aging: utilizing epigenetics to optimize geriatric pharmacotherapy. Pharmacogenomics 2024; 25:41-54. [PMID: 38126340 PMCID: PMC10794944 DOI: 10.2217/pgs-2023-0199] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2023] [Accepted: 12/06/2023] [Indexed: 12/23/2023] Open
Abstract
We explore the relationship between epigenetic aging and drug metabolism. We review current evidence for changes in drug metabolism in normal aging, followed by a description of how epigenetic modifications associated with age can regulate the expression and functionality of genes. In particular, we focus on the role of epigenome-wide studies of human and mouse liver in understanding these age-related processes with respect to xenobiotic processing. We highlight genes encoding drug metabolizing enzymes and transporters revealed to be affected by epigenetic aging in these studies. We conclude that substantial evidence exists for epigenetic aging impacting drug metabolism and transport genes, but more work is needed. We further highlight the promise of pharmacoepigenetics applied to enhancing drug safety in older adults.
Collapse
Affiliation(s)
- Sara Abudahab
- Department of Pharmacotherapy & Outcomes Science, School of Pharmacy, Virginia Commonwealth University, Richmond, VA 23298, USA
| | - Patricia W Slattum
- Department of Pharmacotherapy & Outcomes Science, School of Pharmacy, Virginia Commonwealth University, Richmond, VA 23298, USA
- Virginia Center on Aging, Virginia Commonwealth University, Richmond, VA 23298, USA
| | - Elvin T Price
- Department of Pharmacotherapy & Outcomes Science, School of Pharmacy, Virginia Commonwealth University, Richmond, VA 23298, USA
| | - Joseph L McClay
- Department of Pharmacotherapy & Outcomes Science, School of Pharmacy, Virginia Commonwealth University, Richmond, VA 23298, USA
| |
Collapse
|
36
|
Kalyakulina A, Yusipov I, Moskalev A, Franceschi C, Ivanchenko M. eXplainable Artificial Intelligence (XAI) in aging clock models. Ageing Res Rev 2024; 93:102144. [PMID: 38030090 DOI: 10.1016/j.arr.2023.102144] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2023] [Revised: 11/07/2023] [Accepted: 11/23/2023] [Indexed: 12/01/2023]
Abstract
XAI is a rapidly progressing field of machine learning, aiming to unravel the predictions of complex models. XAI is especially required in sensitive applications, e.g. in health care, when diagnosis, recommendations and treatment choices might rely on the decisions made by artificial intelligence systems. AI approaches have become widely used in aging research as well, in particular, in developing biological clock models and identifying biomarkers of aging and age-related diseases. However, the potential of XAI here awaits to be fully appreciated. We discuss the application of XAI for developing the "aging clocks" and present a comprehensive analysis of the literature categorized by the focus on particular physiological systems.
Collapse
Affiliation(s)
- Alena Kalyakulina
- Institute of Biogerontology, Lobachevsky State University, Nizhny Novgorod 603022, Russia; Research Center for Trusted Artificial Intelligence, The Ivannikov Institute for System Programming of the Russian Academy of Sciences, Moscow 109004, Russia; Department of Applied Mathematics, Institute of Information Technologies, Mathematics and Mechanics, Lobachevsky State University, Nizhny Novgorod 603022, Russia.
| | - Igor Yusipov
- Institute of Biogerontology, Lobachevsky State University, Nizhny Novgorod 603022, Russia; Research Center for Trusted Artificial Intelligence, The Ivannikov Institute for System Programming of the Russian Academy of Sciences, Moscow 109004, Russia; Department of Applied Mathematics, Institute of Information Technologies, Mathematics and Mechanics, Lobachevsky State University, Nizhny Novgorod 603022, Russia
| | - Alexey Moskalev
- Institute of Biogerontology, Lobachevsky State University, Nizhny Novgorod 603022, Russia
| | - Claudio Franceschi
- Institute of Biogerontology, Lobachevsky State University, Nizhny Novgorod 603022, Russia
| | - Mikhail Ivanchenko
- Institute of Biogerontology, Lobachevsky State University, Nizhny Novgorod 603022, Russia; Department of Applied Mathematics, Institute of Information Technologies, Mathematics and Mechanics, Lobachevsky State University, Nizhny Novgorod 603022, Russia
| |
Collapse
|
37
|
Shi L, Hai B, Kuang Z, Wang H, Zhao J. ResnetAge: A Resnet-Based DNA Methylation Age Prediction Method. Bioengineering (Basel) 2023; 11:34. [PMID: 38247911 PMCID: PMC10813502 DOI: 10.3390/bioengineering11010034] [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: 11/08/2023] [Revised: 12/13/2023] [Accepted: 12/26/2023] [Indexed: 01/23/2024] Open
Abstract
Aging is a significant contributing factor to degenerative diseases such as cancer. The extent of DNA methylation in human cells indicates the aging process and screening for age-related methylation sites can be used to construct epigenetic clocks. Thereby, it can be a new aging-detecting marker for clinical diagnosis and treatments. Predicting the biological age of human individuals is conducive to the study of physical aging problems. Although many researchers have developed epigenetic clock prediction methods based on traditional machine learning and even deep learning, higher prediction accuracy is still required to match the clinical applications. Here, we proposed an epigenetic clock prediction method based on a Resnet neuro networks model named ResnetAge. The model accepts 22,278 CpG sites as a sample input, supporting both the Illumina 27K and 450K identification frameworks. It was trained using 32 public datasets containing multiple tissues such as whole blood, saliva, and mouth. The Mean Absolute Error (MAE) of the training set is 1.29 years, and the Median Absolute Deviation (MAD) is 0.98 years. The Mean Absolute Error (MAE) of the validation set is 3.24 years, and the Median Absolute Deviation (MAD) is 2.3 years. Our method has higher accuracy in age prediction in comparison with other methylation-based age prediction methods.
Collapse
Affiliation(s)
- Lijuan Shi
- Key Laboratory of Intelligent Rehabilitation and Barrier-Free for the Disabled (Changchun University), Ministry of Education, Changchun University, Changchun 130012, China; (L.S.); (B.H.)
- Jilin Provincial Key Laboratory of Human Health Status Identification & Function Enhancement, Changchun 130022, China
| | - Boquan Hai
- Key Laboratory of Intelligent Rehabilitation and Barrier-Free for the Disabled (Changchun University), Ministry of Education, Changchun University, Changchun 130012, China; (L.S.); (B.H.)
- Jilin Provincial Key Laboratory of Human Health Status Identification & Function Enhancement, Changchun 130022, China
| | - Zhejun Kuang
- Key Laboratory of Intelligent Rehabilitation and Barrier-Free for the Disabled (Changchun University), Ministry of Education, Changchun University, Changchun 130012, China; (L.S.); (B.H.)
