1
|
Liu C, Dai Y, Li X, Xu T, Li J, Zhao G, Liu S, Li B. A novel metabolomic aging score - better than conventional metrics in predicting short-term mortality. Expert Rev Mol Diagn 2025:1-12. [PMID: 40394731 DOI: 10.1080/14737159.2025.2509027] [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: 03/01/2025] [Revised: 04/27/2025] [Accepted: 05/16/2025] [Indexed: 05/22/2025]
Abstract
INTRODUCTION Accurate prediction of short-term mortality is crucial for optimizing clinical prognosis and providing treatment decisions. Conventional metrics, including physiological indicators, laboratory indexes and scoring systems, suffer from limitations in comprehensiveness, accuracy, and dynamism. In contrast, the metabolomic aging score, as an emerging biomarker, offers substantial promise in short-term mortality prediction. AREAS COVERED By integrating multiple metabolites associated with aging and mortality, the score captures dynamic metabolic shifts, providing a real-time reflection of an individual's health status. This approach enables a more precise assessment of short-term mortality risk across diverse diseases, setting it apart from traditional, disease-specific biomarkers. In addition, the metabolic aging score also shows great application prospects in identifying susceptible populations and providing individualized precision medication. This article discusses the novel role of the metabolomic aging score in mortality prediction, highlighting its superior accuracy compared to conventional metrics. EXPERT OPINION This score has broad application prospects in the future and also faces challenges such as large-scale validation and standardization. Furthermore, the integration of artificial intelligence (AI) is poised to enhance the clinical utility of the metabolomic aging score, advancing its potential to transform healthcare practices.
Collapse
Affiliation(s)
- Chong Liu
- Bioinformatics Centre, National Clinical Research Centre for Geriatric Disorders, Department of Geriatrics, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Yinghong Dai
- Bioinformatics Centre, National Clinical Research Centre for Geriatric Disorders, Department of Geriatrics, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China
| | - Xinxue Li
- Bioinformatics Centre, National Clinical Research Centre for Geriatric Disorders, Department of Geriatrics, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Tiantian Xu
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Jinchen Li
- Bioinformatics Centre, National Clinical Research Centre for Geriatric Disorders, Department of Geriatrics, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Bioinformatics Centre, Furong Laboratory, Changsha, Hunan, China
| | - Guihu Zhao
- Bioinformatics Centre, National Clinical Research Centre for Geriatric Disorders, Department of Geriatrics, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Sijia Liu
- Department of Rheumatology and Immunology, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Bin Li
- Bioinformatics Centre, National Clinical Research Centre for Geriatric Disorders, Department of Geriatrics, Xiangya Hospital, Central South University, Changsha, Hunan, China
| |
Collapse
|
2
|
Kuiper L, Helthuis R, Polinder-Bos H, Lemmens L, Dollé M, Slagboom E, van Rooij J, Verschuren M, van Meurs J. Exploring geriatricians' perspectives on aging biomarkers: A reflexive thematic analysis. Maturitas 2025; 198:108601. [PMID: 40393194 DOI: 10.1016/j.maturitas.2025.108601] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2025] [Revised: 05/07/2025] [Accepted: 05/13/2025] [Indexed: 05/22/2025]
Abstract
OBJECTIVES Aging biomarkers have been developed to identify people at higher risk of age-related decline. The objective of this study is to understand geriatricians' perspectives on aging biomarkers in order to facilitate their integration into geriatric medicine. STUDY DESIGN Using reflexive thematic analysis, this qualitative study explores the views of geriatricians on the potential role of aging biomarkers in clinical practice based on thirteen semi-structured interviews. MAIN OUTCOME MEASURES Geriatricians' views on the role, utility, and challenges of the use of aging biomarkers in geriatric medicine. RESULTS Two main themes were developed: the complexity of geriatric medicine and the importance of trust in biomarkers. Clinicians highlighted the heterogeneity of the older patient population, noting that current assessments, such as the Comprehensive Geriatric Assessment (CGA), rely on the geriatricians' clinical judgment. While most participants saw potential for aging biomarkers to supplement the CGA in assessment of patients' resilience in recovery from invasive treatment, they emphasized the need to prove value beyond current treatment decisions. Furthermore, participants stressed the need for actionable, reliable, and context-specific tools. Concerns included the risk of oversimplifying the assessment of resilience, lack of applicability to the frail clinical population, and the ethical implications for both health care and society more broadly in the implementation of aging biomarkers. CONCLUSIONS This study emphasizes the importance of aligning biomarker development with the reality of geriatric medicine and clinicians' needs. Efforts from geriatricians, aging biomarker researchers, ethicists, and primary treating physicians are needed to successfully adopt aging biomarkers into geriatric medicine.
Collapse
Affiliation(s)
- Lieke Kuiper
- Department of Internal Medicine, Erasmus University Medical Center, Rotterdam, the Netherlands; Center for Prevention, Lifestyle and Health, National Institute for Public Health and Environment (RIVM), Bilthoven, the Netherlands.
| | - Roy Helthuis
- Center for Prevention, Lifestyle and Health, National Institute for Public Health and Environment (RIVM), Bilthoven, the Netherlands
| | - Harmke Polinder-Bos
- Department of Internal Medicine, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Lidwien Lemmens
- Centre for Nutrition, Prevention and Health Services, National Institute for Public Health and Environment (RIVM), Bilthoven, the Netherlands
| | - Martijn Dollé
- Center for Health Protection, National Institute for Public Health and Environment (RIVM), Bilthoven, the Netherlands
| | - Eline Slagboom
- Molecular Epidemiology, Department of Biomedical Data Sciences, Leiden University Medical Centre, Leiden, the Netherlands; Max Planck Institute for the Biology of Ageing, Cologne, Germany
| | - Jeroen van Rooij
- Department of Internal Medicine, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Monique Verschuren
- Center for Prevention, Lifestyle and Health, National Institute for Public Health and Environment (RIVM), Bilthoven, the Netherlands; Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| | - Joyce van Meurs
- Department of Internal Medicine, Erasmus University Medical Center, Rotterdam, the Netherlands; Department of Orthopaedics & Sports, Erasmus Medical Center, Rotterdam, the Netherlands
| |
Collapse
|
3
|
Drouard G, Suhonen S, Heikkinen A, Wang Z, Kaprio J, Ollikainen M. Multi-Omic Associations of Epigenetic Age Acceleration Are Heterogeneously Shaped by Genetic and Environmental Influences. Aging Cell 2025:e70088. [PMID: 40325911 DOI: 10.1111/acel.70088] [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: 08/07/2024] [Revised: 04/03/2025] [Accepted: 04/13/2025] [Indexed: 05/07/2025] Open
Abstract
Connections between the multi-ome and epigenetic age acceleration (EAA), and especially whether these are influenced by genetic or environmental factors, remain underexplored. We therefore quantified associations between the multi-ome comprising four layers-the proteome, metabolome, external exposome (here, sociodemographic factors), and specific exposome (here, lifestyle)-with six different EAA estimates. Two twin cohorts were used in a discovery-replication scheme, comprising, respectively, young (N = 642; mean age = 22.3) and older (N = 354; mean age = 62.3) twins. Within-pair twin designs were used to assess genetic and environmental effects on associations. We identified 40 multi-omic factors, of which 28 were proteins, associated with EAA in the young twins while adjusting for sex, smoking, and body mass index. Within-pair analyses revealed that genetic confounding influenced these associations heterogeneously, with six multi-omic factors -matrix metalloproteinase 9, complement component C6, histidine, glycoprotein acetyls, lactate, and neighborhood percentage of nonagenarians- remaining significantly associated with EAA, independent of genetic effects. Replication analyses showed that some associations assessed in young twins were consistent in older twins. Our study highlights the differential influence of genetic effects on the associations between the multi-ome and EAA and shows that some, but not all, of the associations persist into adulthood.
Collapse
Affiliation(s)
- Gabin Drouard
- Institute for Molecular Medicine Finland (FIMM), HiLIFE, University of Helsinki, Helsinki, Finland
| | - Sannimari Suhonen
- Institute for Molecular Medicine Finland (FIMM), HiLIFE, University of Helsinki, Helsinki, Finland
| | - Aino Heikkinen
- Institute for Molecular Medicine Finland (FIMM), HiLIFE, University of Helsinki, Helsinki, Finland
- Minerva Foundation Institute for Medical Research, Helsinki, Finland
| | - Zhiyang Wang
- Institute for Molecular Medicine Finland (FIMM), HiLIFE, University of Helsinki, Helsinki, Finland
| | - Jaakko Kaprio
- Institute for Molecular Medicine Finland (FIMM), HiLIFE, University of Helsinki, Helsinki, Finland
| | - Miina Ollikainen
- Institute for Molecular Medicine Finland (FIMM), HiLIFE, University of Helsinki, Helsinki, Finland
- Minerva Foundation Institute for Medical Research, Helsinki, Finland
| |
Collapse
|
4
|
Kuiper LM, Picavet HSJ, Rietman ML, Dollé MET, Verschuren WMM. Advanced Glycation End-Products and Metabolomics Are Independently Associated With Frailty: The Longitudinal Doetinchem Cohort Study. J Gerontol A Biol Sci Med Sci 2025; 80:glae272. [PMID: 39607723 PMCID: PMC12086670 DOI: 10.1093/gerona/glae272] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2024] [Indexed: 11/29/2024] Open
Abstract
Skin autofluorescence (SAF), reflecting advanced glycation endproducts' accumulation in tissue, has been proposed as a noninvasive aging biomarker. Yet, SAF has not been compared with well-established blood-based aging biomarkers such as MetaboHealth in association with frailty. Furthermore, no previous study determined the longitudinal association of SAF with frailty. We used 2 382 Doetinchem Cohort Study participants' (aged 46.0-85.4) cross-sectional data, of whom 1 654 had longitudinal SAF measurements. SAF was measured using the AGE Reader. MetaboHealth was calculated on 1H-NMR-metabolomics. Linear regressions were used for the associations of SAF and MetaboHealth on the 36-deficit frailty index and logistic regressions for being pre-frail or frail as determined by the frailty phenotype. Longitudinal associations were determined by an interaction term between age and SAF in a linear mixed model. SAF and MetaboHealth were associated with higher odds of pre-frailty (odd ratios per standard deviation SAF: 1.21 [1.10-1.32], MetaboHealth: 1.35 [1.24-1.49]) and frailty (SAF: 1.70 [1.41-2.06], MetaboHealth: 1.90 [1.57-2.32]). When mutually adjusted, both aging biomarkers remained associated with pre-frailty (SAF: 1.16 [1.05-1.27], MetaboHealth 1.33 [1.21-1.46]) and frailty (SAF: 1.52 [1.25-1.85], MetaboHealth: 1.75 [1.43-2.14]). Additionally, SAF and MetaboHealth were associated with higher frailty index scores (percentage increase per standard deviation SAF: 1.35 [1.00-1.70], MetaboHealth: 1.87 [1.54-2.20]), also after mutual adjustment (SAF: 1.02 [0.68-1.37], MetaboHealth: 1.69 [1.35-2.02]). SAF was also longitudinally associated with the frailty index (percentage per unit/year increase: 0.12 [0.07-0.16]). The mutual independence of SAF and MetaboHealth implies they capture distinct aspects of the aging process. Altogether, these findings emphasize SAF's clinical potential as an age-related decline biomarker, which could be further enhanced when combined with MetaboHealth.
Collapse
Affiliation(s)
- Lieke M Kuiper
- Center for Prevention, Lifestyle and Health, National Institute for Public Health and Environment (RIVM), Bilthoven, the Netherlands
- Department of Internal Medicine, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - H Susan J Picavet
- Center for Prevention, Lifestyle and Health, National Institute for Public Health and Environment (RIVM), Bilthoven, the Netherlands
| | - M Liset Rietman
- Center for Prevention, Lifestyle and Health, National Institute for Public Health and Environment (RIVM), Bilthoven, the Netherlands
| | - Martijn E T Dollé
- Center for Health Protection, National Institute for Public Health and Environment (RIVM), Bilthoven, the Netherlands
| | - W M Monique Verschuren
- Center for Prevention, Lifestyle and Health, National Institute for Public Health and Environment (RIVM), Bilthoven, the Netherlands
- Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| |
Collapse
|
5
|
Thorp EB, Ananthakrishnan A, Lantz CW. Decoding immune cell interactions during cardiac allograft vasculopathy: insights derived from bioinformatic strategies. Front Cardiovasc Med 2025; 12:1568528. [PMID: 40342971 PMCID: PMC12058854 DOI: 10.3389/fcvm.2025.1568528] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2025] [Accepted: 04/04/2025] [Indexed: 05/11/2025] Open
Abstract
Chronic allograft vasculopathy (CAV) is a major cause of late graft failure in heart transplant recipients, characterized by progressive intimal thickening and diffuse narrowing of the coronary arteries. Unlike atherosclerosis, CAV exhibits a distinct cellular composition and lesion distribution, yet its pathogenesis remains incompletely understood. A major challenge in CAV research has been the limited application of advanced "-omics" technologies, which have revolutionized the study of other vascular diseases. Recent advancements in single-cell and spatial transcriptomics, proteomics, and metabolomics have begun to uncover the complex immune-endothelial-stromal interactions driving CAV progression. Notably, single-cell RNA sequencing has identified previously unrecognized immune cell populations and signaling pathways implicated in endothelial injury and vascular remodeling after heart transplantation. Despite these breakthroughs, studies applying these technologies to CAV remain sparse, limiting the translation of these insights into clinical practice. This review aims to bridge this gap by summarizing recent findings from single-cell and multi-omic approaches, highlighting key discoveries, and discussing their implications for understanding CAV pathogenesis.
Collapse
Affiliation(s)
- Edward B. Thorp
- Department of Pathology, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
| | - Aparnaa Ananthakrishnan
- Department of Pathology, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
| | - Connor W. Lantz
- Department of Surgery, Comprehensive Transplant Center, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
| |
Collapse
|
6
|
Lazareva O, Riediger A, Stegle O, Sültmann H, Hohenfellner M, Görtz M. Integrative analysis of blood biomarkers and clinical variables improves early detection of aggressive prostate cancer. Sci Rep 2025; 15:14071. [PMID: 40269068 PMCID: PMC12018954 DOI: 10.1038/s41598-025-98980-3] [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: 12/12/2024] [Accepted: 04/16/2025] [Indexed: 04/25/2025] Open
Abstract
Aggressive prostate cancer (PC) represents a significant health concern. Conventional screening methods, primarily based on prostate-specific antigen (PSA) levels, lack specificity, leading to an urgent need for more accurate diagnostic tools. This study investigates whether integrating clinical and routine blood laboratory parameters can improve the early non-invasive prediction of aggressive PC. In a pilot study of 578 patients with suspicion of PC, 28 laboratory values alongside data on family history, diet, and lifestyle were analyzed. A logistic regression classifier was developed, with model performance evaluated using repeated k-fold cross-validation on the complete dataset (n = 282). Participants were categorized into healthy, moderate PC (ISUP 1-2), and aggressive PC (ISUP 3-5). Significant associations were found between PC aggressiveness and lower levels of androstenedione, dehydroepiandrosterone-sulfate (DHEA-S) and free PSA%, as well as higher levels of sex hormone binding globulin (SHBG). The integration of these serum markers with clinical parameters into a new multi-stage risk classifier significantly improved the predictive accuracy for aggressive PC, outperforming PSA-only methods. The integration of DHEA-S, androstenedione, and SHBG as widely available and cost-effective novel blood biomarkers offers a more targeted, non-invasive prediction of aggressive PC. This approach could reduce reliance on invasive prostate biopsies and expensive magnetic resonance imaging.
Collapse
Affiliation(s)
- Olga Lazareva
- Junior Clinical Cooperation Unit 'Multiparametric Methods for Early Detection of Prostate Cancer', German Cancer Research Center (DKFZ), Heidelberg, Germany
- Division of Computational Genomics and Systems Genetics, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Genome Biology Unit, European Molecular Biology Laboratory, Heidelberg, Germany
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, UK
| | - Anja Riediger
- Junior Clinical Cooperation Unit 'Multiparametric Methods for Early Detection of Prostate Cancer', German Cancer Research Center (DKFZ), Heidelberg, Germany
- Department of Urology, University Hospital Heidelberg, Im Neuenheimer Feld 420, 69120, Heidelberg, Germany
- National Center for Tumor Diseases (NCT), Heidelberg, Germany
- Faculty of Biosciences, Heidelberg University, Heidelberg, Germany
| | - Oliver Stegle
- Division of Computational Genomics and Systems Genetics, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Genome Biology Unit, European Molecular Biology Laboratory, Heidelberg, Germany
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, UK
| | - Holger Sültmann
- National Center for Tumor Diseases (NCT), Heidelberg, Germany
- Division of Cancer Genome Research, German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Markus Hohenfellner
- Department of Urology, University Hospital Heidelberg, Im Neuenheimer Feld 420, 69120, Heidelberg, Germany
| | - Magdalena Görtz
- Junior Clinical Cooperation Unit 'Multiparametric Methods for Early Detection of Prostate Cancer', German Cancer Research Center (DKFZ), Heidelberg, Germany.
- Department of Urology, University Hospital Heidelberg, Im Neuenheimer Feld 420, 69120, Heidelberg, Germany.
- National Center for Tumor Diseases (NCT), Heidelberg, Germany.
| |
Collapse
|
7
|
Liu R, Yang S, Zhong X, Zhu Z, Huang W, Wang W. Metabolomic signature of retinal ageing, polygenetic susceptibility, and major health outcomes. Br J Ophthalmol 2025; 109:619-627. [PMID: 39581638 DOI: 10.1136/bjo-2024-325846] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2024] [Accepted: 10/28/2024] [Indexed: 11/26/2024]
Abstract
BACKGROUND/AIMS To identify the metabolic underpinnings of retinal aging and examine how it is related to mortality and morbidity of common diseases. METHODS The retinal age gap has been established as essential aging indicator for mortality and systemic health. We applied neural network to train the retinal age gap among the participants in UK Biobank and used nuclear magnetic resonance (NMR) to profile plasma metabolites. The metabolomic signature of retinal ageing (MSRA) was identified using an elastic network model. Multivariable Cox regressions were used to assess associations between the signature with 12 serious health conditions. The participants in Guangzhou Diabetic Eye Study (GDES) cohort were analyzed for validation. RESULTS This study included 110 722 participants (mean age 56.5±8.1 years at baseline, 53.8% female), and 28 plasma metabolites associated with retinal ageing were identified. The MSRA revealed significant correlations with each 12 serious health conditions beyond traditional risk factors and genetic predispositions. Each SD increase in MSRA was linked to a 24%-76% higher risk of mortality, cardiovascular diseases, dementia and diabetes mellitus. MSRA showed dose-response relationships with risks of these diseases, with seven showing non-linear and five showing linear increases. Validation in the GDES further established the relation between retinal ageing-related metabolites and increased risks of cardiovascular and chronic kidney diseases (all p<0.05). CONCLUSIONS The metabolic connections between ocular and systemic health offer a novel tool for identifying individuals at high risk of premature ageing, promoting a more holistic view of human health.
Collapse
Affiliation(s)
- Riqian Liu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangdong Provincial Clinical Study Center for Ocular Diseases, Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Shaopeng Yang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangdong Provincial Clinical Study Center for Ocular Diseases, Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Xiaoying Zhong
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangdong Provincial Clinical Study Center for Ocular Diseases, Sun Yat-Sen University, Guangzhou, Guangdong, China
- Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Ziyu Zhu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangdong Provincial Clinical Study Center for Ocular Diseases, Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Wenyong Huang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangdong Provincial Clinical Study Center for Ocular Diseases, Sun Yat-Sen University, Guangzhou, Guangdong, China
- Hainan Eye Hospital and Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Haikou, China
| | - Wei Wang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangdong Provincial Clinical Study Center for Ocular Diseases, Sun Yat-Sen University, Guangzhou, Guangdong, China
- Hainan Eye Hospital and Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Haikou, China
| |
Collapse
|
8
|
Rouskas K, Bocher O, Simistiras A, Emmanouil C, Mantas P, Skoulakis A, Park YC, Dimopoulos A, Glentis S, Kastenmüller G, Zeggini E, Dimas AS. Periodic dietary restriction of animal products induces metabolic reprogramming in humans with effects on cardiometabolic health. NPJ METABOLIC HEALTH AND DISEASE 2025; 3:14. [PMID: 40225784 PMCID: PMC11981922 DOI: 10.1038/s44324-025-00057-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/26/2024] [Accepted: 03/02/2025] [Indexed: 04/15/2025]
Abstract
Dietary interventions constitute powerful approaches for disease prevention and treatment. However, the molecular mechanisms through which diet affects health remain underexplored in humans. Here, we compare plasma metabolomic and proteomic profiles between dietary states for a unique group of individuals who alternate between omnivory and restriction of animal products for religious reasons. We find that short-term restriction drives reductions in levels of lipid classes and of branched-chain amino acids, not detected in a control group of individuals, and results in metabolic profiles associated with decreased risk for all-cause mortality. We show that 23% of proteins whose levels are affected by dietary restriction are druggable targets and reveal that pro-longevity hormone FGF21 and seven additional proteins (FOLR2, SUMF2, HAVCR1, PLA2G1B, OXT, SPP1, HPGDS) display the greatest magnitude of change. Through Mendelian randomization we demonstrate potentially causal effects of FGF21 and HAVCR1 on risk for type 2 diabetes, of HPGDS on BMI, and of OXT on risk for lacunar stroke. Collectively, we find that restriction-associated reprogramming improves metabolic health and emphasise high-value targets for pharmacological intervention.