- Jilin Provincial Key Laboratory of Human Health Status Identification & Function Enhancement, Changchun 130022, China
| | - Han Wang
- The Institution of Computational Biology of Northeast Normal University, Changchun 130000, China;
| | - Jian Zhao
- Key Laboratory of Intelligent Rehabilitation and Barrier-Free for the Disabled (Changchun University), Ministry of Education, Changchun University, Changchun 130012, China; (L.S.); (B.H.)
- Jilin Provincial Key Laboratory of Human Health Status Identification & Function Enhancement, Changchun 130022, China
| |
Collapse
|
38
|
Yu D, Li M, Linghu G, Hu Y, Hajdarovic KH, Wang A, Singh R, Webb AE. CellBiAge: Improved single-cell age classification using data binarization. Cell Rep 2023; 42:113500. [PMID: 38032797 PMCID: PMC10791072 DOI: 10.1016/j.celrep.2023.113500] [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: 05/04/2023] [Revised: 10/20/2023] [Accepted: 11/13/2023] [Indexed: 12/02/2023] Open
Abstract
Aging is a major risk factor for many diseases. Accurate methods for predicting age in specific cell types are essential to understand the heterogeneity of aging and to assess rejuvenation strategies. However, classifying organismal age at single-cell resolution using transcriptomics is challenging due to sparsity and noise. Here, we developed CellBiAge, a robust and easy-to-implement machine learning pipeline, to classify the age of single cells in the mouse brain using single-cell transcriptomics. We show that binarization of gene expression values for the top highly variable genes significantly improved test performance across different models, techniques, sexes, and brain regions, with potential age-related genes identified for model prediction. Additionally, we demonstrate CellBiAge's ability to capture exercise-induced rejuvenation in neural stem cells. This study provides a broadly applicable approach for robust classification of organismal age of single cells in the mouse brain, which may aid in understanding the aging process and evaluating rejuvenation methods.
Collapse
Affiliation(s)
- Doudou Yu
- Molecular Biology, Cell Biology, and Biochemistry Graduate Program, Brown University, Providence, RI 02912, USA; Data Science Institute, Brown University, Providence, RI 02912, USA
| | - Manlin Li
- Data Science Institute, Brown University, Providence, RI 02912, USA
| | - Guanjie Linghu
- Data Science Institute, Brown University, Providence, RI 02912, USA
| | - Yihuan Hu
- Data Science Institute, Brown University, Providence, RI 02912, USA
| | | | - An Wang
- Department of Applied Mathematics & Statistics, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Ritambhara Singh
- Department of Computer Science, Brown University, Providence, RI 02912, USA; Center for Computational Molecular Biology, Brown University, Providence, RI 02912, USA.
| | - Ashley E Webb
- Department of Molecular Biology, Cell Biology, and Biochemistry, Brown University, Providence, RI 02912, USA; Center on the Biology of Aging, Brown University, Providence, RI 02912, USA; Carney Institute for Brain Science, Brown University, Providence, RI 02912, USA; Center for Translational Neuroscience, Brown University, Providence, RI 02912, USA.
| |
Collapse
|
39
|
Pathare ADS, Loid M, Saare M, Gidlöf SB, Zamani Esteki M, Acharya G, Peters M, Salumets A. Endometrial receptivity in women of advanced age: an underrated factor in infertility. Hum Reprod Update 2023; 29:773-793. [PMID: 37468438 PMCID: PMC10628506 DOI: 10.1093/humupd/dmad019] [Citation(s) in RCA: 44] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Revised: 05/24/2023] [Indexed: 07/21/2023] Open
Abstract
BACKGROUND Modern lifestyle has led to an increase in the age at conception. Advanced age is one of the critical risk factors for female-related infertility. It is well known that maternal age positively correlates with the deterioration of oocyte quality and chromosomal abnormalities in oocytes and embryos. The effect of age on endometrial function may be an equally important factor influencing implantation rate, pregnancy rate, and overall female fertility. However, there are only a few published studies on this topic, suggesting that this area has been under-explored. Improving our knowledge of endometrial aging from the biological (cellular, molecular, histological) and clinical perspectives would broaden our understanding of the risks of age-related female infertility. OBJECTIVE AND RATIONALE The objective of this narrative review is to critically evaluate the existing literature on endometrial aging with a focus on synthesizing the evidence for the impact of endometrial aging on conception and pregnancy success. This would provide insights into existing gaps in the clinical application of research findings and promote the development of treatment options in this field. SEARCH METHODS The review was prepared using PubMed (Medline) until February 2023 with the keywords such as 'endometrial aging', 'receptivity', 'decidualization', 'hormone', 'senescence', 'cellular', 'molecular', 'methylation', 'biological age', 'epigenetic', 'oocyte recipient', 'oocyte donation', 'embryo transfer', and 'pregnancy rate'. Articles in a language other than English were excluded. OUTCOMES In the aging endometrium, alterations occur at the molecular, cellular, and histological levels suggesting that aging has a negative effect on endometrial biology and may impair endometrial receptivity. Additionally, advanced age influences cellular senescence, which plays an important role during the initial phase of implantation and is a major obstacle in the development of suitable senolytic agents for endometrial aging. Aging is also accountable for chronic conditions associated with inflammaging, which eventually can lead to increased pro-inflammation and tissue fibrosis. Furthermore, advanced age influences epigenetic regulation in the endometrium, thus altering the relation between its epigenetic and chronological age. The studies in oocyte donation cycles to determine the effect of age on endometrial receptivity with respect to the rates of implantation, clinical pregnancy, miscarriage, and live birth have revealed contradictory inferences indicating the need for future research on the mechanisms and corresponding causal effects of women's age on endometrial receptivity. WIDER IMPLICATIONS Increasing age can be accountable for female infertility and IVF failures. Based on the complied observations and synthesized conclusions in this review, advanced age has been shown to have a negative impact on endometrial functioning. This information can provide recommendations for future research focusing on molecular mechanisms of age-related cellular senescence, cellular composition, and transcriptomic changes in relation to endometrial aging. Additionally, further prospective research is needed to explore newly emerging therapeutic options, such as the senolytic agents that can target endometrial aging without affecting decidualization. Moreover, clinical trial protocols, focusing on oocyte donation cycles, would be beneficial in understanding the direct clinical implications of endometrial aging on pregnancy outcomes.