Collapse
Affiliation(s)
- Konstantinos Rouskas
- Institute for Bioinnovation, Biomedical Sciences Research Center ‘Alexander Fleming’, Fleming 34, 16672 Vari, Greece
- Institute of Applied Biosciences, Centre for Research & Technology Hellas, Thessaloniki, Greece
| | - Ozvan Bocher
- Institute of Translational Genomics, Helmholtz Zentrum München – German Research Center for Environmental Health, Neuherberg, Germany
| | - Alexandros Simistiras
- Institute for Bioinnovation, Biomedical Sciences Research Center ‘Alexander Fleming’, Fleming 34, 16672 Vari, Greece
| | - Christina Emmanouil
- Institute for Bioinnovation, Biomedical Sciences Research Center ‘Alexander Fleming’, Fleming 34, 16672 Vari, Greece
| | - Panagiotis Mantas
- Institute for Bioinnovation, Biomedical Sciences Research Center ‘Alexander Fleming’, Fleming 34, 16672 Vari, Greece
| | - Anargyros Skoulakis
- Institute for Bioinnovation, Biomedical Sciences Research Center ‘Alexander Fleming’, Fleming 34, 16672 Vari, Greece
| | - Young-Chan Park
- Institute of Translational Genomics, Helmholtz Zentrum München – German Research Center for Environmental Health, Neuherberg, Germany
| | - Alexandros Dimopoulos
- Institute for Bioinnovation, Biomedical Sciences Research Center ‘Alexander Fleming’, Fleming 34, 16672 Vari, Greece
| | - Stavros Glentis
- Institute for Bioinnovation, Biomedical Sciences Research Center ‘Alexander Fleming’, Fleming 34, 16672 Vari, Greece
| | - Gabi Kastenmüller
- Institute of Computational Biology, Helmholtz Zentrum München – German Research Center for Environmental Health, Neuherberg, Germany
- German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - Eleftheria Zeggini
- Institute of Translational Genomics, Helmholtz Zentrum München – German Research Center for Environmental Health, Neuherberg, Germany
- Technical University of Munich (TUM) and Klinikum Rechts der Isar, TUM School of Medicine and Health, Munich, Germany
| | - Antigone S. Dimas
- Institute for Bioinnovation, Biomedical Sciences Research Center ‘Alexander Fleming’, Fleming 34, 16672 Vari, Greece
- Institute of Translational Genomics, Helmholtz Zentrum München – German Research Center for Environmental Health, Neuherberg, Germany
| |
Collapse
|
9
|
Menzaghi C, Copetti M, Mantzoros CS, Trischitta V. Prediction models for the implementation of precision medicine in the real world. Some critical issues. Metabolism 2025:156257. [PMID: 40187402 DOI: 10.1016/j.metabol.2025.156257] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/31/2025] [Accepted: 03/31/2025] [Indexed: 04/07/2025]
Affiliation(s)
- Claudia Menzaghi
- Research Unit of Diabetes and Endocrine Diseases, Fondazione Istituto di Ricovero e Cura a Carattere Scientifico "Casa Sollievo della Sofferenza", 71013 San Giovanni Rotondo, Italy
| | - Massimiliano Copetti
- Unit of Biostatistics, Fondazione Istituto di Ricovero e Cura a Carattere Scientifico "Casa Sollievo della Sofferenza", 71013 San Giovanni Rotondo, Italy
| | - Christos S Mantzoros
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Vincenzo Trischitta
- Research Unit of Diabetes and Endocrine Diseases, Fondazione Istituto di Ricovero e Cura a Carattere Scientifico "Casa Sollievo della Sofferenza", 71013 San Giovanni Rotondo, Italy.
| |
Collapse
|
10
|
Sinke L, Beekman M, Raz Y, Gehrmann T, Moustakas I, Boulinguiez A, Lakenberg N, Suchiman E, Bogaards FA, Bizzarri D, van den Akker EB, Waldenberger M, Butler‐Browne G, Trollet C, de Groot CPGM, Heijmans BT, Slagboom PE. Tissue-specific methylomic responses to a lifestyle intervention in older adults associate with metabolic and physiological health improvements. Aging Cell 2025; 24:e14431. [PMID: 39618079 PMCID: PMC11984676 DOI: 10.1111/acel.14431] [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: 08/26/2024] [Revised: 10/24/2024] [Accepted: 11/14/2024] [Indexed: 04/12/2025] Open
Abstract
Across the lifespan, diet and physical activity profiles substantially influence immunometabolic health. DNA methylation, as a tissue-specific marker sensitive to behavioral change, may mediate these effects through modulation of transcription factor binding and subsequent gene expression. Despite this, few human studies have profiled DNA methylation and gene expression simultaneously in multiple tissues or examined how molecular levels react and interact in response to lifestyle changes. The Growing Old Together (GOTO) study is a 13-week lifestyle intervention in older adults, which imparted health benefits to participants. Here, we characterize the DNA methylation response to this intervention at over 750 thousand CpGs in muscle, adipose, and blood. Differentially methylated sites are enriched for active chromatin states, located close to relevant transcription factor binding sites, and associated with changing expression of insulin sensitivity genes and health parameters. In addition, measures of biological age are consistently reduced, with decreases in grimAge associated with observed health improvements. Taken together, our results identify responsive molecular markers and demonstrate their potential to measure progression and finetune treatment of age-related risks and diseases.
Collapse
Affiliation(s)
- Lucy Sinke
- Molecular Epidemiology, Department of Biomedical Data SciencesLeiden University Medical CentreLeidenThe Netherlands
| | - Marian Beekman
- Molecular Epidemiology, Department of Biomedical Data SciencesLeiden University Medical CentreLeidenThe Netherlands
| | - Yotam Raz
- Molecular Epidemiology, Department of Biomedical Data SciencesLeiden University Medical CentreLeidenThe Netherlands
| | - Thies Gehrmann
- Molecular Epidemiology, Department of Biomedical Data SciencesLeiden University Medical CentreLeidenThe Netherlands
- Department of Bioscience Engineering, Research Group Environmental Ecology and Applied MicrobiologyUniversity of AntwerpAntwerpBelgium
| | - Ioannis Moustakas
- Molecular Epidemiology, Department of Biomedical Data SciencesLeiden University Medical CentreLeidenThe Netherlands
- Sequencing Analysis Support Core, Department of Biomedical Data SciencesLeiden University Medical CenterLeidenThe Netherlands
| | - Alexis Boulinguiez
- Myology Center for Research, U974Sorbonne Université, INSERM, AIM, GH Pitié Salpêtrière Bat BabinskiParisFrance
| | - Nico Lakenberg
- Molecular Epidemiology, Department of Biomedical Data SciencesLeiden University Medical CentreLeidenThe Netherlands
| | - Eka Suchiman
- Molecular Epidemiology, Department of Biomedical Data SciencesLeiden University Medical CentreLeidenThe Netherlands
| | - Fatih A. Bogaards
- Molecular Epidemiology, Department of Biomedical Data SciencesLeiden University Medical CentreLeidenThe Netherlands
- Division of Human NutritionWageningen University and ResearchWageningenThe Netherlands
| | - Daniele Bizzarri
- Molecular Epidemiology, Department of Biomedical Data SciencesLeiden University Medical CentreLeidenThe Netherlands
- Delft Bioinformatics Lab, Pattern Recognition and BioinformaticsDelftThe Netherlands
| | - Erik B. van den Akker
- Molecular Epidemiology, Department of Biomedical Data SciencesLeiden University Medical CentreLeidenThe Netherlands
- Delft Bioinformatics Lab, Pattern Recognition and BioinformaticsDelftThe Netherlands
| | - Melanie Waldenberger
- Research Unit Molecular Epidemiology, Institute of EpidemiologyHelmholtz Munich, German Research Center for Environmental HealthNeuherbergGermany
- German Center for Cardiovascular Research (DZHK)Partner Site Munich Heart AllianceMunichGermany
| | - Gillian Butler‐Browne
- Myology Center for Research, U974Sorbonne Université, INSERM, AIM, GH Pitié Salpêtrière Bat BabinskiParisFrance
| | - Capucine Trollet
- Myology Center for Research, U974Sorbonne Université, INSERM, AIM, GH Pitié Salpêtrière Bat BabinskiParisFrance
| | - C. P. G. M. de Groot
- Division of Human NutritionWageningen University and ResearchWageningenThe Netherlands
| | - Bastiaan T. Heijmans
- Molecular Epidemiology, Department of Biomedical Data SciencesLeiden University Medical CentreLeidenThe Netherlands
| | - P. Eline Slagboom
- Molecular Epidemiology, Department of Biomedical Data SciencesLeiden University Medical CentreLeidenThe Netherlands
| |
Collapse
|
11
|
Zonneveld MH, Al Kuhaili N, Mooijaart SP, Slagboom PE, Jukema JW, Noordam R, Trompet S. Increased 1H-NMR metabolomics-based health score associates with declined cognitive performance and functional independence in older adults at risk of cardiovascular disease. GeroScience 2025; 47:2035-2045. [PMID: 39436550 PMCID: PMC11978560 DOI: 10.1007/s11357-024-01391-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2024] [Accepted: 10/11/2024] [Indexed: 10/23/2024] Open
Abstract
The 1-HMR metabolomics-based MetaboHealth score, comprised of 14 serum metabolic markers, associates with disease-specific mortality, but it is unclear whether the score also reflects cognitive changes and functional impairment. We aimed to assess the associations between the MetaboHealth score with cognitive function and functional decline in older adults at increased cardiovascular risk. A total of 5292 older adults free of dementia at baseline with mean age 75.3 years (SD = 3.4) from the Prospective Study of Pravastatin in the Elderly (PROSPER). MetaboHealth score were measured at baseline, and cognitive function and functional independence were measured at baseline and every 3 months during up to 2.5 years follow-up. Cognitive function was assessed using the Stroop test (selective attention), the Letter Digit Coding test (LDCT) (processing speed), and the two versions of the Picture Learning test (delayed and immediate; memory). Two tests of functional independence were used: Barthel Index (BI) and instrumental activities at daily living (IADL). A higher MetaboHealth score was associated with worse cognitive function (in all domains) and with worse functional independence. For example, after full adjustments, a 1-SD higher MetaboHealth score was associated with 9.02 s (95%CI 7.29, 10.75) slower performance on the Stroop test and 2.79 (2.21, 3.26) less digits coded on the LDCT. During follow-up, 1-SD higher MetaboHealth score was associated with an additional decline of 0.53 s (0.23, 0.83) on the Stroop test and - 0.08 (- 0.11, - 0.06) points on the IADL. Metabolic disturbance, as reflected by an increased metabolomics-based health score, may mark future cognitive and functional decline.
Collapse
Affiliation(s)
- Michelle H Zonneveld
- Department of Internal Medicine, Section of Gerontology and Geriatrics, Leiden University Medical Center, PO Box 9600, 2300 RC, Leiden, The Netherlands
- Department of Cardiology, Leiden University Medical Center, Leiden, The Netherlands
| | - Nour Al Kuhaili
- Department of Internal Medicine, Section of Gerontology and Geriatrics, Leiden University Medical Center, PO Box 9600, 2300 RC, Leiden, The Netherlands
| | - Simon P Mooijaart
- Department of Internal Medicine, Section of Gerontology and Geriatrics, Leiden University Medical Center, PO Box 9600, 2300 RC, Leiden, The Netherlands
- LUMC Center for Medicine for Older People, Leiden University Medical Center, Leiden, The Netherlands
| | - P Eline Slagboom
- Department of Biomedical Data Sciences, Section of Molecular Epidemiology, Leiden University Medical Center, Leiden, The Netherlands
| | - J Wouter Jukema
- Department of Cardiology, Leiden University Medical Center, Leiden, The Netherlands
- Netherlands Heart Institute, Utrecht, The Netherlands
| | - Raymond Noordam
- Department of Internal Medicine, Section of Gerontology and Geriatrics, Leiden University Medical Center, PO Box 9600, 2300 RC, Leiden, The Netherlands.
| | - Stella Trompet
- Department of Internal Medicine, Section of Gerontology and Geriatrics, Leiden University Medical Center, PO Box 9600, 2300 RC, Leiden, The Netherlands
| |
Collapse
|
12
|
Li J, Man Q, Wang Y, Cui M, Li J, Xu K, Liu Z, Jin L, Chen X, Suo C, Jiang Y. The metabolic vulnerability index as a novel tool for mortality risk stratification in a large-scale population-based cohort. Redox Biol 2025; 81:103585. [PMID: 40064119 PMCID: PMC11930697 DOI: 10.1016/j.redox.2025.103585] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2025] [Accepted: 03/04/2025] [Indexed: 03/22/2025] Open
Abstract
Metabolic malnutrition and inflammation-key mechanism links to redox imbalance-are fundamental pathologies that accelerate aging and disease progression, ultimately leading to death. The recently proposed metabolic vulnerability index (MVX) integrates multiple circulatory biomarkers closely linked to both metabolic and inflammatory factors. This study aims to assess MVX's potential to predict mortality in community-based population. In this large community-based prospective study, we included UK Biobank participants who underwent plasma metabolomics analysis. Gender-specific MVX scores were calculated based on six established biomarkers of mortality. Linear and non-linear associations between MVX and mortality were assessed using Cox proportional hazards models and restricted cubic spline models, respectively. Among the 274,092 UKB participants, 24,241 all-cause deaths occurred during a median follow-up period of 13.7 years. A significant, graded positive association was observed between MVX quartiles and all-cause mortality risk (P for trend <0.05), with the highest MVX quartile exhibiting the greatest risk (HR = 1.21 and 95 % CI = 1.16-1.25 after full adjustment). Females had higher MVX score than males (P < 0.05), but males with the same MVX score faced a greater mortality risk. Baseline age and comorbidities interacted (P for interaction <0.05 and synergy index >1) with MVX on mortality risk. Longitudinal analyses showed that females with persistently high MVX score had a significantly increased risk of mortality (HR = 1.39 in fully adjusted model). Collectively, these findings highlight MVX as a novel tool that captures metabolic and potential redox vulnerabilities in community residents, and serves as a valuable resource for identifying high-risk individuals of mortality. Further research is warranted to investigate the underlying mechanisms and establish causal relationships.
Collapse
Affiliation(s)
- Jialin Li
- Human Phenome Institute, Research and Innovation Center, Shanghai Pudong Hospital, Zhangjiang Fudan International Innovation Center, Fudan University, Shanghai, 200433, China; Fudan University, Taizhou Institute of Health Sciences, Taizhou, Jiangsu, 225326, China
| | - Qiuhong Man
- Department of Laboratory Medicine, Shanghai Fourth People's Hospital, School of Medicine, Tongji University, Shanghai, 200434, China
| | - Yingzhe Wang
- Department of Neurology, Huashan Hospital, Fudan University, Shanghai, 200040, China
| | - Mei Cui
- Department of Neurology, Huashan Hospital, Fudan University, Shanghai, 200040, China
| | - Jincheng Li
- Human Phenome Institute, Research and Innovation Center, Shanghai Pudong Hospital, Zhangjiang Fudan International Innovation Center, Fudan University, Shanghai, 200433, China; Fudan University, Taizhou Institute of Health Sciences, Taizhou, Jiangsu, 225326, China
| | - Kelin Xu
- Fudan University, Taizhou Institute of Health Sciences, Taizhou, Jiangsu, 225326, China; Ministry of Education Key Laboratory of Public Health Safety, Department of Biostatistics, School of Public Health, Fudan University, Shanghai, 200032, China
| | - Zhenqiu Liu
- Human Phenome Institute, Research and Innovation Center, Shanghai Pudong Hospital, Zhangjiang Fudan International Innovation Center, Fudan University, Shanghai, 200433, China; Fudan University, Taizhou Institute of Health Sciences, Taizhou, Jiangsu, 225326, China
| | - Li Jin
- Fudan University, Taizhou Institute of Health Sciences, Taizhou, Jiangsu, 225326, China; State Key Laboratory of Genetic Engineering, Human Phenome Institute, Zhangjiang Fudan International Innovation Center, and National Clinical Research Center for Aging and Medicine, Huashan Hospital, Fudan University, Shanghai, 200433, China
| | - Xingdong Chen
- Fudan University, Taizhou Institute of Health Sciences, Taizhou, Jiangsu, 225326, China; State Key Laboratory of Genetic Engineering, Human Phenome Institute, Zhangjiang Fudan International Innovation Center, and National Clinical Research Center for Aging and Medicine, Huashan Hospital, Fudan University, Shanghai, 200433, China; Yiwu Research Institute of Fudan University, Yiwu, Zhejiang, 322000, China
| | - Chen Suo
- Fudan University, Taizhou Institute of Health Sciences, Taizhou, Jiangsu, 225326, China; Ministry of Education Key Laboratory of Public Health Safety, Department of Epidemiology, School of Public Health, Fudan University, Shanghai, 200032, China.
| | - Yanfeng Jiang
- Human Phenome Institute, Research and Innovation Center, Shanghai Pudong Hospital, Zhangjiang Fudan International Innovation Center, Fudan University, Shanghai, 200433, China; Fudan University, Taizhou Institute of Health Sciences, Taizhou, Jiangsu, 225326, China.
| |
Collapse
|
13
|
You D, Tang Y, Lange T, Wu Y, Lu M, Shao F, Shen S, Zhang R, Zhou H, Xu H, Yin Y, Wei Y, Chen F, Shen H, Christiani DC, Zhao Y. Systematic analysis of relationships between serum lipids with all-cause and cause-specific mortality: Evidence from prospective cohort studies of UK Biobank and Women's Health Initiative. Clin Nutr 2025; 47:94-102. [PMID: 39999642 DOI: 10.1016/j.clnu.2025.02.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2024] [Revised: 02/07/2025] [Accepted: 02/09/2025] [Indexed: 02/27/2025]
Abstract
BACKGROUND & AIMS Serum lipids, including lipoproteins, cholesterol, and triglycerides, are important modifiable factors influencing human health. However, the associations among different serum lipid profiles and mortality remain insufficiently understood, particularly regarding potential causality and population heterogeneity. This prospective study aims to systematically investigate the relationships between serum lipid concentrations of different densities and sizes with all-cause and cause-specific mortality. METHODS Cox proportional and Fine-Gray subdistribution hazard models were applied to investigate the associations of 54 lipid concentrations with all-cause and cause-specific mortality (including cardiovascular disease (CVD), cancer, and respiratory disease) in the UK Biobank cohort of 441,448 individuals with 17-year follow-up. Cohorts of 120,967 and 44,168 individuals from the Women's Health Initiative (WHI) with 16-year follow-up and a large-scale meta-analysis were utilized for external replication. We further assessed the underlying causality using Mendelian randomization (MR) and possible modifiers using multiple subgroup analyses. RESULTS During a median follow-up of 13.8 years, 39,290 deaths occurred, including 7399 from CVD, 18,928 from cancer, and 2707 from respiratory disease. We identified 160 significant associations between lipid concentrations and all-cause and cause-specific mortality. Importantly, most were inverse, with decreased lipid levels linked to increased risk of premature death [hazard ratios (HRs): 0.70-0.98 per standard deviation (SD)]. In contrast, positives were observed for HDL (large/very large) and triglyceride concentrations [HRs: 1.02-1.25 per SD], indicating increased mortality risk with higher levels. Most lipoproteins and cholesterol exhibited nonlinearly correlations with mortality, especially the significant U-shaped in total/HDL. However, MR showed that elevations in several lipids were associated with increased all-cause and CVD-specific mortality risk. Multiple subgroup analyses revealed that age, sex, and lipid-modifying drugs modified the lipid-mortality relationship; specifically, higher lipid concentrations increased mortality risk in younger adults not taking lipid-modifying drugs, but decreased mortality in older adults taking lipid-modifying drugs. The majority of associations were replicated in the WHI and external cohorts. CONCLUSION Our study systematically reported a large number of associations between serum lipid concentrations and mortality. Subgroup-based population heterogeneity analysis suggests that age, sex, and lipid-modifying drugs could be modifiers for the lipid-mortality relationship. These findings provide more guidance for lipid management and individualized prevention.
Collapse
Affiliation(s)
- Dongfang You
- Department of Biostatistics, School of Public Health, Nanjing Medical University, Nanjing, Jiangsu, 211166, China; Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing, Jiangsu, 210029, China.
| | - Yingdan Tang
- Department of Biostatistics, School of Public Health, Nanjing Medical University, Nanjing, Jiangsu, 211166, China
| | - Theis Lange
- Section of Biostatistics, Department of Public Health, Faculty of Health and Medical Sciences, University of Copenhagen, ØsterFarimagsgade 5, 1353, Copenhagen, Denmark
| | - Yaqian Wu
- Department of Biostatistics, School of Public Health, Nanjing Medical University, Nanjing, Jiangsu, 211166, China
| | - Mengyi Lu
- Department of Biostatistics, School of Public Health, Nanjing Medical University, Nanjing, Jiangsu, 211166, China
| | - Fang Shao
- Department of Biostatistics, School of Public Health, Nanjing Medical University, Nanjing, Jiangsu, 211166, China
| | - Sipeng Shen
- Department of Biostatistics, School of Public Health, Nanjing Medical University, Nanjing, Jiangsu, 211166, China
| | - Ruyang Zhang
- Department of Biostatistics, School of Public Health, Nanjing Medical University, Nanjing, Jiangsu, 211166, China; China International Cooperation Centre for Environment and Human Health, Centre for Global Health, Nanjing Medical University, Nanjing, Jiangsu, 211166, China
| | - Hongwen Zhou
- Department of Endocrinology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, 210029, China
| | - Hongyang Xu
- Department of Critical Care Medicine, Wuxi People's Hospital Affiliated to Nanjing Medical University, Wuxi, Jiangsu, 214023, China
| | - Yongmei Yin
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing, Jiangsu, 210029, China
| | - Yongyue Wei
- Department of Epidemiology and Biostatistics, School of Public Health, Peking University Health Science Centre, Beijing, 100191, China
| | - Feng Chen
- Department of Biostatistics, School of Public Health, Nanjing Medical University, Nanjing, Jiangsu, 211166, China; China International Cooperation Centre for Environment and Human Health, Centre for Global Health, Nanjing Medical University, Nanjing, Jiangsu, 211166, China; The Key Laboratory of Modern Toxicology of Ministry of Education, Nanjing Medical University, Nanjing, Jiangsu, 211166, China; Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Centre for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, Jiangsu, 211166, China
| | - Hongbing Shen
- Department of Epidemiology, School of Public Health, Nanjing Medical University, Nanjing, Jiangsu, 211166, China
| | - David C Christiani
- Department of Environmental Health, Harvard T.H. Chan School of Public Health, and Department of Medicine, Massachusetts General Hospital/Harvard Medical School, Boston, MA, 02115, USA
| | - Yang Zhao
- Department of Biostatistics, School of Public Health, Nanjing Medical University, Nanjing, Jiangsu, 211166, China; China International Cooperation Centre for Environment and Human Health, Centre for Global Health, Nanjing Medical University, Nanjing, Jiangsu, 211166, China; The Key Laboratory of Modern Toxicology of Ministry of Education, Nanjing Medical University, Nanjing, Jiangsu, 211166, China; Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Centre for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, Jiangsu, 211166, China; Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, 210029, China.
| |
Collapse
|
14
|
Kuo CL, Liu P, Drouard G, Vuoksimaa E, Kaprio J, Ollikainen M, Chen Z, Pilling LC, Atkins JL, Fortinsky RH, Kuchel GA, Diniz BS. A proteomic signature of healthspan. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2025:2024.06.26.24309530. [PMID: 38978645 PMCID: PMC11230312 DOI: 10.1101/2024.06.26.24309530] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 07/10/2024]
Abstract
The focus of aging research has shifted from increasing lifespan to enhancing healthspan to reduce the time spent living with disability. Despite significant efforts to develop biomarkers of aging, few studies have focused on biomarkers of healthspan. We developed a proteomics-based signature of healthspan (healthspan proteomic score (HPS)) using proteomic data from the Olink Explore 3072 assay in the UK Biobank Pharma Proteomics Project (53,018 individuals and 2920 proteins). A lower HPS was associated with higher mortality risk and several age-related conditions, such as COPD, diabetes, heart failure, cancer, myocardial infarction, dementia, and stroke. HPS showed superior predictive accuracy for these outcomes compared to other biological age measures. Proteins associated with HPS were enriched in hallmark pathways such as immune response, inflammation, cellular signaling, and metabolic regulation. The external validity was evaluated using the Essential Hypertension Epigenetics study with proteomic data also from the Olink Explore 3072 and complementary epigenetic data, making it a valuable tool for assessing healthspan and as a potential surrogate marker to complement existing proteomic and epigenetic biological age measures in geroscience-guided studies.