Collapse
Affiliation(s)
- Amruta D S Pathare
- Department of Obstetrics and Gynaecology, Institute of Clinical Medicine, University of Tartu, Tartu, Estonia
| | - Marina Loid
- Department of Obstetrics and Gynaecology, Institute of Clinical Medicine, University of Tartu, Tartu, Estonia
- Competence Centre on Health Technologies, Tartu, Estonia
| | - Merli Saare
- Department of Obstetrics and Gynaecology, Institute of Clinical Medicine, University of Tartu, Tartu, Estonia
- Competence Centre on Health Technologies, Tartu, Estonia
| | - Sebastian Brusell Gidlöf
- Division of Obstetrics and Gynaecology, Department of Clinical Science, Intervention and Technology (CLINTEC), Karolinska Institute and Karolinska University Hospital, Stockholm, Sweden
- Department of Gynecology and Reproductive Medicine, Karolinska University Hospital, Stockholm, Sweden
| | - Masoud Zamani Esteki
- Division of Obstetrics and Gynaecology, Department of Clinical Science, Intervention and Technology (CLINTEC), Karolinska Institute and Karolinska University Hospital, Stockholm, Sweden
- Department of Clinical Genetics, Maastricht University Medical Centre+, Maastricht, The Netherlands
- Department of Genetics and Cell Biology, GROW School for Oncology and Reproduction, Maastricht University, Maastricht, The Netherlands
| | - Ganesh Acharya
- Division of Obstetrics and Gynaecology, Department of Clinical Science, Intervention and Technology (CLINTEC), Karolinska Institute and Karolinska University Hospital, Stockholm, Sweden
- Department of Clinical Medicine, Women’s Health and Perinatology Research Group, UiT The Arctic University of Norway, Tromsø, Norway
| | - Maire Peters
- Department of Obstetrics and Gynaecology, Institute of Clinical Medicine, University of Tartu, Tartu, Estonia
- Competence Centre on Health Technologies, Tartu, Estonia
| | - Andres Salumets
- Department of Obstetrics and Gynaecology, Institute of Clinical Medicine, University of Tartu, Tartu, Estonia
- Competence Centre on Health Technologies, Tartu, Estonia
- Division of Obstetrics and Gynaecology, Department of Clinical Science, Intervention and Technology (CLINTEC), Karolinska Institute and Karolinska University Hospital, Stockholm, Sweden
| |
Collapse
|
40
|
Klose D, Needhamsen M, Ringh MV, Hagemann-Jensen M, Jagodic M, Kular L. Smoking affects epigenetic ageing of lung bronchoalveolar lavage cells in Multiple Sclerosis. Mult Scler Relat Disord 2023; 79:104991. [PMID: 37708820 DOI: 10.1016/j.msard.2023.104991] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Revised: 06/18/2023] [Accepted: 09/02/2023] [Indexed: 09/16/2023]
Abstract
BACKGROUND A compelling body of evidence implicates cigarette smoking and lung inflammation in Multiple Sclerosis (MS) susceptibility and progression. Previous studies have reported epigenetic age (DNAm age) acceleration in blood immune cells and in glial cells of people with MS (pwMS) compared to healthy controls (HC). OBJECTIVES We aimed to examine biological ageing in lung immune cells in the context of MS and smoking. METHODS We analyzed age acceleration residuals in lung bronchoalveolar lavage (BAL) cells, constituted of mainly alveolar macrophages, from 17 pwMS and 22 HC in relation to smoking using eight DNA methylation-based clocks, namely AltumAge, Horvath, GrimAge, PhenoAge, Zhang, SkinBlood, Hannum, Monocyte clock as well as two RNA-based clocks, which capture different aspects of biological ageing. RESULTS After adjustment for covariates, five epigenetic clocks showed significant differences between the groups. Four of them, Horvath (Padj = 0.028), GrimAge (Padj = 4.28 × 10-7), SkinBlood (Padj = 0.001) and Zhang (Padj = 0.02), uncovered the sole effect of smoking on ageing estimates, irrespective of the clinical group. The Horvath, SkinBlood and Zhang clocks showed a negative impact of smoking while GrimAge detected smoking-associated age acceleration in BAL cells. On the contrary, the AltumAge clock revealed differences between pwMS and HC and indicated that, in the absence of smoking, BAL cells of pwMS were epigenetically 5.4 years older compared to HC (Padj = 0.028). Smoking further affected epigenetic ageing in BAL cells of pwMS specifically as non-smoking pwMS exhibited a 10.2-year AltumAge acceleration compared to pwMS smokers (Padj = 0.0049). Of note, blood-derived monocytes did not show any MS-specific or smoking-related AltumAge differences. The difference between BAL cells of pwMS smokers and non-smokers was attributable to the differential methylation of 114 AltumAge-CpGs (Padj < 0.05) affecting genes involved in innate immune processes such as cytokine production, defense response and cell motility. These changes functionally translated into transcriptional differences in BAL cells between pwMS smokers and non-smokers. CONCLUSIONS BAL cells of pwMS display inflammation-related and smoking-dependent changes associated to epigenetic ageing captured by the AltumAge clock. Future studies examining potential confounders, such as the distribution of distinct BAL myeloid cell types in pwMS compared to control individuals in relation to smoking may clarify the varying performance and DNAm age estimations among epigenetic clocks.
Collapse
Affiliation(s)
- Dennis Klose
- Department of Clinical Neuroscience, Karolinska Institutet, Center for Molecular Medicine, Karolinska University Hospital, 171 76 Stockholm, Sweden
| | - Maria Needhamsen
- Department of Clinical Neuroscience, Karolinska Institutet, Center for Molecular Medicine, Karolinska University Hospital, 171 76 Stockholm, Sweden
| | - Mikael V Ringh
- Department of Clinical Neuroscience, Karolinska Institutet, Center for Molecular Medicine, Karolinska University Hospital, 171 76 Stockholm, Sweden
| | | | - Maja Jagodic
- Department of Clinical Neuroscience, Karolinska Institutet, Center for Molecular Medicine, Karolinska University Hospital, 171 76 Stockholm, Sweden
| | - Lara Kular
- Department of Clinical Neuroscience, Karolinska Institutet, Center for Molecular Medicine, Karolinska University Hospital, 171 76 Stockholm, Sweden.