Collapse
Affiliation(s)
- Chia-Ling Kuo
- Department of Public Health Sciences, University of Connecticut Health Center, Farmington Connecticut, USA
- The Cato T. Laurencin Institute for Regenerative Engineering, University of Connecticut Health Center, Farmington, Connecticut, USA
- UConn Center on Aging, University of Connecticut Health Center, Farmington, CT, USA
| | - Peiran Liu
- The Cato T. Laurencin Institute for Regenerative Engineering, University of Connecticut Health Center, Farmington, Connecticut, USA
| | - Gabin Drouard
- Minerva Foundation Institute for Medical Research, Helsinki, Finland
| | - Eero Vuoksimaa
- Minerva Foundation Institute for Medical Research, Helsinki, Finland
| | - Jaakko Kaprio
- Minerva Foundation Institute for Medical Research, Helsinki, Finland
| | - Miina Ollikainen
- Minerva Foundation Institute for Medical Research, Helsinki, Finland
- Institute for Molecular Medicine Finland, HiLIFE, University of Helsinki, Helsinki, Finland
| | - Zhiduo Chen
- UConn Center on Aging, University of Connecticut Health Center, Farmington, CT, USA
| | - Luke C Pilling
- Department of Clinical and Biomedical Sciences, University of Exeter, Exeter, UK
| | - Janice L Atkins
- Department of Clinical and Biomedical Sciences, University of Exeter, Exeter, UK
| | - Richard H Fortinsky
- UConn Center on Aging, University of Connecticut Health Center, Farmington, CT, USA
| | - George A Kuchel
- UConn Center on Aging, University of Connecticut Health Center, Farmington, CT, USA
| | - Breno S Diniz
- Department of Public Health Sciences, University of Connecticut Health Center, Farmington Connecticut, USA
- UConn Center on Aging, University of Connecticut Health Center, Farmington, CT, USA
- Department of Psychiatry, University of Connecticut Health Center, Farmington Connecticut, USA
| |
Collapse
|
15
|
Prescott J, Keyser AJ, Litwin P, Dunbar MD, McClelland R, Ruple A, Ernst H, Butler BL, Kauffman M, Avery A, Harrison BR, Partida-Aguilar M, McCoy BM, Slikas E, Greenier AK, Muller E, Algavi YM, Bamberger T, Creevy KE, Borenstein E, Snyder-Mackler N, Promislow DEL. Rationale and design of the Dog Aging Project precision cohort: a multi-omic resource for longitudinal research in geroscience. GeroScience 2025:10.1007/s11357-025-01571-3. [PMID: 40038157 DOI: 10.1007/s11357-025-01571-3] [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: 10/25/2024] [Accepted: 02/17/2025] [Indexed: 03/06/2025] Open
Abstract
A significant challenge in multi-omic geroscience research is the collection of high quality, fit-for-purpose biospecimens from a diverse and well-characterized study population with sufficient sample size to detect age-related changes in physiological biomarkers. The Dog Aging Project designed the precision cohort to study the mechanisms underlying age-related change in the metabolome, microbiome, and epigenome in companion dogs, an emerging model system for translational geroscience research. One thousand dog-owner pairs were recruited into cohort strata based on life stage, sex, size, and geography. We designed and built a novel implementation of the REDCap electronic data capture system to manage study participants, logistics, and biospecimen and survey data collection in a secure online platform. In collaboration with primary care veterinarians, we collected and processed blood, urine, fecal, and hair samples from 976 dogs. The resulting data include complete blood count, chemistry profile, immunophenotyping by flow cytometry, metabolite quantification, fecal microbiome characterization, epigenomic profile, urinalysis, and associated metadata characterizing sample conditions at collection and during lab processing. The project, which has already begun collecting second- and third-year samples from precision cohort dogs, demonstrates that scientifically useful biospecimens can be collected from a geographically dispersed population through collaboration with private veterinary clinics and downstream labs. The data collection infrastructure developed for the precision cohort can be leveraged for future studies. Most important, the Dog Aging Project is an open data project. We encourage researchers around the world to apply for data access and utilize this rich, constantly growing dataset in their own work.
Collapse
Affiliation(s)
- Jena Prescott
- Department of Small Animal Clinical Sciences, Texas a&M University, College Station, TX, USA
| | - Amber J Keyser
- Center for Studies in Demography and Ecology, University of Washington, Seattle, WA, USA
| | - Paul Litwin
- Center for Studies in Demography and Ecology, University of Washington, Seattle, WA, USA
| | - Matthew D Dunbar
- Center for Studies in Demography and Ecology, University of Washington, Seattle, WA, USA
| | - Robyn McClelland
- Biostatistics and Collaborative Health Studies Coordinating Center, University of Washington, Seattle, WA, USA
| | - Audrey Ruple
- Department of Population Health Science, Virginia Tech, Blacksburg, VA, USA
| | - Holley Ernst
- Department of Small Animal Clinical Sciences, Texas a&M University, College Station, TX, USA
| | - Brianna L Butler
- Department of Small Animal Clinical Sciences, Texas a&M University, College Station, TX, USA
| | - Mandy Kauffman
- Center for Studies in Demography and Ecology, University of Washington, Seattle, WA, USA
| | - Anne Avery
- College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, CO, USA
| | - Benjamin R Harrison
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, USA
| | - Maria Partida-Aguilar
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, USA
| | - Brianah M McCoy
- School of Life Sciences, Arizona State University, Tempe, AZ, USA
| | - Elizabeth Slikas
- School of Life Sciences, Arizona State University, Tempe, AZ, USA
| | | | - Efrat Muller
- The Blavatnik School of Computer Science, Tel Aviv University, Tel Aviv, Israel
| | - Yadid M Algavi
- Faculty of Medical & Health Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Tal Bamberger
- Faculty of Medical & Health Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Kate E Creevy
- Department of Small Animal Clinical Sciences, Texas a&M University, College Station, TX, USA
| | - Elhanan Borenstein
- Blavatnik School of Computer Science and Faculty of Medical and Health Sciences, Tel Aviv University, Tel Aviv, Israel
| | | | - Daniel E L Promislow
- Jean Mayer USDA Human Nutrition Research Center on Aging, Tufts University, Boston, MA, USA.
| |
Collapse
|
16
|
Trisolini L, Musio B, Teixeira B, Sgobba MN, Francavilla AL, Volpicella M, Guerra L, De Grassi A, Gallo V, Duarte IF, Pierri CL. Exploring Metabolic Shifts in Kidney Cancer and Non-Cancer Cells Under Pro- and Anti-Apoptotic Treatments Using NMR Metabolomics. Cells 2025; 14:367. [PMID: 40072095 PMCID: PMC11899725 DOI: 10.3390/cells14050367] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2025] [Revised: 02/24/2025] [Accepted: 02/26/2025] [Indexed: 03/14/2025] Open
Abstract
This study investigates the metabolic responses of cancerous (RCC) and non-cancerous (HK2) kidney cells to treatment with Staurosporine (STAU), which has a pro-apoptotic effect, and Bongkrekic acid (BKA), which has an anti-apoptotic effect, individually and in combination, using 1H NMR metabolomics to identify metabolite markers linked to mitochondrial apoptotic pathways. BKA had minimal metabolic effects in RCC cells, suggesting its role in preserving mitochondrial function without significantly altering metabolic pathways. In contrast, STAU induced substantial metabolic reprogramming in RCC cells, disrupting energy production, redox balance, and biosynthesis, thereby triggering apoptotic pathways. The combined treatment of BKA and STAU primarily mirrored the effects of STAU alone, with BKA showing little capacity to counteract the pro-apoptotic effects. In non-cancerous HK2 cells, the metabolic alterations were far less pronounced, highlighting key differences in the metabolic responses of cancerous and non-cancerous cells. RCC cells displayed greater metabolic flexibility, while HK2 cells maintained a more regulated metabolic state. These findings emphasize the potential for targeting cancer-specific metabolic vulnerabilities while sparing non-cancerous cells, underscoring the value of metabolomics in understanding apoptotic and anti-apoptotic mechanisms. Future studies should validate these results in vivo and explore their potential for personalized treatment strategies.
Collapse
Affiliation(s)
- Lucia Trisolini
- Department of Biosciences, Biotechnologies and Environment, University of Bari “Aldo Moro”, Via Orabona, 4, 70125 Bari, Italy; (L.T.); (M.N.S.); (A.L.F.); (M.V.); (L.G.); (A.D.G.)
| | - Biagia Musio
- Department of Civil, Environmental, Land, Building Engineering and Chemistry (DICATECh), Polytechnic University of Bari, Via Orabona, 4, 70125 Bari, Italy; (B.M.); (V.G.)
| | - Beatriz Teixeira
- CICECO-Aveiro Institute of Materials and LAQV-REQUIMTE, Department of Chemistry, University of Aveiro, 3810-193 Aveiro, Portugal;
| | - Maria Noemi Sgobba
- Department of Biosciences, Biotechnologies and Environment, University of Bari “Aldo Moro”, Via Orabona, 4, 70125 Bari, Italy; (L.T.); (M.N.S.); (A.L.F.); (M.V.); (L.G.); (A.D.G.)
| | - Anna Lucia Francavilla
- Department of Biosciences, Biotechnologies and Environment, University of Bari “Aldo Moro”, Via Orabona, 4, 70125 Bari, Italy; (L.T.); (M.N.S.); (A.L.F.); (M.V.); (L.G.); (A.D.G.)
| | - Mariateresa Volpicella
- Department of Biosciences, Biotechnologies and Environment, University of Bari “Aldo Moro”, Via Orabona, 4, 70125 Bari, Italy; (L.T.); (M.N.S.); (A.L.F.); (M.V.); (L.G.); (A.D.G.)
| | - Lorenzo Guerra
- Department of Biosciences, Biotechnologies and Environment, University of Bari “Aldo Moro”, Via Orabona, 4, 70125 Bari, Italy; (L.T.); (M.N.S.); (A.L.F.); (M.V.); (L.G.); (A.D.G.)
| | - Anna De Grassi
- Department of Biosciences, Biotechnologies and Environment, University of Bari “Aldo Moro”, Via Orabona, 4, 70125 Bari, Italy; (L.T.); (M.N.S.); (A.L.F.); (M.V.); (L.G.); (A.D.G.)
| | - Vito Gallo
- Department of Civil, Environmental, Land, Building Engineering and Chemistry (DICATECh), Polytechnic University of Bari, Via Orabona, 4, 70125 Bari, Italy; (B.M.); (V.G.)
- Innovative Solutions S.r.l.—Spin-Off Company of the Polytechnic University of Bari, Zona H 150/B, 70015 Noci, Italy
| | - Iola F. Duarte
- CICECO-Aveiro Institute of Materials and LAQV-REQUIMTE, Department of Chemistry, University of Aveiro, 3810-193 Aveiro, Portugal;
| | - Ciro Leonardo Pierri
- Department of Pharmacy—Pharmaceutical Sciences, University of Bari “Aldo Moro”, Via Orabona, 4, 70125 Bari, Italy
| |
Collapse
|
17
|
Kyhl LK, Nordestgaard BG, Tybjærg-Hansen A, Smith GD, Nielsen SF. VLDL triglycerides and cholesterol in non-alcoholic fatty liver disease and myocardial infarction. Atherosclerosis 2025; 401:119094. [PMID: 39837114 DOI: 10.1016/j.atherosclerosis.2024.119094] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/13/2023] [Revised: 12/12/2024] [Accepted: 12/12/2024] [Indexed: 01/23/2025]
Abstract
BACKGROUND AND AIMS Myocardial infarction is a leading cause of death in individuals with non-alcoholic fatty liver disease (NAFLD). The two diseases share elevated very low-density lipoproteins (VLDL) carrying both triglycerides and cholesterol; however, in NAFLD mainly triglycerides accumulate in liver cells while in myocardial infarction mainly cholesterol accumulates in the atherosclerotic plaque. We hypothesized that VLDL triglycerides preferentially associate with risk of NAFLD, while VLDL cholesterol preferentially associates with risk of myocardial infarction. METHODS We examined 25,428 individuals without clinically diagnosed NAFLD or myocardial infarction at baseline, nested within 109,776 individuals from the prospective Copenhagen General Population Study and followed these individuals for a mean of 10 years. VLDL triglycerides, VLDL cholesterol, and low-density lipoprotein (LDL) cholesterol were determined using nuclear magnetic resonance spectrometry. RESULTS Continuously higher VLDL triglycerides were associated with continuously higher risk of NAFLD; however, this was not the case for VLDL cholesterol, LDL cholesterol, or apolipoprotein B. In contrast, continuously higher VLDL cholesterol, LDL cholesterol, and plasma apolipoprotein B were all associated with continuously higher risk of myocardial infarction. Compared to individuals with both VLDL triglycerides and VLDL cholesterol ≤66th percentile, the hazard ratios for NAFLD in individuals with VLDL triglycerides >66th percentile were 1.61(95 % confidence intervals:1.25-2.06) at high VLDL cholesterol and 1.41(0.90-2.21) at low VLDL cholesterol. Corresponding hazard ratios for myocardial infarction in individuals with VLDL cholesterol >66th percentile were 1.51(1.36-1.67) at high VLDL triglycerides and 1.42(1.18-1.69) at low VLDL triglycerides. CONCLUSIONS VLDL triglycerides predominated in NAFLD while VLDL cholesterol predominated in myocardial infarction; however, VLDL cholesterol was also elevated slightly in NAFLD while VLDL triglycerides was also elevated in myocardial infarction.
Collapse
Affiliation(s)
- Lærke Kristine Kyhl
- Department of Clinical Biochemistry, Copenhagen University Hospital - Herlev Gentofte, Herlev, Denmark; The Copenhagen General Population Study, Copenhagen University Hospital - Herlev Gentofte, Herlev, Denmark; Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Børge Grønne Nordestgaard
- Department of Clinical Biochemistry, Copenhagen University Hospital - Herlev Gentofte, Herlev, Denmark; The Copenhagen General Population Study, Copenhagen University Hospital - Herlev Gentofte, Herlev, Denmark; Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Anne Tybjærg-Hansen
- The Copenhagen General Population Study, Copenhagen University Hospital - Herlev Gentofte, Herlev, Denmark; Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark; Department of Clinical Biochemistry, Copenhagen University Hospital - Rigshospitalet, Copenhagen, Denmark
| | - George Davey Smith
- MRC Integrative Epidemiology Unit (IEU), Bristol Medical School, University of Bristol, United Kingdom; Population Health Sciences, Bristol Medical School, University of Bristol, United Kingdom
| | - Sune Fallgaard Nielsen
- Department of Clinical Biochemistry, Copenhagen University Hospital - Herlev Gentofte, Herlev, Denmark; The Copenhagen General Population Study, Copenhagen University Hospital - Herlev Gentofte, Herlev, Denmark; Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark.
| |
Collapse
|
18
|
Yang S, Xin Z, Cheng W, Zhong P, Liu R, Zhu Z, Zhu LZ, Shang X, Chen S, Huang W, Zhang L, Wang W. Photoreceptor metabolic window unveils eye-body interactions. Nat Commun 2025; 16:697. [PMID: 39814712 PMCID: PMC11736035 DOI: 10.1038/s41467-024-55035-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2024] [Accepted: 11/26/2024] [Indexed: 01/18/2025] Open
Abstract
Photoreceptors are specialized neurons at the core of the retina's functionality, with optical accessibility and exceptional sensitivity to systemic metabolic stresses. Here we show the ability of risk-free, in vivo photoreceptor assessment as a window into systemic health and identify shared metabolic underpinnings of photoreceptor degeneration and multisystem health outcomes. A thinner photoreceptor layer thickness is significantly associated with an increased risk of future mortality and 13 multisystem diseases, while systematic analyses of circulating metabolomics enable the identification of 109 photoreceptor-related metabolites, which in turn elevate or reduce the risk of these health outcomes. To translate these insights into a practical tool, we developed an artificial intelligence (AI)-driven photoreceptor metabolic window framework and an accompanying interpreter that comprehensively captures the metabolic landscape of photoreceptor-systemic health linkages and simultaneously predicts 16 multisystem health outcomes beyond established approaches while retaining interpretability. Our work, replicated across cohorts of diverse ethnicities, reveals the potential of photoreceptors to inform systemic health and advance a multisystem perspective on human health by revealing eye-body connections and shared metabolic influences.
Collapse
Affiliation(s)
- Shaopeng Yang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangdong Provincial Clinical Study Center for Ocular Diseases, Guangzhou, China
| | - Zhuoyao Xin
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, USA
- F.M. Kirby Research Center for Functional Brain Imaging, Kennedy Krieger Institute, Baltimore, MD, USA
- Department of Biomedical Engineering, Columbia University, New York, NY, USA
| | - Weijing Cheng
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangdong Provincial Clinical Study Center for Ocular Diseases, Guangzhou, China
| | - Pingting Zhong
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangdong Provincial Clinical Study Center for Ocular Diseases, Guangzhou, China
| | - Riqian Liu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangdong Provincial Clinical Study Center for Ocular Diseases, Guangzhou, China
| | - Ziyu Zhu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangdong Provincial Clinical Study Center for Ocular Diseases, Guangzhou, China
| | - Lisa Zhuoting Zhu
- Centre for Eye Research Australia, Royal Victorian Eye and Ear Hospital, University of Melbourne, Melbourne, VIC, Australia
| | - Xianwen Shang
- Centre for Eye Research Australia, Royal Victorian Eye and Ear Hospital, University of Melbourne, Melbourne, VIC, Australia
| | - Shida Chen
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangdong Provincial Clinical Study Center for Ocular Diseases, Guangzhou, China
| | - Wenyong Huang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangdong Provincial Clinical Study Center for Ocular Diseases, Guangzhou, China
| | - Lei Zhang
- Clinical Medical Research Center, Children's Hospital of Nanjing Medical University, Nanjing, China
- Artificial Intelligence and Modelling in Epidemiology Program, Melbourne Sexual Health Centre, Alfred Health, Melbourne, Australia
- Central Clinical School, Faculty of Medicine, Nursing and Health Sciences, Monash University, Melbourne, Australia
| | - Wei Wang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangdong Provincial Clinical Study Center for Ocular Diseases, Guangzhou, China.
- Hainan Eye Hospital and Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Haikou, Hainan Province, China.
| |
Collapse
|
19
|
Zhu M, Lamont L, Maas P, Harms AC, Beekman M, Slagboom PE, Dubbelman AC, Hankemeier T. Matrix effect evaluation using multi-component post-column infusion in untargeted hydrophilic interaction liquid chromatography-mass spectrometry plasma metabolomics. J Chromatogr A 2025; 1740:465580. [PMID: 39644743 DOI: 10.1016/j.chroma.2024.465580] [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: 07/26/2024] [Revised: 11/28/2024] [Accepted: 12/02/2024] [Indexed: 12/09/2024]
Abstract
Metabolomics based on hydrophilic interaction liquid chromatography (HILIC) coupled with mass spectrometry (MS) is a powerful tool for polar metabolite identification and quantification to further contribute to biomarker discovery and disease mechanism elucidation. However, matrix effect (ME), which may lead to altered ionization efficiency due to co-eluting compounds, is a significant challenge during biological analysis. Therefore, ME evaluation plays a crucial role during method development. Two approaches to evaluate ME are using stable isotope labelled-internal standards (SIL-IS) and post-column infusion (PCI) of standards. In this study, we developed an untargeted HILIC-MS method by applying four PCI standards for ME evaluation. We found PCI is a compelling approach for ME assessment compared to SIL-IS method due to its advantage in untargeted analysis. Through the ME evaluation and chromatographic performance comparison of 18 SIL standards across three columns and three different mobile phase pH conditions, our findings revealed that the BEH-Z-HILIC column operated at pH 4 with 10 mM ammonium formate exhibited minimal ME and superior performance. The method showed exceptional linearity (R² > 0.98), reliable repeatability (RSD < 15 %), good inter-day precision (RSD < 30 %), and acceptable recovery (>75 %) for all SIL standards. Absolute matrix effect (AME) and relative matrix effect (RME) assessment in three plasma donors revealed a high consistency between PCI and SIL-IS approaches. Finally, this method coupled with the PCI approach was applied to 40 plasma samples. Fifty endogenous compounds were detected and their AME and RME were evaluated. Results showed that many compounds experienced severe ion suppression, though their ME variation between 40 samples is low. In conclusion, PCI method is a robust alternative for monitoring ME and evaluating ME of endogenous compounds during untargeted method optimization and biological analysis.