| |
Collapse
|
41
|
Statsenko Y, Kuznetsov NV, Morozova D, Liaonchyk K, Simiyu GL, Smetanina D, Kashapov A, Meribout S, Gorkom KNV, Hamoudi R, Ismail F, Ansari SA, Emerald BS, Ljubisavljevic M. Reappraisal of the Concept of Accelerated Aging in Neurodegeneration and Beyond. Cells 2023; 12:2451. [PMID: 37887295 PMCID: PMC10605227 DOI: 10.3390/cells12202451] [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: 08/04/2023] [Revised: 09/01/2023] [Accepted: 09/06/2023] [Indexed: 10/28/2023] Open
Abstract
BACKGROUND Genetic and epigenetic changes, oxidative stress and inflammation influence the rate of aging, which diseases, lifestyle and environmental factors can further accelerate. In accelerated aging (AA), the biological age exceeds the chronological age. OBJECTIVE The objective of this study is to reappraise the AA concept critically, considering its weaknesses and limitations. METHODS We reviewed more than 300 recent articles dealing with the physiology of brain aging and neurodegeneration pathophysiology. RESULTS (1) Application of the AA concept to individual organs outside the brain is challenging as organs of different systems age at different rates. (2) There is a need to consider the deceleration of aging due to the potential use of the individual structure-functional reserves. The latter can be restored by pharmacological and/or cognitive therapy, environment, etc. (3) The AA concept lacks both standardised terminology and methodology. (4) Changes in specific molecular biomarkers (MBM) reflect aging-related processes; however, numerous MBM candidates should be validated to consolidate the AA theory. (5) The exact nature of many potential causal factors, biological outcomes and interactions between the former and the latter remain largely unclear. CONCLUSIONS Although AA is commonly recognised as a perspective theory, it still suffers from a number of gaps and limitations that assume the necessity for an updated AA concept.
Collapse
Affiliation(s)
- Yauhen Statsenko
- Department of Radiology, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain P.O. Box 15551, United Arab Emirates; (Y.S.); (G.L.S.); (D.S.); (A.K.); (S.M.); (K.N.-V.G.)
- ASPIRE Precision Medicine Research Institute Abu Dhabi, United Arab Emirates University, Al Ain 27272, United Arab Emirates; (D.M.); (K.L.); (R.H.); (S.A.A.); (B.S.E.); (M.L.)
- Big Data Analytic Center, United Arab Emirates University, Al Ain P.O. Box 15551, United Arab Emirates
| | - Nik V. Kuznetsov
- ASPIRE Precision Medicine Research Institute Abu Dhabi, United Arab Emirates University, Al Ain 27272, United Arab Emirates; (D.M.); (K.L.); (R.H.); (S.A.A.); (B.S.E.); (M.L.)
| | - Daria Morozova
- ASPIRE Precision Medicine Research Institute Abu Dhabi, United Arab Emirates University, Al Ain 27272, United Arab Emirates; (D.M.); (K.L.); (R.H.); (S.A.A.); (B.S.E.); (M.L.)
| | - Katsiaryna Liaonchyk
- ASPIRE Precision Medicine Research Institute Abu Dhabi, United Arab Emirates University, Al Ain 27272, United Arab Emirates; (D.M.); (K.L.); (R.H.); (S.A.A.); (B.S.E.); (M.L.)
| | - Gillian Lylian Simiyu
- Department of Radiology, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain P.O. Box 15551, United Arab Emirates; (Y.S.); (G.L.S.); (D.S.); (A.K.); (S.M.); (K.N.-V.G.)
| | - Darya Smetanina
- Department of Radiology, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain P.O. Box 15551, United Arab Emirates; (Y.S.); (G.L.S.); (D.S.); (A.K.); (S.M.); (K.N.-V.G.)
| | - Aidar Kashapov
- Department of Radiology, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain P.O. Box 15551, United Arab Emirates; (Y.S.); (G.L.S.); (D.S.); (A.K.); (S.M.); (K.N.-V.G.)
| | - Sarah Meribout
- Department of Radiology, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain P.O. Box 15551, United Arab Emirates; (Y.S.); (G.L.S.); (D.S.); (A.K.); (S.M.); (K.N.-V.G.)
| | - Klaus Neidl-Van Gorkom
- Department of Radiology, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain P.O. Box 15551, United Arab Emirates; (Y.S.); (G.L.S.); (D.S.); (A.K.); (S.M.); (K.N.-V.G.)
| | - Rifat Hamoudi
- ASPIRE Precision Medicine Research Institute Abu Dhabi, United Arab Emirates University, Al Ain 27272, United Arab Emirates; (D.M.); (K.L.); (R.H.); (S.A.A.); (B.S.E.); (M.L.)
- Department of Clinical Sciences, College of Medicine, University of Sharjah, Sharjah 27272, United Arab Emirates
- Division of Surgery and Interventional Science, University College London, London NW3 2PS, UK
| | - Fatima Ismail
- Department of Pediatrics, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain P.O. Box 15551, United Arab Emirates;
| | - Suraiya Anjum Ansari
- ASPIRE Precision Medicine Research Institute Abu Dhabi, United Arab Emirates University, Al Ain 27272, United Arab Emirates; (D.M.); (K.L.); (R.H.); (S.A.A.); (B.S.E.); (M.L.)
- Department of Biochemistry and Molecular Biology, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain P.O. Box 15551, United Arab Emirates
| | - Bright Starling Emerald
- ASPIRE Precision Medicine Research Institute Abu Dhabi, United Arab Emirates University, Al Ain 27272, United Arab Emirates; (D.M.); (K.L.); (R.H.); (S.A.A.); (B.S.E.); (M.L.)
- Department of Anatomy, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain P.O. Box 15551, United Arab Emirates
| | - Milos Ljubisavljevic
- ASPIRE Precision Medicine Research Institute Abu Dhabi, United Arab Emirates University, Al Ain 27272, United Arab Emirates; (D.M.); (K.L.); (R.H.); (S.A.A.); (B.S.E.); (M.L.)
- Department of Physiology, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain P.O. Box 15551, United Arab Emirates
| |
Collapse
|
42
|
Hughes BK, Wallis R, Bishop CL. Yearning for machine learning: applications for the classification and characterisation of senescence. Cell Tissue Res 2023; 394:1-16. [PMID: 37016180 PMCID: PMC10558380 DOI: 10.1007/s00441-023-03768-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Accepted: 03/05/2023] [Indexed: 04/06/2023]
Abstract
Senescence is a widely appreciated tumour suppressive mechanism, which acts as a barrier to cancer development by arresting cell cycle progression in response to harmful stimuli. However, senescent cell accumulation becomes deleterious in aging and contributes to a wide range of age-related pathologies. Furthermore, senescence has beneficial roles and is associated with a growing list of normal physiological processes including wound healing and embryonic development. Therefore, the biological role of senescent cells has become increasingly nuanced and complex. The emergence of sophisticated, next-generation profiling technologies, such as single-cell RNA sequencing, has accelerated our understanding of the heterogeneity of senescence, with distinct final cell states emerging within models as well as between cell types and tissues. In order to explore data sets of increasing size and complexity, the senescence field has begun to employ machine learning (ML) methodologies to probe these intricacies. Most notably, ML has been used to aid the classification of cells as senescent, as well as to characterise the final senescence phenotypes. Here, we provide a background to the principles of ML tasks, as well as some of the most commonly used methodologies from both traditional and deep ML. We focus on the application of these within the context of senescence research, by addressing the utility of ML for the analysis of data from different laboratory technologies (microscopy, transcriptomics, proteomics, methylomics), as well as the potential within senolytic drug discovery. Together, we aim to highlight both the progress and potential for the application of ML within senescence research.