Collapse
Affiliation(s)
- Mengle Zhu
- Metabolomics and Analytics Centre, Leiden Academic Centre for Drug Research, Leiden University, Leiden, the Netherlands
| | - Lieke Lamont
- Metabolomics and Analytics Centre, Leiden Academic Centre for Drug Research, Leiden University, Leiden, the Netherlands
| | - Pascal Maas
- Metabolomics and Analytics Centre, Leiden Academic Centre for Drug Research, Leiden University, Leiden, the Netherlands
| | - Amy C Harms
- Metabolomics and Analytics Centre, Leiden Academic Centre for Drug Research, Leiden University, Leiden, the Netherlands
| | - Marian Beekman
- Department of Biomedical Data Sciences, Section of Molecular Epidemiology, Leiden University Medical Center, Leiden, the Netherlands
| | - P Eline Slagboom
- Department of Biomedical Data Sciences, Section of Molecular Epidemiology, Leiden University Medical Center, Leiden, the Netherlands
| | - Anne-Charlotte Dubbelman
- Metabolomics and Analytics Centre, Leiden Academic Centre for Drug Research, Leiden University, Leiden, the Netherlands; Institute of Risk Assessment Sciences, Utrecht University, Utrecht 3584 CM, the Netherlands.
| | - Thomas Hankemeier
- Metabolomics and Analytics Centre, Leiden Academic Centre for Drug Research, Leiden University, Leiden, the Netherlands.
| |
Collapse
|
20
|
Huang H, Chen Y, Xu W, Cao L, Qian K, Bischof E, Kennedy BK, Pu J. Decoding aging clocks: New insights from metabolomics. Cell Metab 2025; 37:34-58. [PMID: 39657675 DOI: 10.1016/j.cmet.2024.11.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/25/2024] [Revised: 09/23/2024] [Accepted: 11/10/2024] [Indexed: 12/12/2024]
Abstract
Chronological age is a crucial risk factor for diseases and disabilities among older adults. However, individuals of the same chronological age often exhibit divergent biological aging states, resulting in distinct individual risk profiles. Chronological age estimators based on omics data and machine learning techniques, known as aging clocks, provide a valuable framework for interpreting molecular-level biological aging. Metabolomics is an intriguing and rapidly growing field of study, involving the comprehensive profiling of small molecules within the body and providing the ultimate genome-environment interaction readout. Consequently, leveraging metabolomics to characterize biological aging holds immense potential. The aim of this review was to provide an overview of metabolomics approaches, highlighting the establishment and interpretation of metabolomic aging clocks while emphasizing their strengths, limitations, and applications, and to discuss their underlying biological significance, which has the potential to drive innovation in longevity research and development.
Collapse
Affiliation(s)
- Honghao Huang
- Division of Cardiology, State Key Laboratory for Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Yifan Chen
- Division of Cardiology, State Key Laboratory for Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Wei Xu
- Division of Cardiology, State Key Laboratory for Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Linlin Cao
- Division of Cardiology, State Key Laboratory for Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Kun Qian
- Division of Cardiology, State Key Laboratory for Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China; School of Biomedical Engineering and Med-X Research Institute, Shanghai Jiao Tong University, Shanghai, China
| | - Evelyne Bischof
- University Hospital of Basel, Division of Internal Medicine, University of Basel, Basel, Switzerland; Shanghai University of Medicine and Health Sciences, College of Clinical Medicine, Shanghai, China
| | - Brian K Kennedy
- Health Longevity Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore; Centre for Healthy Longevity, National University Health System, Singapore, Singapore; Departments of Biochemistry and Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.
| | - Jun Pu
- Division of Cardiology, State Key Laboratory for Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China; Aging Biomarker Consortium, China.
| |
Collapse
|
21
|
Sergeev AV, Kisil OV, Eremin AA, Petrov AS, Zvereva ME. "Aging Clocks" Based on Cell-Free DNA. BIOCHEMISTRY. BIOKHIMIIA 2025; 90:S342-S355. [PMID: 40164165 DOI: 10.1134/s0006297924604076] [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: 08/09/2024] [Revised: 10/08/2024] [Accepted: 10/17/2024] [Indexed: 04/02/2025]
Abstract
Aging is associated with systemic changes in the physiological and molecular parameters of the body. These changes are referred to as biomarkers of aging. Statistical models that link changes in individual biomarkers to biological age are called aging clocks. These tools facilitate a comprehensive evaluation of bodily health and permit the quantitative determination of the rate of aging. A particularly promising area for the development of aging clocks is the analysis of cell-free DNA (cfDNA), which is present in the blood and contains numerous potential biomarkers. This review explores in detail the fragmentomics, topology, and epigenetic landscape of cfDNA as possible biomarkers of aging. The review further underscores the potential of leveraging single-molecule sequencing of cfDNA in conjunction with long-read technologies to simultaneously profile multiple biomarkers, a strategy that could lead to the development of more precise aging clocks.
Collapse
Affiliation(s)
- Aleksandr V Sergeev
- Faculty of Chemistry, Lomonosov Moscow State University, Moscow, 119991, Russia.
- Orekhovich Scientific Research Institute of Biomedical Chemistry, Moscow, 119121, Russia
| | - Olga V Kisil
- Faculty of Chemistry, Lomonosov Moscow State University, Moscow, 119991, Russia
- Gauze Scientific Research Institute of New Antibiotics, Moscow, 119021, Russia
| | - Andrey A Eremin
- Faculty of Chemistry, Lomonosov Moscow State University, Moscow, 119991, Russia
| | - Aleksandr S Petrov
- Faculty of Chemistry, Lomonosov Moscow State University, Moscow, 119991, Russia
| | - Maria E Zvereva
- Faculty of Chemistry, Lomonosov Moscow State University, Moscow, 119991, Russia
| |
Collapse
|
22
|
Luo J, le Cessie S, Willems van Dijk K, Hägg S, Grassmann F, van Heemst D, Noordam R. Mitochondrial DNA abundance and circulating metabolomic profiling: Multivariable-adjusted and Mendelian randomization analyses in UK Biobank. Mitochondrion 2025; 80:101991. [PMID: 39592086 DOI: 10.1016/j.mito.2024.101991] [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: 07/12/2024] [Revised: 11/11/2024] [Accepted: 11/22/2024] [Indexed: 11/28/2024]
Abstract
BACKGROUND Low leukocyte mitochondrial DNA (mtDNA) abundance has been associated with a higher risk of atherosclerotic cardiovascular disease, but through unclear mechanisms. We aimed to investigate whether low mtDNA abundance is associated with worse metabolomic profiling, as being potential intermediate phenotypes, using cross-sectional and genetic studies. METHODS Among 61,186 unrelated European participants from UK Biobank, we performed multivariable-adjusted linear regression analyses to examine the associations between mtDNA abundance and 168 NMR-based circulating metabolomic measures and nine metabolomic principal components (PCs) that collectively covered 91.5% of the total variation of individual metabolomic measures. Subsequently, we conducted Mendelian randomization (MR) to approximate the causal effects of mtDNA abundance on the individual metabolomic measures and their metabolomic PCs. RESULTS After correction for multiple testing, low mtDNA abundance was associated with 130 metabolomic measures, predominantly lower concentrations of some amino acids and higher concentrations of lipids, lipoproteins and fatty acids; moreover, mtDNA abundance was associated with seven out of the nine metabolomic PCs. Using MR, genetically-predicted low mtDNA abundance was associated with lower lactate (standardized beta and 95% confidence interval: -0.17; -0.26, -0.08), and higher acetate (0.15; 0.07,0.23), and unsaturation degree (0.14; 0.08,0.20). Similarly, genetically-predicted low mtDNA abundance was associated with lower metabolomic PC2 (related to lower concentrations of lipids and fatty acids), and higher metabolomic PC9 (related to lower concentrations of glycolysis-related metabolites). CONCLUSION Low mtDNA abundance is associated with metabolomic perturbations, particularly reflecting a pro-atherogenic metabolomic profile, which potentially could link low mtDNA abundance to higher atherosclerosis risk.
Collapse
Affiliation(s)
- Jiao Luo
- Department of Internal Medicine, Section of Gerontology and Geriatrics, Leiden University Medical Center, Leiden, the Netherlands; Department of Clinical Epidemiology, Leiden University Medical Center, Leiden, the Netherlands
| | - Saskia le Cessie
- Department of Internal Medicine, Section of Gerontology and Geriatrics, Leiden University Medical Center, Leiden, the Netherlands; Department of Biomedical Data Sciences, Leiden University Medical Center, Leiden, the Netherlands
| | - Ko Willems van Dijk
- Department of Internal Medicine, Division of Endocrinology, Leiden University Medical Center, Leiden, the Netherlands; Department of Human Genetics, Leiden University Medical Center, Leiden, the Netherlands
| | - Sara Hägg
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - Felix Grassmann
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden; Institute for Clinical Research and Systems Medicine, Health and Medical University, Potsdam, Germany
| | - Diana van Heemst
- Department of Internal Medicine, Section of Gerontology and Geriatrics, Leiden University Medical Center, Leiden, the Netherlands
| | - Raymond Noordam
- Department of Internal Medicine, Section of Gerontology and Geriatrics, Leiden University Medical Center, Leiden, the Netherlands.
| |
Collapse
|
23
|
Mutz J, Iniesta R, Lewis CM. Metabolomic age (MileAge) predicts health and life span: A comparison of multiple machine learning algorithms. SCIENCE ADVANCES 2024; 10:eadp3743. [PMID: 39693428 DOI: 10.1126/sciadv.adp3743] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/21/2024] [Accepted: 11/14/2024] [Indexed: 12/20/2024]
Abstract
Biological aging clocks produce age estimates that can track with age-related health outcomes. This study aimed to benchmark machine learning algorithms, including regularized regression, kernel-based methods, and ensembles, for developing metabolomic aging clocks from nuclear magnetic resonance spectroscopy data. The UK Biobank data, including 168 plasma metabolites from up to N = 225,212 middle-aged and older adults (mean age, 56.97 years), were used to train and internally validate 17 algorithms. Metabolomic age (MileAge) delta, the difference between metabolite-predicted and chronological age, from a Cubist rule-based regression model showed the strongest associations with health and aging markers. Individuals with an older MileAge were frailer, had shorter telomeres, were more likely to suffer from chronic illness, rated their health worse, and had a higher all-cause mortality hazard (HR = 1.51; 95% CI, 1.43 to 1.59; P < 0.001). This metabolomic aging clock (MileAge) can be applied in research and may find use in health assessments, risk stratification, and proactive health tracking.
Collapse
Affiliation(s)
- Julian Mutz
- Department of Biostatistics and Health Informatics, King's College London, London, United Kingdom
| | - Raquel Iniesta
- Department of Biostatistics and Health Informatics, King's College London, London, United Kingdom
| | - Cathryn M Lewis
- Social, Genetic and Developmental Psychiatry Centre, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, United Kingdom
- Department of Medical and Molecular Genetics, Faculty of Life Sciences and Medicine, King's College London, London, United Kingdom
| |
Collapse
|
24
|
Yeh CY, Chini LCS, Davidson JW, Garcia GG, Gallagher MS, Freichels IT, Calubag MF, Rodgers AC, Green CL, Babygirija R, Sonsalla MM, Pak HH, Trautman ME, Hacker TA, Miller RA, Simcox JA, Lamming DW. Late-life protein or isoleucine restriction impacts physiological and molecular signatures of aging. NATURE AGING 2024; 4:1760-1771. [PMID: 39604703 PMCID: PMC11672203 DOI: 10.1038/s43587-024-00744-7] [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: 01/09/2024] [Accepted: 10/10/2024] [Indexed: 11/29/2024]
Abstract
Restricting the intake of protein or the branched-chain amino acid isoleucine promotes healthspan and extends lifespan in young or adult mice. However, their effects when initiated in aged animals are unknown. Here we investigate the consequences of consuming a diet with 67% reduction of all amino acids (low AA) or of isoleucine alone (low Ile), in male and female C57BL/6J.Nia mice starting at 20 months of age. Both dietary regimens effectively promote overall metabolic health without reducing calorie intake. Both low AA and low Ile diets improve aspects of frailty and slow multiple molecular indicators of aging rate; however, the low Ile diet reduces grip strength in both sexes and has mixed, sexually dimorphic effects on the heart. These results demonstrate that low AA and low Ile diets can promote aspects of healthy aging in aged mice and suggest that similar interventions might promote healthy aging in older adults.
Collapse
Affiliation(s)
- Chung-Yang Yeh
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA
- William S. Middleton Memorial Veterans Hospital, Madison, WI, USA
| | - Lucas C S Chini
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA
- William S. Middleton Memorial Veterans Hospital, Madison, WI, USA
| | - Jessica W Davidson
- Department of Biochemistry, University of Wisconsin-Madison, Madison, WI, USA
| | - Gonzalo G Garcia
- Department of Pathology, University of Michigan School of Medicine, Ann Arbor, MI, USA
| | - Meredith S Gallagher
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA
- William S. Middleton Memorial Veterans Hospital, Madison, WI, USA
| | - Isaac T Freichels
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA
- William S. Middleton Memorial Veterans Hospital, Madison, WI, USA
| | - Mariah F Calubag
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA
- William S. Middleton Memorial Veterans Hospital, Madison, WI, USA
- Graduate Program in Cellular and Molecular Biology, University of Wisconsin-Madison, Madison, WI, USA
| | - Allison C Rodgers
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA
- Cardiovascular Physiology Core Facility, University of Wisconsin-Madison, Madison, WI, USA
| | - Cara L Green
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA
- William S. Middleton Memorial Veterans Hospital, Madison, WI, USA
| | - Reji Babygirija
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA
- William S. Middleton Memorial Veterans Hospital, Madison, WI, USA
- Graduate Program in Cellular and Molecular Biology, University of Wisconsin-Madison, Madison, WI, USA
| | - Michelle M Sonsalla
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA
- William S. Middleton Memorial Veterans Hospital, Madison, WI, USA
- Comparative Biomedical Sciences Graduate Training Program, University of Wisconsin-Madison, Madison, WI, USA
| | - Heidi H Pak
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA
- William S. Middleton Memorial Veterans Hospital, Madison, WI, USA
- Nutrition and Metabolism Graduate Program, University of Wisconsin-Madison, Madison, WI, USA
| | - Michaela E Trautman
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA
- William S. Middleton Memorial Veterans Hospital, Madison, WI, USA
- Nutrition and Metabolism Graduate Program, University of Wisconsin-Madison, Madison, WI, USA
| | - Timothy A Hacker
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA
- Cardiovascular Physiology Core Facility, University of Wisconsin-Madison, Madison, WI, USA
| | - Richard A Miller
- Department of Pathology, University of Michigan School of Medicine, Ann Arbor, MI, USA
| | - Judith A Simcox
- Department of Biochemistry, University of Wisconsin-Madison, Madison, WI, USA
- Howard Hughes Medical Institute, Department of Biochemistry, University of Wisconsin-Madison, Madison, WI, USA
| | - Dudley W Lamming
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA.
- William S. Middleton Memorial Veterans Hospital, Madison, WI, USA.
- Graduate Program in Cellular and Molecular Biology, University of Wisconsin-Madison, Madison, WI, USA.
- Comparative Biomedical Sciences Graduate Training Program, University of Wisconsin-Madison, Madison, WI, USA.
- Nutrition and Metabolism Graduate Program, University of Wisconsin-Madison, Madison, WI, USA.
- University of Wisconsin-Madison Comprehensive Diabetes Center, Madison, WI, USA.
- University of Wisconsin Carbone Cancer Center, Madison, WI, USA.
| |
Collapse
|
25
|
Kramer CS, Monsegue A, Morwani-Mangnani J, Grootswagers P, Beekman M, Slagboom PE, Verdijk LB, de Groot LCPGM. Design of the VOILA-intervention study: A 12-week nutrition and resistance exercise intervention in metabolic or mobility compromised Dutch older adults and the response on immune-metabolic, gut and muscle health parameters. Mech Ageing Dev 2024; 222:112002. [PMID: 39490538 DOI: 10.1016/j.mad.2024.112002] [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: 07/25/2024] [Revised: 10/16/2024] [Accepted: 10/24/2024] [Indexed: 11/05/2024]
Abstract
BACKGROUND Exercise and nutrition interventions can slow ageing-induced decline in physiology. However, effects are heterogeneous and usually studied separately per outcome domain. In the VOILA study, we simultaneously study various health outcomes relevant for older adults and the inter-individual heterogeneity in response to a lifestyle intervention. METHODS VOILA is a 12-week lifestyle intervention in 3 groups of older adults (≥60 years), with compromised mobility (n=50), compromised metabolic health (n=50), or recovering from total knee replacement (TKR, n=70, of which 20 randomized to standard care only). The intervention includes high-intensity resistance exercise training thrice weekly, nutritional counselling, and nutritional supplements every morning and evening (including 20-25 g whey protein and (evening only) 5.5 g Biotis™ GOS). We measure immune-metabolic, gut health, muscle mass and physical functioning at baseline and after completion of the intervention/standard care. An additional reference group of healthy older adults (n=50) will undergo baseline measurements only. DISCUSSION Improvements in various physiological systems are expected, but with differences between groups/individuals. This study will provide insights into how the physiological state of older adults influences the extent of lifestyle-induced health improvements to create better tailored interventions to attenuate biological ageing and improve the health span of subgroups and individuals.
Collapse
Affiliation(s)
- C S Kramer
- Wageningen University & Research, Wageningen Campus, Agrotechnology and Food Sciences Group, Division of Human Nutrition and Health, PO Box 17, Wageningen 6700 AA, the Netherlands.
| | - A Monsegue
- Maastricht University Medical Center+, Department of Human Biology, NUTRIM Institute of nutrition and translational research in metabolism, PO Box 616, Maastricht 6200 MD, the Netherlands.
| | - J Morwani-Mangnani
- Leiden University Medical Centre, Department of Biomedical Data Sciences, Section of Molecular Epidemiology, Einthovenweg 20, Leiden 2333 ZC, the Netherlands.
| | - P Grootswagers
- Wageningen University & Research, Wageningen Campus, Agrotechnology and Food Sciences Group, Division of Human Nutrition and Health, PO Box 17, Wageningen 6700 AA, the Netherlands.
| | - M Beekman
- Leiden University Medical Centre, Department of Biomedical Data Sciences, Section of Molecular Epidemiology, Einthovenweg 20, Leiden 2333 ZC, the Netherlands.
| | - P E Slagboom
- Leiden University Medical Centre, Department of Biomedical Data Sciences, Section of Molecular Epidemiology, Einthovenweg 20, Leiden 2333 ZC, the Netherlands.
| | - L B Verdijk
- Maastricht University Medical Center+, Department of Human Biology, NUTRIM Institute of nutrition and translational research in metabolism, PO Box 616, Maastricht 6200 MD, the Netherlands.
| | - L C P G M de Groot
- Wageningen University & Research, Wageningen Campus, Agrotechnology and Food Sciences Group, Division of Human Nutrition and Health, PO Box 17, Wageningen 6700 AA, the Netherlands.
| |
Collapse
|
26
|
van Holstein Y, Mooijaart SP, van Oevelen M, van Deudekom FJ, Vojinovic D, Bizzarri D, van den Akker EB, Noordam R, Deelen J, van Heemst D, de Glas NA, Holterhues C, Labots G, van den Bos F, Beekman M, Slagboom PE, van Munster BC, Portielje JEA, Trompet S. The performance of metabolomics-based prediction scores for mortality in older patients with solid tumors. GeroScience 2024; 46:5615-5627. [PMID: 38963649 PMCID: PMC11493906 DOI: 10.1007/s11357-024-01261-6] [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: 04/10/2024] [Accepted: 06/23/2024] [Indexed: 07/05/2024] Open
Abstract
Prognostic information is needed to balance benefits and risks of cancer treatment in older patients. Metabolomics-based scores were previously developed to predict 5- and 10-year mortality (MetaboHealth) and biological age (MetaboAge). This study aims to investigate the association of MetaboHealth and MetaboAge with 1-year mortality in older patients with solid tumors, and to study their predictive value for mortality in addition to established clinical predictors. This prospective cohort study included patients aged ≥ 70 years with a solid malignant tumor, who underwent blood sampling and a geriatric assessment before treatment initiation. The outcome was all-cause 1-year mortality. Of the 192 patients, the median age was 77 years. With each SD increase of MetaboHealth, patients had a 2.32 times increased risk of mortality (HR 2.32, 95% CI 1.59-3.39). With each year increase in MetaboAge, there was a 4% increased risk of mortality (HR 1.04, 1.01-1.07). MetaboHealth and MetaboAge showed an AUC of 0.66 (0.56-0.75) and 0.60 (0.51-0.68) for mortality prediction accuracy, respectively. The AUC of a predictive model containing age, primary tumor site, distant metastasis, comorbidity, and malnutrition was 0.76 (0.68-0.83). Addition of MetaboHealth increased AUC to 0.80 (0.74-0.87) (p = 0.09) and AUC did not change with MetaboAge (0.76 (0.69-0.83) (p = 0.89)). Higher MetaboHealth and MetaboAge scores were associated with 1-year mortality. The addition of MetaboHealth to established clinical predictors only marginally improved mortality prediction in this cohort with various types of tumors. MetaboHealth may potentially improve identification of older patients vulnerable for adverse events, but numbers were too small for definitive conclusions. The TENT study is retrospectively registered at the Netherlands Trial Register (NTR), trial number NL8107. Date of registration: 22-10-2019.
Collapse
Affiliation(s)
- Yara van Holstein
- Department of Internal Medicine, Section of Gerontology and Geriatrics, Leiden University Medical Center, PO box 9600, 2300 RC, Leiden, The Netherlands.
| | - Simon P Mooijaart
- Department of Internal Medicine, Section of Gerontology and Geriatrics, Leiden University Medical Center, PO box 9600, 2300 RC, Leiden, The Netherlands
- LUMC Center for Medicine for Older People, Leiden University Medical Center, Leiden, The Netherlands
| | - Mathijs van Oevelen
- Department of Internal Medicine, Section of Nephrology, Leiden University Medical Center, Leiden, The Netherlands
| | - Floor J van Deudekom
- Department of Geriatric Medicine, OLVG Hospitals Amsterdam, Amsterdam, The Netherlands
| | - Dina Vojinovic
- Department of Biomedical Data Sciences, Section of Molecular Epidemiology, Leiden University Medical Center, Leiden, The Netherlands
- Department of Epidemiology, Erasmus Medical Center, University Medical Centre, Rotterdam, The Netherlands
| | - Daniele Bizzarri
- Department of Biomedical Data Sciences, Section of Molecular Epidemiology, Leiden University Medical Center, Leiden, The Netherlands
- Delft Bioinformatics Lab, Delft University of Technology, Delft, The Netherlands
| | - Erik B van den Akker
- Department of Biomedical Data Sciences, Section of Molecular Epidemiology, Leiden University Medical Center, Leiden, The Netherlands
- Delft Bioinformatics Lab, Delft University of Technology, Delft, The Netherlands
| | - Raymond Noordam
- Department of Internal Medicine, Section of Gerontology and Geriatrics, Leiden University Medical Center, PO box 9600, 2300 RC, Leiden, The Netherlands
| | - Joris Deelen
- Max Planck Institute for Biology of Ageing, Cologne, Germany
- Cologne Excellence Cluster On Cellular Stress Responses in Ageing-Associated Diseases (CECAD), University of Cologne, Cologne, Germany
| | - Diana van Heemst
- Department of Internal Medicine, Section of Gerontology and Geriatrics, Leiden University Medical Center, PO box 9600, 2300 RC, Leiden, The Netherlands
| | - Nienke A de Glas
- Department of Medical Oncology, Leiden University Medical Center, Leiden, The Netherlands
| | - Cynthia Holterhues
- Department of Internal Medicine, Haga Hospital, The Hague, The Netherlands
| | - Geert Labots
- Department of Internal Medicine, Haga Hospital, The Hague, The Netherlands
| | - Frederiek van den Bos
- Department of Internal Medicine, Section of Gerontology and Geriatrics, Leiden University Medical Center, PO box 9600, 2300 RC, Leiden, The Netherlands
| | - Marian Beekman
- Department of Biomedical Data Sciences, Section of Molecular Epidemiology, Leiden University Medical Center, Leiden, The Netherlands
| | - P Eline Slagboom
- Department of Biomedical Data Sciences, Section of Molecular Epidemiology, Leiden University Medical Center, Leiden, The Netherlands
| | - Barbara C van Munster
- Department of Internal Medicine, University Medical Center Groningen, Groningen, The Netherlands
| | | | - Stella Trompet
- Department of Internal Medicine, Section of Gerontology and Geriatrics, Leiden University Medical Center, PO box 9600, 2300 RC, Leiden, The Netherlands
| |
Collapse
|
27
|
Yang S, Liu R, Xin Z, Zhu Z, Chu J, Zhong P, Zhu Z, Shang X, Huang W, Zhang L, He M, Wang W. Plasma Metabolomics Identifies Key Metabolites and Improves Prediction of Diabetic Retinopathy: Development and Validation across Multinational Cohorts. Ophthalmology 2024; 131:1436-1446. [PMID: 38972358 DOI: 10.1016/j.ophtha.2024.07.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2024] [Revised: 05/13/2024] [Accepted: 07/01/2024] [Indexed: 07/09/2024] Open
Abstract
PURPOSE To identify longitudinal metabolomic fingerprints of diabetic retinopathy (DR) and to evaluate their usefulness in predicting DR development and progression. DESIGN Multicenter, multiethnic cohort study. PARTICIPANTS This study included 17 675 participants from the UK Biobank (UKB) who had baseline prediabetes or diabetes, identified in accordance with the 2021 American Diabetes Association guidelines, and were free of baseline DR and an additional 638 participants with type 2 diabetes mellitus from the Guangzhou Diabetic Eye Study (GDES) for external validation. Diabetic retinopathy was determined by ICD-10 codes in the UKB cohort and revised ETDRS grading criteria in the GDES cohort. METHODS Longitudinal DR metabolomic fingerprints were identified through nuclear magnetic resonance (NMR) assay in UKB participants. The predictive value of these fingerprints for predicting DR development were assessed in a fully withheld test set. External validation and extrapolation analyses of DR progression and microvascular damage were conducted in the GDES cohort using NMR technology. Model assessments included the concordance (C) statistic, net classification improvement (NRI), integrated discrimination improvement (IDI), calibration, and clinical usefulness in both cohorts. MAIN OUTCOME MEASURES DR development and progression and retinal microvascular damage. RESULTS Of 168 metabolites, 118 were identified as candidate metabolomic fingerprints for future DR development. These fingerprints significantly improved the predictability for DR development beyond traditional indicators (C statistic, 0.802 [95% confidence interval (CI), 0.760-0.843] vs. 0.751 [95% CI, 0.706-0.796]; P = 5.56 × 10-4). Glucose, lactate, and citrate were among the fingerprints validated in the GDES cohort. Using these parsimonious and replicable fingerprints yielded similar improvements for predicting DR development (C statistic, 0.807 [95% CI, 0.711-0.903] vs. 0.617 [95% CI, 0.494-0.740]; P = 1.68 × 10-4) and progression (C statistic, 0.797 [95% CI, 0.712-0.882] vs. 0.665 [95% CI, 0.545-0.784]; P = 0.003) in the external GDES cohort. Improvements in NRIs, IDIs, and clinical usefulness also were evident in both cohorts (all P < 0.05). In addition, lactate and citrate were associated with microvascular damage across macular and optic nerve head regions among Chinese GDES (all P < 0.05). CONCLUSIONS Metabolomic profiling may be effective in identifying robust fingerprints for predicting future DR development and progression, providing novel insights into the early and advanced stages of DR pathophysiology. FINANCIAL DISCLOSURE(S) The author(s) have no proprietary or commercial interest in any materials discussed in this article.
Collapse
Affiliation(s)
- Shaopeng Yang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangdong Provincial Clinical Research Center for Ocular Diseases, Guangzhou, China
| | - Riqian Liu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangdong Provincial Clinical Research Center for Ocular Diseases, Guangzhou, China
| | - Zhuoyao Xin
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, Maryland; F. M. Kirby Research Center for Functional Brain Imaging, Kennedy Krieger Institute, Baltimore, Maryland; Department of Biomedical Engineering, Columbia University, New York, New York
| | - Ziyu Zhu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangdong Provincial Clinical Research Center for Ocular Diseases, Guangzhou, China
| | - Jiaqing Chu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangdong Provincial Clinical Research Center for Ocular Diseases, Guangzhou, China
| | - Pingting Zhong
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangdong Provincial Clinical Research Center for Ocular Diseases, Guangzhou, China
| | - Zhuoting Zhu
- Centre for Eye Research Australia, Royal Victorian Eye and Ear Hospital, Melbourne, Australia
| | - Xianwen Shang
- Centre for Eye Research Australia, Royal Victorian Eye and Ear Hospital, Melbourne, Australia
| | - Wenyong Huang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangdong Provincial Clinical Research Center for Ocular Diseases, Guangzhou, China
| | - Lei Zhang
- Clinical Medical Research Center, Children's Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China; Artificial Intelligence and Modelling in Epidemiology Program, Melbourne Sexual Health Centre, Alfred Health, Melbourne, Australia; Central Clinical School, Faculty of Medicine, Nursing and Health Sciences, Monash University, Melbourne, Australia
| | - Mingguang He
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangdong Provincial Clinical Research Center for Ocular Diseases, Guangzhou, China; Experimental Ophthalmology, The Hong Kong Polytechnic University, Hong Kong, China
| | - Wei Wang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangdong Provincial Clinical Research Center for Ocular Diseases, Guangzhou, China; Hainan Eye Hospital and Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Haikou, Hainan Province, China.
| |
Collapse
|
28
|
Sebastiani P, Monti S, Lustgarten MS, Song Z, Ellis D, Tian Q, Schwaiger-Haber M, Stancliffe E, Leshchyk A, Short MI, Ardisson Korat AV, Gurinovich A, Karagiannis T, Li M, Lords HJ, Xiang Q, Marron MM, Bae H, Feitosa MF, Wojczynski MK, O'Connell JR, Montasser ME, Schupf N, Arbeev K, Yashin A, Schork N, Christensen K, Andersen SL, Ferrucci L, Rappaport N, Perls TT, Patti GJ. Metabolite signatures of chronological age, aging, survival, and longevity. Cell Rep 2024; 43:114913. [PMID: 39504246 PMCID: PMC11656345 DOI: 10.1016/j.celrep.2024.114913] [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: 09/22/2023] [Revised: 07/05/2024] [Accepted: 10/10/2024] [Indexed: 11/08/2024] Open
Abstract
Metabolites that mark aging are not fully known. We analyze 408 plasma metabolites in Long Life Family Study participants to characterize markers of age, aging, extreme longevity, and mortality. We identify 308 metabolites associated with age, 258 metabolites that change over time, 230 metabolites associated with extreme longevity, and 152 metabolites associated with mortality risk. We replicate many associations in independent studies. By summarizing the results into 19 signatures, we differentiate between metabolites that may mark aging-associated compensatory mechanisms from metabolites that mark cumulative damage of aging and from metabolites that characterize extreme longevity. We generate and validate a metabolomic clock that predicts biological age. Network analysis of the age-associated metabolites reveals a critical role of essential fatty acids to connect lipids with other metabolic processes. These results characterize many metabolites involved in aging and point to nutrition as a source of intervention for healthy aging therapeutics.
Collapse
Affiliation(s)
- Paola Sebastiani
- Institute for Clinical Research and Health Policy Studies, Tufts Medical Center, Boston, MA 02111, USA; Department of Medicine, School of Medicine, Tufts University, Boston, MA 02111, USA.
| | - Stefano Monti
- Department of Medicine, Chobanian & Avedisian School of Medicine, Boston University, Boston, MA 02118, USA; Bioinformatics Program, Boston University, Boston, MA 02215, USA
| | - Michael S Lustgarten
- Jean Mayer USDA Human Nutrition Research Center on Aging, Tufts University, Boston, MA 02111, USA
| | - Zeyuan Song
- Institute for Clinical Research and Health Policy Studies, Tufts Medical Center, Boston, MA 02111, USA
| | - Dylan Ellis
- Institute for Systems Biology, Seattle, WA 98109, USA
| | - Qu Tian
- Longitudinal Studies Section, Translational Gerontology Branch, National Institute on Aging, Baltimore, MD 21224, USA
| | | | - Ethan Stancliffe
- Department of Chemistry, Washington University in St. Louis, St. Louis, MO 63130, USA
| | | | - Meghan I Short
- Institute for Clinical Research and Health Policy Studies, Tufts Medical Center, Boston, MA 02111, USA; Department of Medicine, School of Medicine, Tufts University, Boston, MA 02111, USA
| | - Andres V Ardisson Korat
- Jean Mayer USDA Human Nutrition Research Center on Aging, Tufts University, Boston, MA 02111, USA
| | - Anastasia Gurinovich
- Institute for Clinical Research and Health Policy Studies, Tufts Medical Center, Boston, MA 02111, USA; Department of Medicine, School of Medicine, Tufts University, Boston, MA 02111, USA
| | - Tanya Karagiannis
- Institute for Clinical Research and Health Policy Studies, Tufts Medical Center, Boston, MA 02111, USA; Department of Medicine, School of Medicine, Tufts University, Boston, MA 02111, USA
| | - Mengze Li
- Bioinformatics Program, Boston University, Boston, MA 02215, USA
| | - Hannah J Lords
- Bioinformatics Program, Boston University, Boston, MA 02215, USA
| | - Qingyan Xiang
- Institute for Clinical Research and Health Policy Studies, Tufts Medical Center, Boston, MA 02111, USA
| | - Megan M Marron
- Department of Epidemiology, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Harold Bae
- Biostatistics Program, College of Health, Oregon State University, Corvallis, OR 97331, USA
| | - Mary F Feitosa
- Department of Genetics, Washington University School of Medicine, St. Louis, MO 63130, USA
| | - Mary K Wojczynski
- Department of Genetics, Washington University School of Medicine, St. Louis, MO 63130, USA
| | - Jeffrey R O'Connell
- Department of Medicine, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - May E Montasser
- Department of Medicine, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Nicole Schupf
- Department of Epidemiology, Columbia University Medical Center, New York, NY 10032, USA
| | - Konstantin Arbeev
- Social Science Research Institute, Duke University, Durham, NC 27708, USA
| | - Anatoliy Yashin
- Social Science Research Institute, Duke University, Durham, NC 27708, USA
| | - Nicholas Schork
- The Translational Genomics Research Institute, Phoenix, AZ 85004, USA
| | - Kaare Christensen
- Danish Aging Research Center, University of Southern Denmark, 5000 Odense, Denmark
| | - Stacy L Andersen
- Department of Medicine, Chobanian & Avedisian School of Medicine, Boston University, Boston, MA 02118, USA
| | - Luigi Ferrucci
- Longitudinal Studies Section, Translational Gerontology Branch, National Institute on Aging, Baltimore, MD 21224, USA
| | - Noa Rappaport
- Institute for Systems Biology, Seattle, WA 98109, USA
| | - Thomas T Perls
- Department of Medicine, Chobanian & Avedisian School of Medicine, Boston University, Boston, MA 02118, USA
| | - Gary J Patti
- Department of Chemistry, Washington University in St. Louis, St. Louis, MO 63130, USA
| |
Collapse
|
29
|
Huang K, G C de Sá A, Thomas N, Phair RD, Gooley PR, Ascher DB, Armstrong CW. Discriminating Myalgic Encephalomyelitis/Chronic Fatigue Syndrome and comorbid conditions using metabolomics in UK Biobank. COMMUNICATIONS MEDICINE 2024; 4:248. [PMID: 39592839 PMCID: PMC11599898 DOI: 10.1038/s43856-024-00669-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2023] [Accepted: 11/06/2024] [Indexed: 11/28/2024] Open
Abstract
BACKGROUND Diagnosing complex illnesses like Myalgic Encephalomyelitis/Chronic Fatigue Syndrome (ME/CFS) is complicated due to the diverse symptomology and presence of comorbid conditions. ME/CFS patients often present with multiple health issues, therefore, incorporating comorbidities into research can provide a more accurate understanding of the condition's symptomatology and severity, to better reflect real-life patient experiences. METHODS We performed association studies and machine learning on 1194 ME/CFS individuals with blood plasma nuclear magnetic resonance (NMR) metabolomics profiles, and seven exclusive comorbid cohorts: hypertension (n = 13,559), depression (n = 2522), asthma (n = 6406), irritable bowel syndrome (n = 859), hay fever (n = 3025), hypothyroidism (n = 1226), migraine (n = 1551) and a non-diseased control group (n = 53,009). RESULTS We present a lipoprotein perspective on ME/CFS pathophysiology, highlighting gender-specific differences and identifying overlapping associations with comorbid conditions, specifically surface lipids, and ketone bodies from 168 significant individual biomarker associations. Additionally, we searched for, trained, and optimised a machine learning algorithm, resulting in a predictive model using 19 baseline characteristics and nine NMR biomarkers which could identify ME/CFS with an AUC of 0.83 and recall of 0.70. A multi-variable score was subsequently derived from the same 28 features, which exhibited ~2.5 times greater association than the top individual biomarker. CONCLUSIONS This study provides an end-to-end analytical workflow that explores the potential clinical utility that association scores may have for ME/CFS and other difficult to diagnose conditions.
Collapse
Affiliation(s)
- Katherine Huang
- Department of Biochemistry and Pharmacology, Bio21 Molecular Science and Biotechnology Institute, University of Melbourne, Parkville, VIC, Australia
| | - Alex G C de Sá
- School of Chemistry and Molecular Biosciences, University of Queensland, Brisbane City, QLD, Australia
- Computational Biology and Clinical Informatics, Baker Heart and Diabetes Institute, Melbourne, VIC, Australia
- Baker Department of Cardiometabolic Health, University of Melbourne, Parkville, VIC, Australia
| | - Natalie Thomas
- Department of Biochemistry and Pharmacology, Bio21 Molecular Science and Biotechnology Institute, University of Melbourne, Parkville, VIC, Australia
| | - Robert D Phair
- Integrative Bioinformatics, Inc., Mountain View, CA, USA
| | - Paul R Gooley
- Department of Biochemistry and Pharmacology, Bio21 Molecular Science and Biotechnology Institute, University of Melbourne, Parkville, VIC, Australia
| | - David B Ascher
- School of Chemistry and Molecular Biosciences, University of Queensland, Brisbane City, QLD, Australia
- Computational Biology and Clinical Informatics, Baker Heart and Diabetes Institute, Melbourne, VIC, Australia
- Baker Department of Cardiometabolic Health, University of Melbourne, Parkville, VIC, Australia
| | - Christopher W Armstrong
- Department of Biochemistry and Pharmacology, Bio21 Molecular Science and Biotechnology Institute, University of Melbourne, Parkville, VIC, Australia.
| |
Collapse
|
30
|
Zhu Z, Lyu J, Hao X, Guo H, Zhang X, He M, Cheng X, Cheng S, Wang C. Estimation of physiological aging based on routine clinical biomarkers: a prospective cohort study in elderly Chinese and the UK Biobank. BMC Med 2024; 22:552. [PMID: 39578829 PMCID: PMC11583456 DOI: 10.1186/s12916-024-03769-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Accepted: 11/13/2024] [Indexed: 11/24/2024] Open
Abstract
BACKGROUND Chronological age (CA) does not reflect individual variation in the aging process. However, existing biological age predictors are mostly based on European populations and overlook the widespread nonlinear effects of clinical biomarkers. METHODS Using data from the prospective Dongfeng-Tongji (DFTJ) cohort of elderly Chinese, we propose a physiological aging index (PAI) based on 36 routine clinical biomarkers to measure aging progress. We first determined the optimal level of each biomarker by restricted cubic spline Cox models. For biomarkers with a U-shaped relationship with mortality, we derived new variables to model their distinct effects below and above the optimal levels. We defined PAI as a weighted sum of variables predictive of mortality selected by a LASSO Cox model. To measure aging acceleration, we defined ΔPAI as the residual of PAI after regressing on CA. We evaluated the predictive value of ΔPAI on cardiovascular diseases (CVD) in the DFTJ cohort, as well as nine major chronic diseases in the UK Biobank (UKB). RESULTS In the DFTJ training set (n = 12,769, median follow-up: 10.38 years), we identified 25 biomarkers with significant nonlinear associations with mortality, of which 11 showed insignificant linear associations. By incorporating nonlinear effects, we selected CA and 17 clinical biomarkers to calculate PAI. In the DFTJ testing set (n = 15,904, 5.87 years), PAI predict mortality with a concordance index (C-index) of 0.816 (95% confidence interval, [0.796, 0.837]), better than CA (C-index = 0.771 [0.755, 0.788]) and PhenoAge (0.799 [0.784, 0.814]). ΔPAI was predictive of incident CVD and its subtypes, independent of traditional risk factors. In the external validation set of UKB (n = 296,931, 12.80 years), PAI achieved a C-index of 0.749 (0.746, 0.752) to predict mortality, remaining better than CA (0.706 [0.702, 0.709]) and PhenoAge (0.743 [0.739, 0.746]). In both DFTJ and UKB, PAI calibrated better than PhenoAge when comparing the predicted and observed survival probabilities. Furthermore, ΔPAI outperformed any single biomarker to predict incident risks of eight age-related chronic diseases. CONCLUSIONS Our results highlight the potential of PAI and ΔPAI as integrative biomarkers to evaluate aging acceleration and facilitate the development of targeted intervention strategies for healthy aging.
Collapse
Affiliation(s)
- Ziwei Zhu
- Ministry of Education Key Laboratory of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Department of Epidemiology and Biostatistics, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jingjing Lyu
- Ministry of Education Key Laboratory of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Department of Epidemiology and Biostatistics, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xingjie Hao
- Ministry of Education Key Laboratory of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Department of Epidemiology and Biostatistics, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Huan Guo
- Ministry of Education Key Laboratory of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Department of Occupational and Environmental Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiaomin Zhang
- Ministry of Education Key Laboratory of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Department of Occupational and Environmental Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Meian He
- Ministry of Education Key Laboratory of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Department of Occupational and Environmental Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiang Cheng
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Shanshan Cheng
- Ministry of Education Key Laboratory of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
- Department of Epidemiology and Biostatistics, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - Chaolong Wang
- Ministry of Education Key Laboratory of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
- Department of Epidemiology and Biostatistics, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| |
Collapse
|
31
|
Nightingale Health Biobank Collaborative Group, Barrett JC, Esko T, Fischer K, Jostins-Dean L, Jousilahti P, Julkunen H, Jääskeläinen T, Kangas A, Kerimov N, Kerminen S, Kolde A, Koskela H, Kronberg J, Lundgren SN, Lundqvist A, Mäkelä V, Nybo K, Perola M, Salomaa V, Schut K, Soikkeli M, Soininen P, Tiainen M, Tillmann T, Würtz P. Metabolomic and genomic prediction of common diseases in 700,217 participants in three national biobanks. Nat Commun 2024; 15:10092. [PMID: 39572536 PMCID: PMC11582662 DOI: 10.1038/s41467-024-54357-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2023] [Accepted: 11/08/2024] [Indexed: 11/24/2024] Open
Abstract
Identifying individuals at high risk of chronic diseases via easily measured biomarkers could enhance efforts to prevent avoidable illness and death. Using 'omic data can stratify risk for many diseases simultaneously from a single measurement that captures multiple molecular predictors of risk. Here we present nuclear magnetic resonance metabolomics in blood samples from 700,217 participants in three national biobanks. We built metabolomic scores that identify high-risk groups for diseases that cause the most morbidity in high-income countries and show consistent cross-biobank replication of the relative risk of disease for these groups. We show that these metabolomic scores are more strongly associated with disease onset than polygenic scores for most of these diseases. In a subset of 18,709 individuals with metabolomic biomarkers measured at two time points we show that people whose scores change have different risk of disease, suggesting that repeat measurements capture changes both to health status and disease risk possibly due to treatment, lifestyle changes or other factors. Lastly, we assessed the incremental predictive value of metabolomic scores over existing clinical risk scores for multiple diseases and found modest improvements in discrimination for several diseases whose clinical utility, while promising, remains to be determined.
Collapse
|
32
|
Pimentel E, Banoei MM, Kaur J, Lee CH, Winston BW. Metabolomic Insights into COVID-19 Severity: A Scoping Review. Metabolites 2024; 14:617. [PMID: 39590853 PMCID: PMC11596841 DOI: 10.3390/metabo14110617] [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: 09/28/2024] [Revised: 10/29/2024] [Accepted: 11/07/2024] [Indexed: 11/28/2024] Open
Abstract
BACKGROUND In 2019, SARS-CoV-2, the novel coronavirus, entered the world scene, presenting a global health crisis with a broad spectrum of clinical manifestations. Recognizing the significance of metabolomics as the omics closest to symptomatology, it has become a useful tool for predicting clinical outcomes. Several metabolomic studies have indicated variations in the metabolome corresponding to different disease severities, highlighting the potential of metabolomics to unravel crucial insights into the pathophysiology of SARS-CoV-2 infection. METHODS The PRISMA guidelines were followed for this scoping review. Three major scientific databases were searched: PubMed, the Directory of Open Access Journals (DOAJ), and BioMed Central, from 2020 to 2024. Initially, 2938 articles were identified and vetted with specific inclusion and exclusion criteria. Of these, 42 articles were retrieved for analysis and summary. RESULTS Metabolites were identified that were repeatedly noted to change with COVID-19 and its severity. Phenylalanine, glucose, and glutamic acid increased with severity, while tryptophan, proline, and glutamine decreased, highlighting their association with COVID-19 severity. Additionally, pathway analysis revealed that phenylalanine, tyrosine and tryptophan biosynthesis, and arginine biosynthesis were the most significantly impacted pathways in COVID-19 severity. CONCLUSIONS COVID-19 severity is intricately linked to significant metabolic alterations that span amino acid metabolism, energy production, immune response modulation, and redox balance.