Collapse
Affiliation(s)
- Bethany K Hughes
- Blizard Institute, Barts and The London Faculty of Medicine and Dentistry, Queen Mary University of London, 4 Newark Street, London, E1 2AT, UK
| | - Ryan Wallis
- Blizard Institute, Barts and The London Faculty of Medicine and Dentistry, Queen Mary University of London, 4 Newark Street, London, E1 2AT, UK
| | - Cleo L Bishop
- Blizard Institute, Barts and The London Faculty of Medicine and Dentistry, Queen Mary University of London, 4 Newark Street, London, E1 2AT, UK.
| |
Collapse
|
43
|
Varshavsky M, Harari G, Glaser B, Dor Y, Shemer R, Kaplan T. Accurate age prediction from blood using a small set of DNA methylation sites and a cohort-based machine learning algorithm. CELL REPORTS METHODS 2023; 3:100567. [PMID: 37751697 PMCID: PMC10545910 DOI: 10.1016/j.crmeth.2023.100567] [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: 01/20/2023] [Revised: 06/18/2023] [Accepted: 08/03/2023] [Indexed: 09/28/2023]
Abstract
Chronological age prediction from DNA methylation sheds light on human aging, health, and lifespan. Current clocks are mostly based on linear models and rely upon hundreds of sites across the genome. Here, we present GP-age, an epigenetic non-linear cohort-based clock for blood, based upon 11,910 methylomes. Using 30 CpG sites alone, GP-age outperforms state-of-the-art models, with a median accuracy of ∼2 years on held-out blood samples, for both array and sequencing-based data. We show that aging-related changes occur at multiple neighboring CpGs, with implications for using fragment-level analysis of sequencing data in aging research. By training three independent clocks, we show enrichment of donors with consistent deviation between predicted and actual age, suggesting individual rates of biological aging. Overall, we provide a compact yet accurate alternative to array-based clocks for blood, with applications in longitudinal aging research, forensic profiling, and monitoring epigenetic processes in transplantation medicine and cancer.
Collapse
Affiliation(s)
- Miri Varshavsky
- School of Computer Science and Engineering, The Hebrew University of Jerusalem, Jerusalem, Israel; The Center for Computational Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Gil Harari
- School of Computer Science and Engineering, The Hebrew University of Jerusalem, Jerusalem, Israel; Department of Developmental Biology and Cancer Research, Institute for Medical Research Israel-Canada, The Hebrew University-Hadassah Medical School, Jerusalem, Israel
| | - Benjamin Glaser
- Department of Endocrinology and Metabolism, Hadassah Medical Center and Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Yuval Dor
- Department of Developmental Biology and Cancer Research, Institute for Medical Research Israel-Canada, The Hebrew University-Hadassah Medical School, Jerusalem, Israel; The Center for Computational Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Ruth Shemer
- Department of Developmental Biology and Cancer Research, Institute for Medical Research Israel-Canada, The Hebrew University-Hadassah Medical School, Jerusalem, Israel
| | - Tommy Kaplan
- School of Computer Science and Engineering, The Hebrew University of Jerusalem, Jerusalem, Israel; Department of Developmental Biology and Cancer Research, Institute for Medical Research Israel-Canada, The Hebrew University-Hadassah Medical School, Jerusalem, Israel; The Center for Computational Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel.
| |
Collapse
|
44
|
Urban A, Sidorenko D, Zagirova D, Kozlova E, Kalashnikov A, Pushkov S, Naumov V, Sarkisova V, Leung GHD, Leung HW, Pun FW, Ozerov IV, Aliper A, Ren F, Zhavoronkov A. Precious1GPT: multimodal transformer-based transfer learning for aging clock development and feature importance analysis for aging and age-related disease target discovery. Aging (Albany NY) 2023; 15:4649-4666. [PMID: 37315204 PMCID: PMC10292881 DOI: 10.18632/aging.204788] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2023] [Accepted: 05/24/2023] [Indexed: 06/16/2023]
Abstract
Aging is a complex and multifactorial process that increases the risk of various age-related diseases and there are many aging clocks that can accurately predict chronological age, mortality, and health status. These clocks are disconnected and are rarely fit for therapeutic target discovery. In this study, we propose a novel approach to multimodal aging clock we call Precious1GPT utilizing methylation and transcriptomic data for interpretable age prediction and target discovery developed using a transformer-based model and transfer learning for case-control classification. While the accuracy of the multimodal transformer is lower within each individual data type compared to the state of art specialized aging clocks based on methylation or transcriptomic data separately it may have higher practical utility for target discovery. This method provides the ability to discover novel therapeutic targets that hypothetically may be able to reverse or accelerate biological age providing a pathway for therapeutic drug discovery and validation using the aging clock. In addition, we provide a list of promising targets annotated using the PandaOmics industrial target discovery platform.