Collapse
Affiliation(s)
- Eric Pimentel
- Department of Critical Care, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 4Z6, Canada; (E.P.); (M.M.B.); (J.K.); (C.H.L.)
| | - Mohammad Mehdi Banoei
- Department of Critical Care, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 4Z6, Canada; (E.P.); (M.M.B.); (J.K.); (C.H.L.)
- Department of Biological Sciences, University of Calgary, Calgary, AB T2N 4Z6, Canada
| | - Jasnoor Kaur
- Department of Critical Care, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 4Z6, Canada; (E.P.); (M.M.B.); (J.K.); (C.H.L.)
| | - Chel Hee Lee
- Department of Critical Care, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 4Z6, Canada; (E.P.); (M.M.B.); (J.K.); (C.H.L.)
- Department of Mathematics and Statistics, Faculty of Science, University of Calgary, Calgary, AB T2N 5A1, Canada
| | - Brent W. Winston
- Department of Critical Care, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 4Z6, Canada; (E.P.); (M.M.B.); (J.K.); (C.H.L.)
- Departments of Medicine, Biochemistry and Molecular Biology, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 4Z6, Canada
| |
Collapse
|
33
|
Debik J, Mrowiec K, Kurczyk A, Widłak P, Jelonek K, Bathen TF, Giskeødegård GF. Sources of variation in the serum metabolome of female participants of the HUNT2 study. Commun Biol 2024; 7:1450. [PMID: 39506131 PMCID: PMC11541904 DOI: 10.1038/s42003-024-07137-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Accepted: 10/24/2024] [Indexed: 11/08/2024] Open
Abstract
The aim of this study was to explore the intricate relationship between serum metabolomics and lifestyle factors, shedding light on their impact on health in the context of breast cancer risk. Detailed metabolic profiles of 2283 female participants in the Trøndelag Health Study (HUNT study) were obtained through nuclear magnetic resonance (NMR) spectroscopy and mass spectrometry (MS).We show that lifestyle-related variables can explain up to 30% of the variance in individual metabolites. Age and obesity were the primary factors affecting the serum metabolic profile, both associated with increased levels of triglyceride-rich very low-density lipoproteins (VLDL) and intermediate-density lipoproteins (IDL), amino acids and glycolysis-related metabolites, and decreased levels of high-density lipoproteins (HDL). Moreover, factors like hormonal changes associated with menstruation and contraceptive use or education level influence the metabolite levels.Participants were clustered into three distinct clusters based on lifestyle-related factors, revealing metabolic similarities between obese and older individuals, despite diverse lifestyle factors, suggesting accelerated metabolic aging with obesity. Our results show that metabolic associations to cancer risk may partly be explained by modifiable lifestyle factors.
Collapse
Affiliation(s)
- Julia Debik
- Department of Public Health and Nursing, Norwegian University of Science and Technology, Trondheim, Norway.
| | - Katarzyna Mrowiec
- Center for Translational Research and Molecular Biology of Cancer, Maria Skłodowska-Curie National Research Institute of Oncology, Gliwice Branch, Gliwice, Poland
| | - Agata Kurczyk
- Department of Biostatistics and Bioinformatics, Maria Skłodowska-Curie National Research Institute of Oncology, Gliwice Branch, Gliwice, Poland
| | - Piotr Widłak
- 2nd Radiology Department, Medical University of Gdańsk, Gdańsk, Poland
| | - Karol Jelonek
- Center for Translational Research and Molecular Biology of Cancer, Maria Skłodowska-Curie National Research Institute of Oncology, Gliwice Branch, Gliwice, Poland
| | - Tone F Bathen
- Department of Circulation and Medical Imaging, Norwegian University of Science and Technology, Trondheim, Norway
- Department of Radiology and Nuclear Medicine, St. Olav's University Hospital, Trondheim, Norway
| | - Guro F Giskeødegård
- Department of Public Health and Nursing, Norwegian University of Science and Technology, Trondheim, Norway.
- Clinic of Surgery, St. Olav's University Hospital, Trondheim, Norway.
| |
Collapse
|
34
|
Smit AP, Herber GCM, Kuiper LM, Rietman ML, Wesenhagen KEJ, Picavet HSJ, Slagboom PE, Verschuren WMM. Association between metabolomics-based biomarker scores and 10-year cognitive decline in men and women. The Doetinchem Cohort Study. Age Ageing 2024; 53:afae256. [PMID: 39558869 PMCID: PMC11574050 DOI: 10.1093/ageing/afae256] [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: 05/15/2024] [Revised: 08/22/2024] [Indexed: 11/20/2024] Open
Abstract
BACKGROUND Metabolomic scores based on age (MetaboAge) and mortality (MetaboHealth) are considered indicators of overall health, but their association with cognition in the general population is unknown. Therefore, the association between MetaboAge/MetaboHealth and level and decline in cognition was studied, as were differences between men and women. METHODS Data of 2821 participants (50% women, age range 45-75) from the Doetinchem Cohort Study was used. MetaboAge and MetaboHealth were calculated from 1H-NMR metabolomics data at baseline. Cognitive domain scores (memory, flexibility and processing speed) and global cognitive functioning were available over a 10-year period. The association between MetaboAge/MetaboHealth and level of cognitive functioning was studied using linear regressions while for the association between MetaboAge/MetaboHealth and cognitive decline longitudinal linear mixed models were used. Analyses were adjusted for demographics and lifestyle factors. RESULTS Higher MetaboAge, indicating poorer metabolomic ageing, was only associated with lower levels of processing speed in men. Higher MetaboHealth, indicating poorer immune-metabolic health, was associated with lower levels of cognitive functioning for all three domains and global cognitive functioning in both men and women. Only in men, MetaboHealth was also associated with 10-year decline in flexibility, processing speed and global cognition. Metabolites that contributed to the observed associations were in men mainly markers of protein metabolism, and in women mainly markers of lipid metabolism and inflammatory metabolites. CONCLUSIONS MetaboHealth, not MetaboAge, was associated with cognitive functioning independent of conventional risk factors. Individual metabolites affect cognitive functioning differently in men and women, suggesting sex-specific pathophysiological pathways underlying cognitive functioning.
Collapse
Affiliation(s)
- Annelot P Smit
- National Institute for Public Health and the Environment, Center for Prevention, Lifestyle and Health, Bilthoven, The Netherlands
- Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Gerrie-Cor M Herber
- National Institute for Public Health and the Environment, Center for Prevention, Lifestyle and Health, Bilthoven, The Netherlands
| | - Lieke M Kuiper
- National Institute for Public Health and the Environment, Center for Prevention, Lifestyle and Health, Bilthoven, The Netherlands
- Department of Internal Medicine, Erasmus MC, University Medical Center, Rotterdam, The Netherlands
| | - M Liset Rietman
- National Institute for Public Health and the Environment, Center for Prevention, Lifestyle and Health, Bilthoven, The Netherlands
| | - Kirsten E J Wesenhagen
- National Institute for Public Health and the Environment, Center for Prevention, Lifestyle and Health, Bilthoven, The Netherlands
| | - H Susan J Picavet
- National Institute for Public Health and the Environment, Center for Prevention, Lifestyle and Health, Bilthoven, The Netherlands
| | - P Eline Slagboom
- Molecular Epidemiology, Department of Biomedical Data Sciences, Leiden University Medical Centre, Leiden, The Netherlands
- Max Planck Institute for the Biology of Ageing, Cologne, Germany
| | - W M Monique Verschuren
- National Institute for Public Health and the Environment, Center for Prevention, Lifestyle and Health, Bilthoven, The Netherlands
- Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| |
Collapse
|
35
|
van Holstein Y, Trompet S, van Munster BC, van den Berkmortel PJE, van Heemst D, de Glas NA, Slingerland M, Slagboom PE, Holterhues C, Labots G, Mooijaart SP, Portielje JEA, van den Bos F. Association of Glasgow Prognostic Score with frailty, mortality and adverse health outcomes in older patients with cancer: A prospective cohort study. J Geriatr Oncol 2024; 15:102075. [PMID: 39414486 DOI: 10.1016/j.jgo.2024.102075] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2024] [Revised: 09/09/2024] [Accepted: 09/25/2024] [Indexed: 10/18/2024]
Abstract
INTRODUCTION To balance benefits and risks of cancer treatment in older patients, prognostic information is needed. The Glasgow Prognostic Score (GPS), composed of albumin and C-reactive protein (CRP), might provide such information. This study first aims to investigate the association between GPS and frailty, functional decline, and health-related quality of life (HRQoL) decline as indicators of health problems in older patients with cancer. The second aim is to study the predictive value of GPS for mortality, in addition to clinical predictors. MATERIALS AND METHODS This prospective cohort study included patients aged ≥70 years with a solid malignant tumor who underwent a geriatric assessment and blood sampling before treatment initiation. GPS was calculated using serum albumin and CRP measured in batch, categorized into normal (0) and abnormal GPS (1-2). Outcomes were all-cause mortality and a composite outcome of decline in daily functioning and/or HRQoL, or mortality at one year follow-up. Daily functioning was assessed by Activities of Daily Living and Instrumental Activities of Daily Living questionnaires and HRQoL by the EQ-5D-3L and EQ-VAS questionnaires. RESULTS In total, 192 patients with a median age of 77 years (interquartile range 72.3-81.0) were included. Patients with abnormal GPS were more often frail compared to those with normal GPS (79 % vs. 63 %, p = 0.03). Patients with abnormal GPS had higher mortality rates after one year compared to those with normal GPS (48 % vs. 23 %, p < 0.01) in unadjusted analysis. Abnormal GPS was associated with increased mortality risk (hazard ratio 2.8, 95 % CI 1.7-4.8). The area under the receiver operating characteristics curve of age, distant metastasis, tumor site, comorbidity, and malnutrition combined was 0.73 (0.68-0.83) for mortality prediction, and changed to 0.78 (0.73-0.86) with GPS (p = 0.10). The composite outcome occurred in 88 % of patients with abnormal GPS versus 83 % with normal GPS (p = 0.44). DISCUSSION Abnormal GPS was associated with frailty and mortality. The addition of GPS to clinical predictors showed a numerically superior mortality prediction in this cohort of older patients with cancer, although not statistically significant. While GPS may improve the stratification of future older patients with cancer, larger studies including older patients with similar tumor types are necessary to evaluate its clinical usefulness. TRIAL REGISTRATION The TENT study is retrospectively registered at the Netherlands Trial Register (NTR), trial number NL8107. Date of registration: 22-10-2019.
Collapse
Affiliation(s)
- Yara van Holstein
- Department of Internal Medicine, Section of Gerontology and Geriatrics, Leiden University Medical Center, the Netherlands.
| | - Stella Trompet
- Department of Internal Medicine, Section of Gerontology and Geriatrics, Leiden University Medical Center, the Netherlands
| | - Barbara C van Munster
- Department of Internal Medicine, University Medical Center Groningen, the Netherlands
| | - P Janne E van den Berkmortel
- Department of Internal Medicine, Section of Gerontology and Geriatrics, Leiden University Medical Center, the Netherlands
| | - Diana van Heemst
- Department of Internal Medicine, Section of Gerontology and Geriatrics, Leiden University Medical Center, the Netherlands
| | - Nienke A de Glas
- Department of Medical Oncology, Leiden University Medical Center, the Netherlands
| | - Marije Slingerland
- Department of Medical Oncology, Leiden University Medical Center, the Netherlands
| | - P Eline Slagboom
- Department of Biomedical Data Sciences, Section of Molecular Epidemiology, Leiden University Medical Center, the Netherlands
| | - Cynthia Holterhues
- Department of Internal Medicine, Haga Hospital, The Hague, the Netherlands
| | - Geert Labots
- Department of Internal Medicine, Haga Hospital, The Hague, the Netherlands
| | - Simon P Mooijaart
- Department of Internal Medicine, Section of Gerontology and Geriatrics, Leiden University Medical Center, the Netherlands; LUMC Center for Medicine for Older People, Leiden University Medical Center, Leiden, the Netherlands
| | | | - Frederiek van den Bos
- Department of Internal Medicine, Section of Gerontology and Geriatrics, Leiden University Medical Center, the Netherlands
| |
Collapse
|
36
|
Li Y, Wang H, Xiao Y, Yang H, Wang S, Liu L, Cai H, Zhang X, Tang H, Wu T, Qiu G. Lipidomics identified novel cholesterol-independent predictors for risk of incident coronary heart disease: Mediation of risk from diabetes and aggravation of risk by ambient air pollution. J Adv Res 2024; 65:273-282. [PMID: 38104795 PMCID: PMC11519734 DOI: 10.1016/j.jare.2023.12.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Revised: 09/16/2023] [Accepted: 12/10/2023] [Indexed: 12/19/2023] Open
Abstract
INTRODUCTION Previous lipidomics studies have identified various lipid predictors for cardiovascular risk, however, with limited predictive increment, sometimes using too many predictor variables at the expense of practical efficiency. OBJECTIVES To search for lipid predictors of future coronary heart disease (CHD) with stronger predictive power and efficiency to guide primary intervention. METHODS We conducted a prospective nested case-control study involving 1,621 incident CHD cases and 1:1 matched controls. Lipid profiling of 161 lipid species for baseline fasting plasma was performed by liquid chromatography-mass spectrometry. RESULTS In search of CHD predictors, seven lipids were selected by elastic-net regression during over 90% of 1000 cross-validation repetitions, and the derived composite lipid score showed an adjusted odds ratio of 3.75 (95% confidence interval: 3.15, 4.46) per standard deviation increase. Addition of the lipid score into traditional risk model increased c-statistic to 0.736 by an increment of 0.077 (0.063, 0.092). From the seven lipids, we found mediation of CHD risk from baseline diabetes through sphingomyelin (SM) 41:1b with a considerable mediation proportion of 36.97% (P < 0.05). We further found that the positive associations of phosphatidylcholine (PC) 36:0a, SM 41:1b, lysophosphatidylcholine (LPC) 18:0 and LPC 20:3 were more pronounced among participants with higher exposure to fine particulate matter or its certain components, also to ozone for LPC 18:0 and LPC 20:3, while the negative association of cholesteryl ester (CE) 18:2 was attenuated with higher black carbon exposure (P < 0.05). CONCLUSION We identified seven lipid species with greatest predictive increment so-far achieved for incident CHD, and also found novel biomarkers for CHD risk stratification among individuals with diabetes or heavy air pollution exposure.
Collapse
Affiliation(s)
- Yingmei Li
- Ministry of Education and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Hao Wang
- Ministry of Education and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Yang Xiao
- Ministry of Education and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Handong Yang
- Department of Cardiovascular Disease, Sinopharm Dongfeng General Hospital, Hubei University of Medicine, Shiyan, China
| | - Sihan Wang
- Ministry of Education and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Ling Liu
- Ministry of Education and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Hao Cai
- Ministry of Education and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Xiaomin Zhang
- Ministry of Education and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Huiru Tang
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Metabonomics and Systems Biology Laboratory at Shanghai International Centre for Molecular Phenomics, Human Phenome Institute, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Tangchun Wu
- Ministry of Education and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China.
| | - Gaokun Qiu
- Ministry of Education and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China.
| |
Collapse
|
37
|
Jia X, Fan J, Wu X, Cao X, Ma L, Abdelrahman Z, Zhao F, Zhu H, Bizzarri D, Akker EBVD, Slagboom PE, Deelen J, Zhou D, Liu Z. A Novel Metabolomic Aging Clock Predicting Health Outcomes and Its Genetic and Modifiable Factors. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2406670. [PMID: 39331845 DOI: 10.1002/advs.202406670] [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: 06/16/2024] [Revised: 08/22/2024] [Indexed: 09/29/2024]
Abstract
Existing metabolomic clocks exhibit deficiencies in capturing the heterogeneous aging rates among individuals with the same chronological age. Yet, the modifiable and non-modifiable factors in metabolomic aging have not been systematically studied. Here, a new aging measure-MetaboAgeMort-is developed using metabolomic profiles from 239,291 UK Biobank participants for 10-year all-cause mortality prediction. The MetaboAgeMort showed significant associations with all-cause mortality, cause-specific mortality, and diverse incident diseases. Adding MetaboAgeMort to a conventional risk factors model improved the predictive ability of 10-year mortality. A total of 99 modifiable factors across seven categories are identified for MetaboAgeMort. Among these, 16 factors representing pulmonary function, body composition, socioeconomic status, dietary quality, smoking status, alcohol intake, and disease status showed quantitatively stronger associations. The genetic analyses revealed 99 genomic risk loci and 271 genes associated with MetaboAgeMort. The tissue-enrichment analysis showed significant enrichment in liver. While the external validation of the MetaboAgeMort is required, this study illuminates heterogeneous metabolomic aging across the same age, providing avenues for identifying high-risk individuals, developing anti-aging therapies, and personalizing interventions, thus promoting healthy aging and longevity.
Collapse
Affiliation(s)
- Xueqing Jia
- Center for Clinical Big Data and Analytics of the Second Affiliated Hospital and Department of Big Data in Health Science School of Public Health, Zhejiang Key Laboratory of Intelligent Preventive Medicine, Zhejiang University School of Medicine, Hangzhou, 310058, China
| | - Jiayao Fan
- Center for Clinical Big Data and Analytics of the Second Affiliated Hospital and Department of Big Data in Health Science School of Public Health, Zhejiang Key Laboratory of Intelligent Preventive Medicine, Zhejiang University School of Medicine, Hangzhou, 310058, China
| | - Xucheng Wu
- Center for Clinical Big Data and Analytics of the Second Affiliated Hospital and Department of Big Data in Health Science School of Public Health, Zhejiang Key Laboratory of Intelligent Preventive Medicine, Zhejiang University School of Medicine, Hangzhou, 310058, China
| | - Xingqi Cao
- Center for Clinical Big Data and Analytics of the Second Affiliated Hospital and Department of Big Data in Health Science School of Public Health, Zhejiang Key Laboratory of Intelligent Preventive Medicine, Zhejiang University School of Medicine, Hangzhou, 310058, China
- Department of General Practice, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, 310016, China
| | - Lina Ma
- Department of Geriatrics, National Clinical Research Center for Geriatric Medicine, Xuanwu Hospital, Capital Medical University, Beijing, 100053, China
| | - Zeinab Abdelrahman
- Molecular Epidemiology and Public Health Research Group, Centre for Public Health, Queen's University Belfast, Institute for Clinical Sciences A, Royal Victoria Hospital, Belfast, BT12 6BA, UK
| | - Fei Zhao
- Hangzhou Meilian Medical Co., Ltd., Hangzhou, 311200, China
| | - Haitao Zhu
- Hangzhou Meilian Medical Co., Ltd., Hangzhou, 311200, China
| | - Daniele Bizzarri
- Molecular Epidemiology, Department of Biomedical Data Sciences, Leiden University Medical Center, Leiden, 2333 ZC, The Netherlands
- The Delft Bioinformatics Lab, Pattern Recognition & Bioinformatics, Delft University of Technology, Delft, 2628 CC, The Netherlands
| | - Erik B van den Akker
- Molecular Epidemiology, Department of Biomedical Data Sciences, Leiden University Medical Center, Leiden, 2333 ZC, The Netherlands
- The Delft Bioinformatics Lab, Pattern Recognition & Bioinformatics, Delft University of Technology, Delft, 2628 CC, The Netherlands
| | - P Eline Slagboom
- Molecular Epidemiology, Department of Biomedical Data Sciences, Leiden University Medical Center, Leiden, 2333 ZC, The Netherlands
| | - Joris Deelen
- Max Planck Institute for Biology of Ageing, 50931, Cologne, Germany
- Cologne Excellence Cluster on Cellular Stress Responses in Ageing-Associated Diseases (CECAD), University of Cologne, 50931, Cologne, Germany
| | - Dan Zhou
- Center for Clinical Big Data and Analytics of the Second Affiliated Hospital and Department of Big Data in Health Science School of Public Health, Zhejiang Key Laboratory of Intelligent Preventive Medicine, Zhejiang University School of Medicine, Hangzhou, 310058, China
| | - Zuyun Liu
- Center for Clinical Big Data and Analytics of the Second Affiliated Hospital and Department of Big Data in Health Science School of Public Health, Zhejiang Key Laboratory of Intelligent Preventive Medicine, Zhejiang University School of Medicine, Hangzhou, 310058, China
| |
Collapse
|
38
|
Oravilahti A, Vangipurapu J, Laakso M, Fernandes Silva L. Metabolomics-Based Machine Learning for Predicting Mortality: Unveiling Multisystem Impacts on Health. Int J Mol Sci 2024; 25:11636. [PMID: 39519188 PMCID: PMC11546733 DOI: 10.3390/ijms252111636] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2024] [Revised: 10/22/2024] [Accepted: 10/29/2024] [Indexed: 11/16/2024] Open
Abstract
Reliable predictors of long-term all-cause mortality are needed for middle-aged and older populations. Previous metabolomics mortality studies have limitations: a low number of participants and metabolites measured, measurements mainly using nuclear magnetic spectroscopy, and the use only of conventional statistical methods. To overcome these challenges, we applied liquid chromatography-tandem mass spectrometry and measured >1000 metabolites in the METSIM study including 10,197 men. We applied the machine learning approach together with conventional statistical methods to identify metabolites associated with all-cause mortality. The three independent machine learning methods (logistic regression, XGBoost, and Welch's t-test) identified 32 metabolites having the most impactful associations with all-cause mortality (25 increasing and 7 decreasing the risk). From these metabolites, 20 were novel and encompassed various metabolic pathways, impacting the cardiovascular, renal, respiratory, endocrine, and central nervous systems. In the Cox regression analyses (hazard ratios and their 95% confidence intervals), clinical and laboratory risk factors increased the risk of all-cause mortality by 1.76 (1.60-1.94), the 25 metabolites by 1.89 (1.68-2.12), and clinical and laboratory risk factors combined with the 25 metabolites by 2.00 (1.81-2.22). In our study, the main causes of death were cancers (28%) and cardiovascular diseases (25%). We did not identify any metabolites associated with cancer but found 13 metabolites associated with an increased risk of cardiovascular diseases. Our study reports several novel metabolites associated with an increased risk of mortality and shows that these 25 metabolites improved the prediction of all-cause mortality beyond and above clinical and laboratory measurements.
Collapse
Affiliation(s)
- Anniina Oravilahti
- Institute of Clinical Medicine, Internal Medicine, University of Eastern Finland, 70210 Kuopio, Finland; (A.O.); (J.V.); (M.L.)
| | - Jagadish Vangipurapu
- Institute of Clinical Medicine, Internal Medicine, University of Eastern Finland, 70210 Kuopio, Finland; (A.O.); (J.V.); (M.L.)
| | - Markku Laakso
- Institute of Clinical Medicine, Internal Medicine, University of Eastern Finland, 70210 Kuopio, Finland; (A.O.); (J.V.); (M.L.)