Collapse
Affiliation(s)
- Anatoly Urban
- Insilico Medicine, Pak Shek Kok, New Territories, Hong Kong
| | | | - Diana Zagirova
- Insilico Medicine, Pak Shek Kok, New Territories, Hong Kong
| | | | | | - Stefan Pushkov
- Insilico Medicine, Pak Shek Kok, New Territories, Hong Kong
| | | | | | | | - Hoi Wing Leung
- Insilico Medicine, Pak Shek Kok, New Territories, Hong Kong
| | - Frank W. Pun
- Insilico Medicine, Pak Shek Kok, New Territories, Hong Kong
| | - Ivan V. Ozerov
- Insilico Medicine, Pak Shek Kok, New Territories, Hong Kong
| | - Alex Aliper
- Insilico Medicine, Pak Shek Kok, New Territories, Hong Kong
- Insilico Medicine, Masdar City, United Arab Emirates
| | - Feng Ren
- Insilico Medicine, Shanghai, China
| | - Alex Zhavoronkov
- Insilico Medicine, Pak Shek Kok, New Territories, Hong Kong
- Insilico Medicine, Masdar City, United Arab Emirates
| |
Collapse
|
45
|
Doherty T, Dempster E, Hannon E, Mill J, Poulton R, Corcoran D, Sugden K, Williams B, Caspi A, Moffitt TE, Delany SJ, Murphy TM. A comparison of feature selection methodologies and learning algorithms in the development of a DNA methylation-based telomere length estimator. BMC Bioinformatics 2023; 24:178. [PMID: 37127563 PMCID: PMC10152624 DOI: 10.1186/s12859-023-05282-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Accepted: 04/11/2023] [Indexed: 05/03/2023] Open
Abstract
BACKGROUND The field of epigenomics holds great promise in understanding and treating disease with advances in machine learning (ML) and artificial intelligence being vitally important in this pursuit. Increasingly, research now utilises DNA methylation measures at cytosine-guanine dinucleotides (CpG) to detect disease and estimate biological traits such as aging. Given the challenge of high dimensionality of DNA methylation data, feature-selection techniques are commonly employed to reduce dimensionality and identify the most important subset of features. In this study, our aim was to test and compare a range of feature-selection methods and ML algorithms in the development of a novel DNA methylation-based telomere length (TL) estimator. We utilised both nested cross-validation and two independent test sets for the comparisons. RESULTS We found that principal component analysis in advance of elastic net regression led to the overall best performing estimator when evaluated using a nested cross-validation analysis and two independent test cohorts. This approach achieved a correlation between estimated and actual TL of 0.295 (83.4% CI [0.201, 0.384]) on the EXTEND test data set. Contrastingly, the baseline model of elastic net regression with no prior feature reduction stage performed less well in general-suggesting a prior feature-selection stage may have important utility. A previously developed TL estimator, DNAmTL, achieved a correlation of 0.216 (83.4% CI [0.118, 0.310]) on the EXTEND data. Additionally, we observed that different DNA methylation-based TL estimators, which have few common CpGs, are associated with many of the same biological entities. CONCLUSIONS The variance in performance across tested approaches shows that estimators are sensitive to data set heterogeneity and the development of an optimal DNA methylation-based estimator should benefit from the robust methodological approach used in this study. Moreover, our methodology which utilises a range of feature-selection approaches and ML algorithms could be applied to other biological markers and disease phenotypes, to examine their relationship with DNA methylation and predictive value.
Collapse
Affiliation(s)
- Trevor Doherty
- School of Biological, Health and Sports Sciences, Technological University Dublin, Dublin, Ireland.
- SFI Centre for Research Training in Machine Learning, Technological University Dublin, Dublin, Ireland.
| | - Emma Dempster
- University of Exeter Medical School, University of Exeter, Exeter, UK
| | - Eilis Hannon
- University of Exeter Medical School, University of Exeter, Exeter, UK
| | - Jonathan Mill
- University of Exeter Medical School, University of Exeter, Exeter, UK
| | - Richie Poulton
- Department of Psychology, University of Otago, Dunedin, 9016, New Zealand
| | - David Corcoran
- Center for Genomic and Computational Biology, Duke University, Durham, NC, 27708, USA
| | - Karen Sugden
- Department of Psychology and Neuroscience, Duke University, Durham, NC, USA
| | - Ben Williams
- Department of Psychology and Neuroscience, Duke University, Durham, NC, USA
| | - Avshalom Caspi
- Social, Genetic and Developmental Psychiatry Centre, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
- Department of Psychology and Neuroscience, Duke University, Durham, NC, USA
| | - Terrie E Moffitt
- Social, Genetic and Developmental Psychiatry Centre, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
- Department of Psychology and Neuroscience, Duke University, Durham, NC, USA
| | - Sarah Jane Delany
- School of Computer Science, Technological University Dublin, Dublin, Ireland
| | - Therese M Murphy
- School of Biological, Health and Sports Sciences, Technological University Dublin, Dublin, Ireland
| |
Collapse
|
46
|
Martínez-Magaña JJ, Krystal JH, Girgenti MJ, Núnez-Ríos DL, Nagamatsu ST, Andrade-Brito DE, Montalvo-Ortiz JL. Decoding the role of transcriptomic clocks in the human prefrontal cortex. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2023:2023.04.19.23288765. [PMID: 37163025 PMCID: PMC10168432 DOI: 10.1101/2023.04.19.23288765] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/11/2023]
Abstract
Aging is a complex process with interindividual variability, which can be measured by aging biological clocks. Aging clocks are machine-learning algorithms guided by biological information and associated with mortality risk and a wide range of health outcomes. One of these aging clocks are transcriptomic clocks, which uses gene expression data to predict biological age; however, their functional role is unknown. Here, we profiled two transcriptomic clocks (RNAAgeCalc and knowledge-based deep neural network clock) in a large dataset of human postmortem prefrontal cortex (PFC) samples. We identified that deep-learning transcriptomic clock outperforms RNAAgeCalc to predict transcriptomic age in the human PFC. We identified associations of transcriptomic clocks with psychiatric-related traits. Further, we applied system biology algorithms to identify common gene networks among both clocks and performed pathways enrichment analyses to assess its functionality and prioritize genes involved in the aging processes. Identified gene networks showed enrichment for diseases of signal transduction by growth factor receptors and second messenger pathways. We also observed enrichment of genome-wide signals of mental and physical health outcomes and identified genes previously associated with human brain aging. Our findings suggest a link between transcriptomic aging and health disorders, including psychiatric traits. Further, it reveals functional genes within the human PFC that may play an important role in aging and health risk.