- Department of Medicine, Kuopio University Hospital, 70200 Kuopio, Finland
| | - Lilian Fernandes Silva
- Institute of Clinical Medicine, Internal Medicine, University of Eastern Finland, 70210 Kuopio, Finland; (A.O.); (J.V.); (M.L.)
- Department of Medicine, Division of Cardiology, David Geffen School of Medicine, University of California, Los Angeles, CA 90095, USA
| |
Collapse
|
39
|
Xin Q, Liang X, Yang J, Wang X, Hu F, Jiang M, Liu Y, Gong J, Pan Y, Liu L, Xu J, Cui Y, Qin H, Bai H, Li Y, Ma J, Zhang C, Shi B. Metabolomic alterations in the plasma of patients with various clinical manifestations of COVID-19. Virol J 2024; 21:266. [PMID: 39468659 PMCID: PMC11520427 DOI: 10.1186/s12985-024-02523-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2024] [Accepted: 09/27/2024] [Indexed: 10/30/2024] Open
Abstract
BACKGROUND The metabolomic profiles of individuals with different clinical manifestations of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection have not been clearly characterized. METHODS We performed metabolomics analysis of 166 individuals, including 62 healthy controls, 16 individuals with asymptomatic SARS-CoV-2 infection, and 88 patients with moderate (n = 42) and severe (n = 46) symptomatic 2019 coronavirus disease (COVID-19; 17 with short-term and 34 with long-term nucleic-acid test positivity). By examining differential expression, we identified candidate metabolites associated with different SARS-CoV-2 infection presentations. Functional and machine learning analyses were performed to explore the metabolites' functions and verify their candidacy as biomarkers. RESULTS A total of 417 metabolites were detected. We discovered 70 differentially expressed metabolites that may help differentiate asymptomatic infections from healthy controls and COVID-19 patients with different disease severity. Cyclamic acid and N-Acetylneuraminic Acid were identified to distinguish symptomatic infected patients and asymptomatic infected patients. Shikimic Acid, Glycyrrhetinic acid and 3-Hydroxybutyrate can supply significant insights for distinguishing short-term and long-term nucleic-acid test positivity. CONCLUSION Metabolomic profiling may highlight novel biomarkers for the identification of individuals with asymptomatic SARS-CoV-2 infection and further our understanding of the molecular pathogenesis of COVID-19.
Collapse
Affiliation(s)
- Qi Xin
- Precision Medicine Center, The First Affiliated Hospital of Xi'an Jiaotong University, 277 Yanta West Road, Xi'an, 710061, China
| | - Xiao Liang
- Precision Medicine Center, The First Affiliated Hospital of Xi'an Jiaotong University, 277 Yanta West Road, Xi'an, 710061, China
- Cancer Center, The First Affiliated Hospital of Xi'an Jiaotong University, 277 Yanta West Road, Xi'an, 710061, China
| | - Jin Yang
- Precision Medicine Center, The First Affiliated Hospital of Xi'an Jiaotong University, 277 Yanta West Road, Xi'an, 710061, China
- Cancer Center, The First Affiliated Hospital of Xi'an Jiaotong University, 277 Yanta West Road, Xi'an, 710061, China
- Department of Medical Oncology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Xiaorui Wang
- Precision Medicine Center, The First Affiliated Hospital of Xi'an Jiaotong University, 277 Yanta West Road, Xi'an, 710061, China
| | - Fang Hu
- Precision Medicine Center, The First Affiliated Hospital of Xi'an Jiaotong University, 277 Yanta West Road, Xi'an, 710061, China
| | - Meng Jiang
- Precision Medicine Center, The First Affiliated Hospital of Xi'an Jiaotong University, 277 Yanta West Road, Xi'an, 710061, China
| | - Yijia Liu
- Precision Medicine Center, The First Affiliated Hospital of Xi'an Jiaotong University, 277 Yanta West Road, Xi'an, 710061, China
| | - Jin Gong
- Precision Medicine Center, The First Affiliated Hospital of Xi'an Jiaotong University, 277 Yanta West Road, Xi'an, 710061, China
- Cancer Center, The First Affiliated Hospital of Xi'an Jiaotong University, 277 Yanta West Road, Xi'an, 710061, China
| | - Yiwen Pan
- Precision Medicine Center, The First Affiliated Hospital of Xi'an Jiaotong University, 277 Yanta West Road, Xi'an, 710061, China
| | - Lijuan Liu
- Precision Medicine Center, The First Affiliated Hospital of Xi'an Jiaotong University, 277 Yanta West Road, Xi'an, 710061, China
| | - Jiao Xu
- Precision Medicine Center, The First Affiliated Hospital of Xi'an Jiaotong University, 277 Yanta West Road, Xi'an, 710061, China
| | - Yuxin Cui
- Precision Medicine Center, The First Affiliated Hospital of Xi'an Jiaotong University, 277 Yanta West Road, Xi'an, 710061, China
| | - Hongyu Qin
- Precision Medicine Center, The First Affiliated Hospital of Xi'an Jiaotong University, 277 Yanta West Road, Xi'an, 710061, China
| | - Han Bai
- The MED-X Institute, The First Affiliated Hospital of Xi'an Jiaotong University, Building 21, Western China Science and Technology Innovation Harbor, Xi'an, 710000, China
| | - Yixin Li
- Precision Medicine Center, The First Affiliated Hospital of Xi'an Jiaotong University, 277 Yanta West Road, Xi'an, 710061, China
- The MED-X Institute, The First Affiliated Hospital of Xi'an Jiaotong University, Building 21, Western China Science and Technology Innovation Harbor, Xi'an, 710000, China
| | - Junpeng Ma
- Precision Medicine Center, The First Affiliated Hospital of Xi'an Jiaotong University, 277 Yanta West Road, Xi'an, 710061, China
| | - Chengsheng Zhang
- Precision Medicine Center, The First Affiliated Hospital of Xi'an Jiaotong University, 277 Yanta West Road, Xi'an, 710061, China.
- The MED-X Institute, The First Affiliated Hospital of Xi'an Jiaotong University, Building 21, Western China Science and Technology Innovation Harbor, Xi'an, 710000, China.
| | - Bingyin Shi
- Department of Endocrinology, The First Affiliated Hospital of Xi'an Jiaotong University, 277 Yanta West Road, Xi'an, 710061, China.
| |
Collapse
|
40
|
Wang RZ, Zhang WS, Jiang CQ, Zhu F, Jin YL, Xu L. Inflammatory age and its impact on age-related health in older Chinese adults. Arch Gerontol Geriatr 2024; 125:105476. [PMID: 38761528 DOI: 10.1016/j.archger.2024.105476] [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: 02/23/2024] [Revised: 03/27/2024] [Accepted: 05/05/2024] [Indexed: 05/20/2024]
Abstract
INTRODUCTION A standardized measure for inflammaging is lacking. We introduced the inflammatory age (iAge) as a quantification method and explored its associations with age-related traits and diseases in an older Chinese cohort. METHODS Inflammatory markers including white blood cell count (WBC), neutrophils, lymphocytes, monocytes, C-reactive protein, platelets and albumin were measured. Quantitative real-time polymerase chain reaction was used to measure telomere length. Traditional multivariable linear, partial least squares, and logistic regression were used. RESULTS iAge was constructed based on WBC, neutrophils, monocytes and albumin, which were associated with telomere length independently. A higher iAge indicated a heavier aging-related inflammation burden. Per 1-year increase in iAge was associated with higher body mass index (β 0.86 (95 % CI 0.67, 1.05) kg/m2), waist circumference (β 2.37 (95 % CI 1.85, 2.90) cm), glycosylated hemoglobin A1c (β 0.06 (95 % CI 0.02, 0.10) %), systolic blood pressure (β 1.06 (95 % CI 0.10, 2.03) mmHg), triglycerides (β 0.05 (95 % CI 0.01, 0.08) mmol/L), 10-year cardiovascular diseases risk (β 0.05 (95 % CI 0.02, 0.08) %), diabetes (OR 1.22 (95 % CI 1.02, 1.46)), hypertension (OR 1.21 (95 % CI 1.04, 1.42)) and metabolic syndrome risks (OR 1.25 (95 % CI 1.04, 1.51)), and lower fasting plasma glucose (β -0.016 (95 % CI -0.024, -0.007) mmol/L), total cholesterol (β -0.06 (95 % CI -0.12, -0.01) mmol/L) and high-density lipoprotein cholesterol (β -0.05 (95 % CI -0.07, -0.03) mmol/L). CONCLUSION The newly introduced iAge, derived from inflammatory markers and telomere length, aligns with various metabolic dysfunctions and age-related disease risks, underscoring its potential ability in identifying aging-related phenotypes.
Collapse
Affiliation(s)
- Rui Zhen Wang
- School of Public Health, Sun Yat-Sen University, Guangzhou, China
| | - Wei Sen Zhang
- Guangzhou Twelfth People's Hospital, Guangzhou, China.
| | | | - Feng Zhu
- Guangzhou Twelfth People's Hospital, Guangzhou, China
| | - Ya Li Jin
- Guangzhou Twelfth People's Hospital, Guangzhou, China
| | - Lin Xu
- School of Public Health, Sun Yat-Sen University, Guangzhou, China; School of Public Health, the University of Hong Kong, Hong Kong, China; Institute of Applied Health Research, University of Birmingham, Birmingham, UK.
| |
Collapse
|
41
|
Zhang S, Wang Z, Wang Y, Zhu Y, Zhou Q, Jian X, Zhao G, Qiu J, Xia K, Tang B, Mutz J, Li J, Li B. A metabolomic profile of biological aging in 250,341 individuals from the UK Biobank. Nat Commun 2024; 15:8081. [PMID: 39278973 PMCID: PMC11402978 DOI: 10.1038/s41467-024-52310-9] [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: 01/22/2024] [Accepted: 09/02/2024] [Indexed: 09/18/2024] Open
Abstract
The metabolomic profile of aging is complex. Here, we analyse 325 nuclear magnetic resonance (NMR) biomarkers from 250,341 UK Biobank participants, identifying 54 representative aging-related biomarkers associated with all-cause mortality. We conduct genome-wide association studies (GWAS) for these 325 biomarkers using whole-genome sequencing (WGS) data from 95,372 individuals and perform multivariable Mendelian randomization (MVMR) analyses, discovering 439 candidate "biomarker - disease" causal pairs at the nominal significance level. We develop a metabolomic aging score that outperforms other aging metrics in predicting short-term mortality risk and exhibits strong potential for discriminating aging-accelerated populations and improving disease risk prediction. A longitudinal analysis of 13,263 individuals enables us to calculate a metabolomic aging rate which provides more refined aging assessments and to identify candidate anti-aging and pro-aging NMR biomarkers. Taken together, our study has presented a comprehensive aging-related metabolomic profile and highlighted its potential for personalized aging monitoring and early disease intervention.
Collapse
Affiliation(s)
- Shiyu Zhang
- National Clinical Research Center for Geriatric Disorders, Department of Geriatrics, Xiangya Hospital & Center for Medical Genetics, School of Life Sciences, Central South University, Changsha, Hunan, 410008, China
- Xiangya School of Medicine, Central South University, Changsha, Hunan, 410013, China
| | - Zheng Wang
- National Clinical Research Center for Geriatric Disorders, Department of Geriatrics, Xiangya Hospital & Center for Medical Genetics, School of Life Sciences, Central South University, Changsha, Hunan, 410008, China
- Key Laboratory of Hunan Province in Neurodegenerative Disorders, Department of Neurology, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
- Hunan Key Laboratory of Molecular Precision Medicine, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
| | - Yijing Wang
- National Clinical Research Center for Geriatric Disorders, Department of Geriatrics, Xiangya Hospital & Center for Medical Genetics, School of Life Sciences, Central South University, Changsha, Hunan, 410008, China
| | - Yixiao Zhu
- National Clinical Research Center for Geriatric Disorders, Department of Geriatrics, Xiangya Hospital & Center for Medical Genetics, School of Life Sciences, Central South University, Changsha, Hunan, 410008, China
| | - Qiao Zhou
- National Clinical Research Center for Geriatric Disorders, Department of Geriatrics, Xiangya Hospital & Center for Medical Genetics, School of Life Sciences, Central South University, Changsha, Hunan, 410008, China
| | - Xingxing Jian
- National Clinical Research Center for Geriatric Disorders, Department of Geriatrics, Xiangya Hospital & Center for Medical Genetics, School of Life Sciences, Central South University, Changsha, Hunan, 410008, China
| | - Guihu Zhao
- National Clinical Research Center for Geriatric Disorders, Department of Geriatrics, Xiangya Hospital & Center for Medical Genetics, School of Life Sciences, Central South University, Changsha, Hunan, 410008, China
| | - Jian Qiu
- National Clinical Research Center for Geriatric Disorders, Department of Geriatrics, Xiangya Hospital & Center for Medical Genetics, School of Life Sciences, Central South University, Changsha, Hunan, 410008, China
- Key Laboratory of Hunan Province in Neurodegenerative Disorders, Department of Neurology, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
- Hunan Key Laboratory of Molecular Precision Medicine, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
| | - Kun Xia
- MOE Key Laboratory of Pediatric Rare Diseases & Hunan Key Laboratory of Medical Genetics, Central South University, Changsha, Hunan, 410008, China
| | - Beisha Tang
- National Clinical Research Center for Geriatric Disorders, Department of Geriatrics, Xiangya Hospital & Center for Medical Genetics, School of Life Sciences, Central South University, Changsha, Hunan, 410008, China
- Key Laboratory of Hunan Province in Neurodegenerative Disorders, Department of Neurology, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
- Department of Neurology & Multi-omics Research Center for Brain Disorders, The First Affiliated Hospital University of South China, Hengyang, Hunan, China
| | - Julian Mutz
- Social, Genetic and Developmental Psychiatry Centre, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, UK.
| | - Jinchen Li
- National Clinical Research Center for Geriatric Disorders, Department of Geriatrics, Xiangya Hospital & Center for Medical Genetics, School of Life Sciences, Central South University, Changsha, Hunan, 410008, China.
- Key Laboratory of Hunan Province in Neurodegenerative Disorders, Department of Neurology, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China.
- Bioinformatics Center, Xiangya Hospital & Furong Laboratory, Changsha, Hunan, 410008, China.
| | - Bin Li
- National Clinical Research Center for Geriatric Disorders, Department of Geriatrics, Xiangya Hospital & Center for Medical Genetics, School of Life Sciences, Central South University, Changsha, Hunan, 410008, China.
| |
Collapse
|
42
|
Bizzarri D, Reinders MJT, Kuiper L, Beekman M, Deelen J, van Meurs JBJ, van Dongen J, Pool R, Boomsma DI, Ghanbari M, Franke L, Slagboom PE, van den Akker EB. NMR metabolomics-guided DNA methylation mortality predictors. EBioMedicine 2024; 107:105279. [PMID: 39154540 PMCID: PMC11378104 DOI: 10.1016/j.ebiom.2024.105279] [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: 03/14/2024] [Revised: 07/25/2024] [Accepted: 07/29/2024] [Indexed: 08/20/2024] Open
Abstract
BACKGROUND 1H-NMR metabolomics and DNA methylation in blood are widely known biomarkers predicting age-related physiological decline and mortality yet exert mutually independent mortality and frailty signals. METHODS Leveraging multi-omics data in four Dutch population studies (N = 5238, ∼40% of which male) we investigated whether the mortality signal captured by 1H-NMR metabolomics could guide the construction of DNA methylation-based mortality predictors. FINDINGS We trained DNA methylation-based surrogates for 64 metabolomic analytes and found that analytes marking inflammation, fluid balance, or HDL/VLDL metabolism could be accurately reconstructed using DNA-methylation assays. Interestingly, a previously reported multi-analyte score indicating mortality risk (MetaboHealth) could also be accurately reconstructed. Sixteen of our derived surrogates, including the MetaboHealth surrogate, showed significant associations with mortality, independent of relevant covariates. INTERPRETATION The addition of our metabolic analyte-derived surrogates to the well-established epigenetic clock GrimAge demonstrates that our surrogates potentially represent valuable mortality signal. FUNDING BBMRI-NL, X-omics, VOILA, Medical Delta, NWO, ERC.
Collapse
Affiliation(s)
- Daniele Bizzarri
- Molecular Epidemiology, Department of Biomedical Data Sciences, Leiden University Medical Center, Leiden, the Netherlands; Leiden Computational Biology Center, Department of Biomedical Data Sciences, Leiden University Medical Center, Leiden, the Netherlands; Delft Bioinformatics Lab, TU Delft, Delft, the Netherlands
| | - Marcel J T Reinders
- Leiden Computational Biology Center, Department of Biomedical Data Sciences, Leiden University Medical Center, Leiden, the Netherlands; Delft Bioinformatics Lab, TU Delft, Delft, the Netherlands
| | - Lieke Kuiper
- Department of Internal Medicine, Erasmus MC, Rotterdam, the Netherlands; Center for Nutrition, Prevention and Health Services, National Institute for Public Health and Environment (RIVM), Bilthoven, the Netherlands
| | - Marian Beekman
- Molecular Epidemiology, Department of Biomedical Data Sciences, Leiden University Medical Center, Leiden, the Netherlands
| | - Joris Deelen
- Molecular Epidemiology, Department of Biomedical Data Sciences, Leiden University Medical Center, Leiden, the Netherlands; Max Planck Institute for the Biology of Ageing, Cologne, Germany; Cologne Excellence Cluster on Cellular Stress Responses in Aging Associated Diseases, University of Cologne, Cologne, Germany
| | - Joyce B J van Meurs
- Department of Internal Medicine, Erasmus MC, Rotterdam, the Netherlands; Department of Orthopaedics & Sports, Erasmus Medical Center, Rotterdam, the Netherlands
| | - Jenny van Dongen
- Department of Biological Psychology, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands; Amsterdam Reproduction and Development (AR&D) Research Institute, Amsterdam, the Netherlands; Amsterdam Public Health Research Institute, Amsterdam, the Netherlands
| | - René Pool
- Department of Biological Psychology, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands; Amsterdam Public Health Research Institute, Amsterdam, the Netherlands
| | - Dorret I Boomsma
- Department of Biological Psychology, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands; Amsterdam Reproduction and Development (AR&D) Research Institute, Amsterdam, the Netherlands; Amsterdam Public Health Research Institute, Amsterdam, the Netherlands
| | - Mohsen Ghanbari
- Department of Epidemiology, Erasmus MC, Rotterdam, the Netherlands
| | - Lude Franke
- Department of Genetics, University Medical Center Groningen, Groningen, the Netherlands
| | - Pieternella E Slagboom
- Molecular Epidemiology, Department of Biomedical Data Sciences, Leiden University Medical Center, Leiden, the Netherlands; Max Planck Institute for the Biology of Ageing, Cologne, Germany
| | - Erik B van den Akker
- Molecular Epidemiology, Department of Biomedical Data Sciences, Leiden University Medical Center, Leiden, the Netherlands; Leiden Computational Biology Center, Department of Biomedical Data Sciences, Leiden University Medical Center, Leiden, the Netherlands; Delft Bioinformatics Lab, TU Delft, Delft, the Netherlands.
| |
Collapse
|
43
|
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
|
44
|
Abdelhafez YG, Wang G, Li S, Pellegrinelli V, Chaudhari AJ, Ramirez A, Sen F, Vidal-Puig A, Sidossis LS, Klein S, Badawi RD, Chondronikola M. The role of brown adipose tissue in branched-chain amino acid clearance in people. iScience 2024; 27:110559. [PMID: 39175781 PMCID: PMC11340589 DOI: 10.1016/j.isci.2024.110559] [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: 09/29/2023] [Revised: 06/24/2024] [Accepted: 07/17/2024] [Indexed: 08/24/2024] Open
Abstract
Brown adipose tissue (BAT) in rodents appears to be an important tissue for the clearance of plasma branched-chain amino acids (BCAAs) contributing to improved metabolic health. However, the role of human BAT in plasma BCAA clearance is poorly understood. Here, we evaluate patients with prostate cancer who underwent positron emission tomography-computed tomography imaging after an injection of 18F-fluciclovine (L-leucine analog). Supraclavicular adipose tissue (AT; primary location of human BAT) has a higher net uptake rate for 18F-fluciclovine compared to subcutaneous abdominal and upper chest AT. Supraclavicular AT 18F-fluciclovine net uptake rate is lower in patients with obesity and type 2 diabetes. Finally, the expression of genes involved in BCAA catabolism is higher in the supraclavicular AT of healthy people with high BAT volume compared to those with low BAT volume. These findings support the notion that BAT can potentially function as a metabolic sink for plasma BCAA clearance in people.
Collapse
Affiliation(s)
- Yasser G. Abdelhafez
- Department of Radiology, University of California Davis, Sacramento, CA 95817, USA
- Nuclear Medicine Unit, South Egypt Cancer Institute, Assiut University, El Fateh 71111, Egypt
| | - Guobao Wang
- Department of Radiology, University of California Davis, Sacramento, CA 95817, USA
| | - Siqi Li
- Department of Radiology, University of California Davis, Sacramento, CA 95817, USA
| | - Vanessa Pellegrinelli
- Institute of Metabolic Science-Metabolic Research Laboratories, Medical Research Council Metabolic Diseases Unit, University of Cambridge, Cambridge CB2 0QQ, UK
| | - Abhijit J. Chaudhari
- Department of Radiology, University of California Davis, Sacramento, CA 95817, USA
| | - Anthony Ramirez
- Department of Nutrition, University of California Davis, Davis, CA 95616, USA
| | - Fatma Sen
- Department of Radiology, University of California Davis, Sacramento, CA 95817, USA
| | - Antonio Vidal-Puig
- Institute of Metabolic Science-Metabolic Research Laboratories, Medical Research Council Metabolic Diseases Unit, University of Cambridge, Cambridge CB2 0QQ, UK
| | - Labros S. Sidossis
- Department of Kinesiology and Health, Rutgers University, New Brunswick, NJ 08901, USA
| | - Samuel Klein
- Center for Human Nutrition, Washington University School of Medicine, Saint Louis, MO 63110, USA
| | - Ramsey D. Badawi
- Department of Radiology, University of California Davis, Sacramento, CA 95817, USA
| | - Maria Chondronikola
- Department of Radiology, University of California Davis, Sacramento, CA 95817, USA
- Institute of Metabolic Science-Metabolic Research Laboratories, Medical Research Council Metabolic Diseases Unit, University of Cambridge, Cambridge CB2 0QQ, UK
- Department of Nutrition, University of California Davis, Davis, CA 95616, USA
- Department of Nutrition and Dietetics, Harokopio University of Athens, 17778 Athens, Greece
| |
Collapse
|
45
|
Adolph TE, Tilg H. Western diets and chronic diseases. Nat Med 2024; 30:2133-2147. [PMID: 39085420 DOI: 10.1038/s41591-024-03165-6] [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: 03/01/2024] [Accepted: 06/28/2024] [Indexed: 08/02/2024]
Abstract
'Westernization', which incorporates industrial, cultural and dietary trends, has paralleled the rise of noncommunicable diseases across the globe. Today, the Western-style diet emerges as a key stimulus for gut microbial vulnerability, chronic inflammation and chronic diseases, affecting mainly the cardiovascular system, systemic metabolism and the gut. Here we review the diet of modern times and evaluate the threat it poses for human health by summarizing recent epidemiological, translational and clinical studies. We discuss the links between diet and disease in the context of obesity and type 2 diabetes, cardiovascular diseases, gut and liver diseases and solid malignancies. We collectively interpret the evidence and its limitations and discuss future challenges and strategies to overcome these. We argue that healthcare professionals and societies must react today to the detrimental effects of the Western diet to bring about sustainable change and improved outcomes in the future.