Collapse
Affiliation(s)
- José J. Martínez-Magaña
- Division of Human Genetics, Department of Psychiatry, Yale University School of Medicine, New Haven
- National Center for PTSD, US Department of Veterans Affairs, West Haven, CT, USA
| | - John H. Krystal
- Division of Human Genetics, Department of Psychiatry, Yale University School of Medicine, New Haven
- National Center for PTSD, US Department of Veterans Affairs, West Haven, CT, USA
- Psychiatry Service, VA Connecticut Health Care System, West Haven, CT, USA
| | - Matthew J. Girgenti
- Division of Human Genetics, Department of Psychiatry, Yale University School of Medicine, New Haven
- National Center for PTSD, US Department of Veterans Affairs, West Haven, CT, USA
| | - Diana L. Núnez-Ríos
- Division of Human Genetics, Department of Psychiatry, Yale University School of Medicine, New Haven
- National Center for PTSD, US Department of Veterans Affairs, West Haven, CT, USA
| | - Sheila T. Nagamatsu
- Division of Human Genetics, Department of Psychiatry, Yale University School of Medicine, New Haven
- National Center for PTSD, US Department of Veterans Affairs, West Haven, CT, USA
| | - Diego E. Andrade-Brito
- Division of Human Genetics, Department of Psychiatry, Yale University School of Medicine, New Haven
- National Center for PTSD, US Department of Veterans Affairs, West Haven, CT, USA
| | | | - Janitza L. Montalvo-Ortiz
- Division of Human Genetics, Department of Psychiatry, Yale University School of Medicine, New Haven
- National Center for PTSD, US Department of Veterans Affairs, West Haven, CT, USA
- Psychiatry Service, VA Connecticut Health Care System, West Haven, CT, USA
| |
Collapse
|
47
|
Silveira PP, Meaney MJ. Examining the biological mechanisms of human mental disorders resulting from gene-environment interdependence using novel functional genomic approaches. Neurobiol Dis 2023; 178:106008. [PMID: 36690304 DOI: 10.1016/j.nbd.2023.106008] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Revised: 12/30/2022] [Accepted: 01/18/2023] [Indexed: 01/21/2023] Open
Abstract
We explore how functional genomics approaches that integrate datasets from human and non-human model systems can improve our understanding of the effect of gene-environment interplay on the risk for mental disorders. We start by briefly defining the G-E paradigm and its challenges and then discuss the different levels of regulation of gene expression and the corresponding data existing in humans (genome wide genotyping, transcriptomics, DNA methylation, chromatin modifications, chromosome conformational changes, non-coding RNAs, proteomics and metabolomics), discussing novel approaches to the application of these data in the study of the origins of mental health. Finally, we discuss the multilevel integration of diverse types of data. Advance in the use of functional genomics in the context of a G-E perspective improves the detection of vulnerabilities, informing the development of preventive and therapeutic interventions.
Collapse
Affiliation(s)
- Patrícia Pelufo Silveira
- Department of Psychiatry, Faculty of Medicine and Health Sciences, McGill University, Montreal, QC, Canada; Ludmer Centre for Neuroinformatics and Mental Health, Douglas Mental Health University Institute, McGill University, Montreal, QC, Canada.
| | - Michael J Meaney
- Department of Psychiatry, Faculty of Medicine and Health Sciences, McGill University, Montreal, QC, Canada; Translational Neuroscience Program, Singapore Institute for Clinical Sciences, Agency for Science, Technology and Research (ASTAR), Singapore; Brain - Body Initiative, Agency for Science, Technology and Research (ASTAR), Singapore.
| |
Collapse
|
48
|
Bernabeu E, McCartney DL, Gadd DA, Hillary RF, Lu AT, Murphy L, Wrobel N, Campbell A, Harris SE, Liewald D, Hayward C, Sudlow C, Cox SR, Evans KL, Horvath S, McIntosh AM, Robinson MR, Vallejos CA, Marioni RE. Refining epigenetic prediction of chronological and biological age. Genome Med 2023; 15:12. [PMID: 36855161 PMCID: PMC9976489 DOI: 10.1186/s13073-023-01161-y] [Citation(s) in RCA: 47] [Impact Index Per Article: 23.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2022] [Accepted: 02/06/2023] [Indexed: 03/02/2023] Open
Abstract
BACKGROUND Epigenetic clocks can track both chronological age (cAge) and biological age (bAge). The latter is typically defined by physiological biomarkers and risk of adverse health outcomes, including all-cause mortality. As cohort sample sizes increase, estimates of cAge and bAge become more precise. Here, we aim to develop accurate epigenetic predictors of cAge and bAge, whilst improving our understanding of their epigenomic architecture. METHODS First, we perform large-scale (N = 18,413) epigenome-wide association studies (EWAS) of chronological age and all-cause mortality. Next, to create a cAge predictor, we use methylation data from 24,674 participants from the Generation Scotland study, the Lothian Birth Cohorts (LBC) of 1921 and 1936, and 8 other cohorts with publicly available data. In addition, we train a predictor of time to all-cause mortality as a proxy for bAge using the Generation Scotland cohort (1214 observed deaths). For this purpose, we use epigenetic surrogates (EpiScores) for 109 plasma proteins and the 8 component parts of GrimAge, one of the current best epigenetic predictors of survival. We test this bAge predictor in four external cohorts (LBC1921, LBC1936, the Framingham Heart Study and the Women's Health Initiative study). RESULTS Through the inclusion of linear and non-linear age-CpG associations from the EWAS, feature pre-selection in advance of elastic net regression, and a leave-one-cohort-out (LOCO) cross-validation framework, we obtain cAge prediction with a median absolute error equal to 2.3 years. Our bAge predictor was found to slightly outperform GrimAge in terms of the strength of its association to survival (HRGrimAge = 1.47 [1.40, 1.54] with p = 1.08 × 10-52, and HRbAge = 1.52 [1.44, 1.59] with p = 2.20 × 10-60). Finally, we introduce MethylBrowsR, an online tool to visualise epigenome-wide CpG-age associations. CONCLUSIONS The integration of multiple large datasets, EpiScores, non-linear DNAm effects, and new approaches to feature selection has facilitated improvements to the blood-based epigenetic prediction of biological and chronological age.
Collapse
Affiliation(s)
- Elena Bernabeu
- Centre for Genomic and Experimental Medicine, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, UK
| | - Daniel L McCartney
- Centre for Genomic and Experimental Medicine, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, UK
| | - Danni A Gadd
- Centre for Genomic and Experimental Medicine, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, UK
| | - Robert F Hillary
- Centre for Genomic and Experimental Medicine, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, UK
| | - Ake T Lu
- Department of Human Genetics, David Geffen School of Medicine, University of California, Los Angeles, CA, USA
- Altos Labs, San Diego, USA
| | - Lee Murphy
- Edinburgh Clinical Research Facility, University of Edinburgh, Edinburgh, UK
| | - Nicola Wrobel
- Edinburgh Clinical Research Facility, University of Edinburgh, Edinburgh, UK
| | - Archie Campbell
- Centre for Genomic and Experimental Medicine, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, UK
| | - Sarah E Harris
- Department of Psychology, Lothian Birth Cohorts, University of Edinburgh, Edinburgh, UK
| | - David Liewald
- Department of Psychology, Lothian Birth Cohorts, University of Edinburgh, Edinburgh, UK
| | - Caroline Hayward
- Centre for Genomic and Experimental Medicine, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, UK
- Medical Research Council Human Genetics Unit, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, UK
| | - Cathie Sudlow
- Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, UK
- BHF Data Science Centre, Health Data Research UK, London, UK
- Edinburgh Medical School, Usher Institute, University of Edinburgh, Edinburgh, UK
| | - Simon R Cox
- Department of Psychology, Lothian Birth Cohorts, University of Edinburgh, Edinburgh, UK
| | - Kathryn L Evans
- Centre for Genomic and Experimental Medicine, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, UK
| | - Steve Horvath
- Department of Human Genetics, David Geffen School of Medicine, University of California, Los Angeles, CA, USA
- Altos Labs, San Diego, USA
| | - Andrew M McIntosh
- Centre for Genomic and Experimental Medicine, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, UK
- Division of Psychiatry, University of Edinburgh, Royal Edinburgh Hospital, Edinburgh, UK
| | | | - Catalina A Vallejos
- Medical Research Council Human Genetics Unit, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, UK
- The Alan Turing Institute, London, UK
| | - Riccardo E Marioni
- Centre for Genomic and Experimental Medicine, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, UK.