Collapse
Affiliation(s)
- Timon E Adolph
- Department of Internal Medicine I, Gastroenterology, Hepatology, Endocrinology and Metabolism, Medical University of Innsbruck, Innsbruck, Austria.
| | - Herbert Tilg
- Department of Internal Medicine I, Gastroenterology, Hepatology, Endocrinology and Metabolism, Medical University of Innsbruck, Innsbruck, Austria.
| |
Collapse
|
46
|
Kuiper LM, Smit AP, Bizzarri D, van den Akker EB, Reinders MJT, Ghanbari M, van Rooij JGJ, Voortman T, Rivadeneira F, Dollé MET, Herber GCM, Rietman ML, Picavet HSJ, van Meurs JBJ, Verschuren WMM. Lifestyle factors and metabolomic aging biomarkers: Meta-analysis of cross-sectional and longitudinal associations in three prospective cohorts. Mech Ageing Dev 2024; 220:111958. [PMID: 38950629 DOI: 10.1016/j.mad.2024.111958] [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: 02/07/2024] [Revised: 06/18/2024] [Accepted: 06/24/2024] [Indexed: 07/03/2024]
Abstract
Biological age uses biophysiological information to capture a person's age-related risk of adverse outcomes. MetaboAge and MetaboHealth are metabolomics-based biomarkers of biological age trained on chronological age and mortality risk, respectively. Lifestyle factors contribute to the extent chronological and biological age differ. The association of lifestyle factors with MetaboAge and MetaboHealth, potential sex differences in these associations, and MetaboAge's and MetaboHealth's sensitivity to lifestyle changes have not been studied yet. Linear regression analyses and mixed-effect models were used to examine the cross-sectional and longitudinal associations of scaled lifestyle factors with scaled MetaboAge and MetaboHealth in 24,332 middle-aged participants from the Doetinchem Cohort Study, Rotterdam Study, and UK Biobank. Random-effect meta-analyses were performed across cohorts. Repeated metabolomics measurements had a ten-year interval in the Doetinchem Cohort Study and a five-year interval in the UK Biobank. In the first study incorporating longitudinal information on MetaboAge and MetaboHealth, we demonstrate associations between current smoking, sleeping ≥8 hours/day, higher BMI, and larger waist circumference were associated with higher MetaboHealth, the latter two also with higher MetaboAge. Furthermore, adhering to the dietary and physical activity guidelines were inversely associated with MetaboHealth. Lastly, we observed sex differences in the associations between alcohol use and MetaboHealth.
Collapse
Affiliation(s)
- L M Kuiper
- Center for Prevention, Lifestyle and Health, National Institute for Public Health and Environment (RIVM), Bilthoven, the Netherlands; Department of Internal Medicine, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - A P Smit
- Center for Prevention, Lifestyle and Health, National Institute for Public Health and Environment (RIVM), Bilthoven, the Netherlands; Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| | - D Bizzarri
- Molecular Epidemiology, Department of Biomedical Data Sciences, Leiden University Medical Center, Leiden, the Netherlands; Leiden Computational Biology Center, Department of Biomedical Data Sciences, Leiden University Medical Center, Leiden, the Netherlands; Delft Bioinformatics Lab, TU Delft, Delft, the Netherlands
| | - E B van den Akker
- Molecular Epidemiology, Department of Biomedical Data Sciences, Leiden University Medical Center, Leiden, the Netherlands; Leiden Computational Biology Center, Department of Biomedical Data Sciences, Leiden University Medical Center, Leiden, the Netherlands; Delft Bioinformatics Lab, TU Delft, Delft, the Netherlands
| | - M J T Reinders
- Molecular Epidemiology, Department of Biomedical Data Sciences, Leiden University Medical Center, Leiden, the Netherlands; Leiden Computational Biology Center, Department of Biomedical Data Sciences, Leiden University Medical Center, Leiden, the Netherlands; Delft Bioinformatics Lab, TU Delft, Delft, the Netherlands
| | - M Ghanbari
- Department of Epidemiology, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - J G J van Rooij
- Department of Internal Medicine, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - T Voortman
- Department of Epidemiology, Erasmus University Medical Center, Rotterdam, the Netherlands; Meta-Research Innovation Center at Stanford (METRICS), Stanford University, California, USA
| | - F Rivadeneira
- Department of Internal Medicine, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - M E T Dollé
- Center for Health Protection, National Institute for Public Health and Environment (RIVM), Bilthoven, the Netherlands
| | - G C M Herber
- Center for Prevention, Lifestyle and Health, National Institute for Public Health and Environment (RIVM), Bilthoven, the Netherlands
| | - M L Rietman
- Center for Prevention, Lifestyle and Health, National Institute for Public Health and Environment (RIVM), Bilthoven, the Netherlands
| | - H S J Picavet
- Center for Prevention, Lifestyle and Health, National Institute for Public Health and Environment (RIVM), Bilthoven, the Netherlands
| | - J B J van Meurs
- Department of Internal Medicine, Erasmus University Medical Center, Rotterdam, the Netherlands; Department of Orthopaedics & Sports, Erasmus Medical Center, Rotterdam, the Netherlands
| | - W M M Verschuren
- Center for Prevention, Lifestyle and Health, National Institute for Public Health and Environment (RIVM), Bilthoven, the Netherlands; Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands.
| |
Collapse
|
47
|
Liang N, Nho K, Newman JW, Arnold M, Huynh K, Meikle PJ, Borkowski K, Kaddurah-Daouk R. Peripheral inflammation is associated with brain atrophy and cognitive decline linked to mild cognitive impairment and Alzheimer's disease. Sci Rep 2024; 14:17423. [PMID: 39075118 PMCID: PMC11286782 DOI: 10.1038/s41598-024-67177-5] [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: 02/01/2024] [Accepted: 07/09/2024] [Indexed: 07/31/2024] Open
Abstract
Inflammation is an important factor in Alzheimer's disease (AD). An NMR measurement in plasma, glycoprotein acetyls (GlycA), captures the overall level of protein production and glycosylation implicated in systemic inflammation. With its additional advantage of reducing biological variability, GlycA might be useful in monitoring the relationship between peripheral inflammation and brain changes relevant to AD. However, the associations between GlycA and these brain changes have not been fully evaluated. Here, we performed Spearman's correlation analyses to evaluate these associations cross-sectionally and determined whether GlycA can inform AD-relevant longitudinal measurements among participants in the Alzheimer's Disease Neuroimaging Initiative (n = 1506), with additional linear models and stratification analyses to evaluate the influences of sex or diagnosis status and confirm findings from Spearman's correlation analyses. We found that GlycA was elevated in AD patients compared to cognitively normal participants. GlycA correlated negatively with multiple concurrent regional brain volumes in females diagnosed with late mild cognitive impairment (LMCI) or AD. Baseline GlycA level was associated with executive function decline at 3-9 year follow-up in participants diagnosed with LMCI at baseline, with similar but not identical trends observed in the future decline of memory and entorhinal cortex volume. Results here indicated that GlycA is an inflammatory biomarker relevant to AD pathogenesis and that the stage of LMCI might be relevant to inflammation-related intervention.
Collapse
Affiliation(s)
- Nuanyi Liang
- West Coast Metabolomics Center, Genome Center, University of California-Davis, Davis, CA, 95616, USA
| | - Kwangsik Nho
- Department of Radiology and Imaging Sciences and the Indiana Alzheimer Disease Center, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - John W Newman
- West Coast Metabolomics Center, Genome Center, University of California-Davis, Davis, CA, 95616, USA
- Department of Nutrition, University of California-Davis, Davis, CA, 95616, USA
- Western Human Nutrition Research Center, United States Department of Agriculture-Agriculture Research Service, Davis, CA, 95616, USA
| | - Matthias Arnold
- Department of Psychiatry and Behavioral Sciences, Duke University, Durham, NC, 27708, USA
- Institute of Computational Biology, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
| | - Kevin Huynh
- Baker Heart and Diabetes Institute, Melbourne, VIC, 3004, Australia
- Baker Department of Cardiovascular Research, Translation and Implementation, La Trobe University, Bundoora, VIC, 3086, Australia
| | - Peter J Meikle
- Baker Heart and Diabetes Institute, Melbourne, VIC, 3004, Australia
- Baker Department of Cardiovascular Research, Translation and Implementation, La Trobe University, Bundoora, VIC, 3086, Australia
| | - Kamil Borkowski
- West Coast Metabolomics Center, Genome Center, University of California-Davis, Davis, CA, 95616, USA.
| | - Rima Kaddurah-Daouk
- Department of Psychiatry and Behavioral Sciences, Duke University, Durham, NC, 27708, USA.
- Duke Institute of Brain Sciences, Duke University, Durham, NC, USA.
- Department of Medicine, Duke University, Durham, NC, USA.
| |
Collapse
|
48
|
Zheng Z, Li J, Liu T, Fan Y, Zhai QC, Xiong M, Wang QR, Sun X, Zheng QW, Che S, Jiang B, Zheng Q, Wang C, Liu L, Ping J, Wang S, Gao DD, Ye J, Yang K, Zuo Y, Ma S, Yang YG, Qu J, Zhang F, Jia P, Liu GH, Zhang W. DNA methylation clocks for estimating biological age in Chinese cohorts. Protein Cell 2024; 15:575-593. [PMID: 38482631 PMCID: PMC11259550 DOI: 10.1093/procel/pwae011] [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: 11/11/2023] [Accepted: 01/10/2024] [Indexed: 07/21/2024] Open
Abstract
Epigenetic clocks are accurate predictors of human chronological age based on the analysis of DNA methylation (DNAm) at specific CpG sites. However, a systematic comparison between DNA methylation data and other omics datasets has not yet been performed. Moreover, available DNAm age predictors are based on datasets with limited ethnic representation. To address these knowledge gaps, we generated and analyzed DNA methylation datasets from two independent Chinese cohorts, revealing age-related DNAm changes. Additionally, a DNA methylation aging clock (iCAS-DNAmAge) and a group of DNAm-based multi-modal clocks for Chinese individuals were developed, with most of them demonstrating strong predictive capabilities for chronological age. The clocks were further employed to predict factors influencing aging rates. The DNAm aging clock, derived from multi-modal aging features (compositeAge-DNAmAge), exhibited a close association with multi-omics changes, lifestyles, and disease status, underscoring its robust potential for precise biological age assessment. Our findings offer novel insights into the regulatory mechanism of age-related DNAm changes and extend the application of the DNAm clock for measuring biological age and aging pace, providing the basis for evaluating aging intervention strategies.
Collapse
Affiliation(s)
- Zikai Zheng
- CAS Key Laboratory of Genomic and Precision Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences and China National Center for Bioinformation, Beijing 100101, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Jiaming Li
- CAS Key Laboratory of Genomic and Precision Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences and China National Center for Bioinformation, Beijing 100101, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Tianzi Liu
- CAS Key Laboratory of Genomic and Precision Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences and China National Center for Bioinformation, Beijing 100101, China
- CAS Key Laboratory of Computational Biology, Shanghai Institute of Nutrition and Health, Chinese Academy of Sciences, Shanghai 200031, China
| | - Yanling Fan
- CAS Key Laboratory of Genomic and Precision Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences and China National Center for Bioinformation, Beijing 100101, China
| | - Qiao-Cheng Zhai
- Division of Orthopaedics, Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou 324000, China
- The Joint Innovation Center for Engineering in Medicine, Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou 324000, China
| | - Muzhao Xiong
- CAS Key Laboratory of Genomic and Precision Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences and China National Center for Bioinformation, Beijing 100101, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Qiao-Ran Wang
- CAS Key Laboratory of Genomic and Precision Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences and China National Center for Bioinformation, Beijing 100101, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Xiaoyan Sun
- CAS Key Laboratory of Genomic and Precision Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences and China National Center for Bioinformation, Beijing 100101, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Qi-Wen Zheng
- CAS Key Laboratory of Genomic and Precision Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences and China National Center for Bioinformation, Beijing 100101, China
| | - Shanshan Che
- CAS Key Laboratory of Genomic and Precision Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences and China National Center for Bioinformation, Beijing 100101, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Beier Jiang
- The Joint Innovation Center for Engineering in Medicine, Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou 324000, China
| | - Quan Zheng
- The Joint Innovation Center for Engineering in Medicine, Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou 324000, China
| | - Cui Wang
- CAS Key Laboratory of Genomic and Precision Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences and China National Center for Bioinformation, Beijing 100101, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Lixiao Liu
- CAS Key Laboratory of Genomic and Precision Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences and China National Center for Bioinformation, Beijing 100101, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Jiale Ping
- CAS Key Laboratory of Genomic and Precision Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences and China National Center for Bioinformation, Beijing 100101, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Si Wang
- Advanced Innovation Center for Human Brain Protection, and National Clinical Research Center for Geriatric Disorders, Xuanwu Hospital Capital Medical University, Beijing 100053, China
- Aging Translational Medicine Center, International Center for Aging and Cancer, Xuanwu Hospital, Capital Medical University, Beijing 100053, China
- Aging Biomarker Consortium, Beijing 100101, China
| | - Dan-Dan Gao
- The Joint Innovation Center for Engineering in Medicine, Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou 324000, China
| | - Jinlin Ye
- The Joint Innovation Center for Engineering in Medicine, Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou 324000, China
| | - Kuan Yang
- CAS Key Laboratory of Genomic and Precision Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences and China National Center for Bioinformation, Beijing 100101, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yuesheng Zuo
- CAS Key Laboratory of Genomic and Precision Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences and China National Center for Bioinformation, Beijing 100101, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Shuai Ma
- Aging Biomarker Consortium, Beijing 100101, China
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
- Key Laboratory of Organ Regeneration and Reconstruction, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing 100101, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing 100101, China
| | - Yun-Gui Yang
- CAS Key Laboratory of Genomic and Precision Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences and China National Center for Bioinformation, Beijing 100101, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Jing Qu
- University of Chinese Academy of Sciences, Beijing 100049, China
- Aging Biomarker Consortium, Beijing 100101, China
- Key Laboratory of Organ Regeneration and Reconstruction, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing 100101, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing 100101, China
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
| | - Feng Zhang
- Division of Orthopaedics, Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou 324000, China
| | - Peilin Jia
- CAS Key Laboratory of Genomic and Precision Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences and China National Center for Bioinformation, Beijing 100101, China
- University of Chinese Academy of Sciences, Beijing 100049, China
- National Genomics Data Center, Beijing Institute of Genomics, Chinese Academy of Sciences and China National Center for Bioinformation, Beijing 100101, China
| | - Guang-Hui Liu
- University of Chinese Academy of Sciences, Beijing 100049, China
- Advanced Innovation Center for Human Brain Protection, and National Clinical Research Center for Geriatric Disorders, Xuanwu Hospital Capital Medical University, Beijing 100053, China
- Aging Translational Medicine Center, International Center for Aging and Cancer, Xuanwu Hospital, Capital Medical University, Beijing 100053, China
- Aging Biomarker Consortium, Beijing 100101, China
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
- Key Laboratory of Organ Regeneration and Reconstruction, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing 100101, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing 100101, China
| | - Weiqi Zhang
- CAS Key Laboratory of Genomic and Precision Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences and China National Center for Bioinformation, Beijing 100101, China
- University of Chinese Academy of Sciences, Beijing 100049, China
- Aging Biomarker Consortium, Beijing 100101, China
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing 100101, China
| |
Collapse
|
49
|
Arsenault BJ, Carpentier AC, Poirier P, Després JP. Adiposity, type 2 diabetes and atherosclerotic cardiovascular disease risk: Use and abuse of the body mass index. Atherosclerosis 2024; 394:117546. [PMID: 38692978 DOI: 10.1016/j.atherosclerosis.2024.117546] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Revised: 03/29/2024] [Accepted: 04/10/2024] [Indexed: 05/03/2024]
Abstract
The worldwide prevalence of individuals with an elevated body weight has increased steadily over the past five decades. Billions of research dollars have been invested to improve our understanding of the causes and consequences of having an elevated body weight. All this knowledge has, however, failed to influence populational body weight trajectories of most countries around the world. Research on the definition of "obesity" has also evolved. Body mass index (BMI), the most commonly used tool to make its diagnosis, has major limitations. In this review article, we will highlight evidence from observational studies, genetic association studies and randomized clinical trials that have shown the remarkable inter-individual differences in the way humans store energy as body fat. Increasing evidence also suggests that, as opposed to weight inclusive, lifestyle-based approaches, weight-centric approaches advising people to simply eat less and move more are not sustainable for most people for long-term weight loss and maintenance. It is time to recognize that this outdated approach may have produced more harm than good. On the basis of pathophysiological, genetic and clinical evidence presented in this review, we propose that it may be time to shift away from the traditional clinical approach, which is BMI-centric. Rather, emphasis should be placed on actionable lifestyle-related risk factors aiming at improving overall diet quality and increasing physical activity level in the general population.
Collapse
Affiliation(s)
- Benoit J Arsenault
- Centre de recherche de l'Institut universitaire de cardiologie et de pneumologie de Québec - Université Laval, Québec (QC), Canada; Department of Medicine, Faculty of Medicine, Université Laval, Québec (QC), Canada
| | - André C Carpentier
- Division of Endocrinology, Department of Medicine, Centre de recherche du Centre hospitalier universitaire de Sherbrooke, Université de Sherbrooke, Sherbrooke (QC), Canada
| | - Paul Poirier
- Centre de recherche de l'Institut universitaire de cardiologie et de pneumologie de Québec - Université Laval, Québec (QC), Canada; Faculté de pharmacie, Université Laval, Québec (QC), Canada
| | - Jean-Pierre Després
- Centre de recherche de l'Institut universitaire de cardiologie et de pneumologie de Québec - Université Laval, Québec (QC), Canada; VITAM - Centre de recherche en santé durable, CIUSSS de la Capitale-Nationale, Québec (QC), Canada; Department of Kinesiology, Faculty of Medicine, Université Laval, Québec (QC), Canada.
| |
Collapse
|
50
|
Gadd DA, Hillary RF, Kuncheva Z, Mangelis T, Cheng Y, Dissanayake M, Admanit R, Gagnon J, Lin T, Ferber KL, Runz H, Foley CN, Marioni RE, Sun BB. Blood protein assessment of leading incident diseases and mortality in the UK Biobank. NATURE AGING 2024; 4:939-948. [PMID: 38987645 PMCID: PMC11257969 DOI: 10.1038/s43587-024-00655-7] [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: 03/15/2023] [Accepted: 05/22/2024] [Indexed: 07/12/2024]
Abstract
The circulating proteome offers insights into the biological pathways that underlie disease. Here, we test relationships between 1,468 Olink protein levels and the incidence of 23 age-related diseases and mortality in the UK Biobank (n = 47,600). We report 3,209 associations between 963 protein levels and 21 incident outcomes. Next, protein-based scores (ProteinScores) are developed using penalized Cox regression. When applied to test sets, six ProteinScores improve the area under the curve estimates for the 10-year onset of incident outcomes beyond age, sex and a comprehensive set of 24 lifestyle factors, clinically relevant biomarkers and physical measures. Furthermore, the ProteinScore for type 2 diabetes outperforms a polygenic risk score and HbA1c-a clinical marker used to monitor and diagnose type 2 diabetes. The performance of scores using metabolomic and proteomic features is also compared. These data characterize early proteomic contributions to major age-related diseases, demonstrating the value of the plasma proteome for risk stratification.
Collapse
Affiliation(s)
- Danni A Gadd
- Optima Partners, Edinburgh, UK
- Centre for Genomic and Experimental Medicine, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, UK
| | - Robert F Hillary
- Optima Partners, Edinburgh, UK
- Centre for Genomic and Experimental Medicine, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, UK
| | - Zhana Kuncheva
- Optima Partners, Edinburgh, UK
- Bayes Centre, University of Edinburgh, Edinburgh, UK
| | - Tasos Mangelis
- Optima Partners, Edinburgh, UK
- Bayes Centre, University of Edinburgh, Edinburgh, UK
| | - Yipeng Cheng
- Centre for Genomic and Experimental Medicine, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, UK
| | - Manju Dissanayake
- Optima Partners, Edinburgh, UK
- Bayes Centre, University of Edinburgh, Edinburgh, UK
| | - Romi Admanit
- Biostatistics, Research and Development, Biogen Inc., Cambridge, MA, USA
| | - Jake Gagnon
- Biostatistics, Research and Development, Biogen Inc., Cambridge, MA, USA
| | - Tinchi Lin
- Biostatistics, Research and Development, Biogen Inc., Cambridge, MA, USA
| | - Kyle L Ferber
- Biostatistics, Research and Development, Biogen Inc., Cambridge, MA, USA
| | - Heiko Runz
- Translational Sciences, Research and Development, Biogen Inc., Cambridge, MA, USA
| | - Christopher N Foley
- Optima Partners, Edinburgh, UK.
- Bayes Centre, University of Edinburgh, Edinburgh, UK.
| | - Riccardo E Marioni
- Optima Partners, Edinburgh, UK.
- Centre for Genomic and Experimental Medicine, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, UK.
| | - Benjamin B Sun
- Translational Sciences, Research and Development, Biogen Inc., Cambridge, MA, USA.
- Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK.
| |
Collapse
|