| |
Collapse
|
49
|
Sugino RP, Ohira M, Mansai SP, Kamijo T. Comparative epigenomics by machine learning approach for neuroblastoma. BMC Genomics 2022; 23:852. [PMID: 36572864 PMCID: PMC9793522 DOI: 10.1186/s12864-022-09061-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Accepted: 12/02/2022] [Indexed: 12/28/2022] Open
Abstract
BACKGROUND Neuroblastoma (NB) is the second most common pediatric solid tumor. Because the number of genetic mutations found in tumors are small, even in some patients with unfavorable NB, epigenetic variation is expected to play an important role in NB progression. DNA methylation is a major epigenetic mechanism, and its relationship with NB prognosis has been a concern. One limitation with the analysis of variation in DNA methylation is the lack of a suitable analytical model. Therefore, in this study, we performed a random forest (RF) analysis of the DNA methylome data of NB from multiple databases. RESULTS RF is a popular machine learning model owing to its simplicity, intuitiveness, and computational cost. RF analysis identified novel intermediate-risk patient groups with characteristic DNA methylation patterns within the low-risk group. Feature selection analysis based on probe annotation revealed that enhancer-annotated regions had strong predictive power, particularly for MYCN-amplified NBs. We developed a gene-based analytical model to identify candidate genes related to disease progression, such as PRDM8 and FAM13A-AS1. RF analysis revealed sufficient predictive power compared to other machine learning models. CONCLUSIONS RF is a useful tool for DNA methylome analysis in cancer epigenetic studies, and has potential to identify a novel cancer-related genes.
Collapse
Affiliation(s)
- Ryuichi P. Sugino
- grid.416695.90000 0000 8855 274XResearch Institute for Clinical Oncology, Saitama Cancer Center, Ina, Saitama, 362-0806 Japan
| | - Miki Ohira
- grid.416695.90000 0000 8855 274XResearch Institute for Clinical Oncology, Saitama Cancer Center, Ina, Saitama, 362-0806 Japan
| | - Sayaka P. Mansai
- grid.416695.90000 0000 8855 274XResearch Institute for Clinical Oncology, Saitama Cancer Center, Ina, Saitama, 362-0806 Japan
| | - Takehiko Kamijo
- grid.416695.90000 0000 8855 274XResearch Institute for Clinical Oncology, Saitama Cancer Center, Ina, Saitama, 362-0806 Japan ,grid.263023.60000 0001 0703 3735Laboratory of Tumor Molecular Biology, Department of Graduate School of Science and Engineering, Saitama University, Kita-Urawa, Saitama, Japan
| |
Collapse
|
50
|
Deryabin PI, Borodkina AV. Epigenetic clocks provide clues to the mystery of uterine ageing. Hum Reprod Update 2022; 29:259-271. [PMID: 36515535 DOI: 10.1093/humupd/dmac042] [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: 08/12/2022] [Revised: 11/28/2022] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Rising maternal ages and age-related fertility decline are a global challenge for modern reproductive medicine. Clinicians and researchers pay specific attention to ovarian ageing and hormonal insufficiency in this regard. However, uterine ageing is often left out of the picture, with the majority of reproductive clinicians being close to unanimous on the absence of age-related functional decline in the uterine tissues. Therefore, most existing techniques to treat an age-related decline in implantation rates are based primarily on hormonal supplementation and oocyte donation. Solving the issue of uterine ageing might lead to an adjustment to these methods. OBJECTIVE AND RATIONALE A focus on uterine ageing and the possibility of slowing it emerged with the development of the information theory of ageing, which identifies genomic instability and erosion of the epigenetic landscape as important drivers of age-related decline in the functionality of most cells and tissues. Age-related smoothing of this landscape and a decline in tissue function can be assessed by measuring the ticking of epigenetic clocks. Within this review, we explore whether the uterus experiences age-related alterations using this elegant approach. We analyse existing data on epigenetic clocks in the endometrium, highlight approaches to improve the accuracy of the clocks in this cycling tissue, speculate on the endometrial pathologies whose progression might be predicted by the altered speed of epigenetic clocks and discuss the possibilities of slowing down the ticking of these clocks. SEARCH METHODS Data for this review were identified by searches of Medline, PubMed and Google Scholar. References from relevant articles using the search terms 'ageing', 'maternal age', 'female reproduction', 'uterus', 'endometrium', 'implantation', 'decidualization', 'epigenetic clock', 'biological age', 'DNA methylation', 'fertility' and 'infertility' were selected. A total of 95 articles published in English between 1985 and 2022 were included, six of which describe the use of the epigenetic clock to evaluate uterine/endometrium ageing. OUTCOMES Application of the Horvath and DNAm PhenoAge epigenetic clocks demonstrated a poor correlation with chronological age in the endometrium. Several approaches were suggested to enhance the predictive power of epigenetic clocks for the endometrium. The first was to increase the number of samples in the training dataset, as for the Zang clock, or to use more sophisticated clock-building algorithms, as for the AltumAge clock. The second method is to adjust the clocks according to the dynamic nature of the endometrium. Using either approach revealed a strong correlation with chronological age in the endometrium, providing solid evidence for age-related functional decline in this tissue. Furthermore, age acceleration/deceleration, as estimated by epigenetic clocks, might be a promising tool to predict or to gain insights into the origin of various endometrial pathologies, including recurrent implantation failure, cancer and endometriosis. Finally, there are several strategies to slow down or even reverse epigenetic clocks that might be applied to reduce the risk of age-related uterine impairments. WIDER IMPLICATIONS The uterine factor should be considered, along with ovarian issues, to correct for the decline in female fertility with age. Epigenetic clocks can be tested to gain a deeper understanding of various endometrial disorders.
Collapse
Affiliation(s)
- Pavel I Deryabin
- Mechanisms of Cellular Senescence Group, Institute of Cytology of the Russian Academy of Sciences, Saint-Petersburg, Russia
| | - Aleksandra V Borodkina
- Mechanisms of Cellular Senescence Group, Institute of Cytology of the Russian Academy of Sciences, Saint-Petersburg, Russia
| |
Collapse
|