1
|
Zhang Y, Guo R, Yin M, Shi M, Zhong T, Liu S, Li X. The immunological age prediction of monocytes indicates that gestational diabetes mellitus accelerates the aging of monocytes in offspring. Immun Ageing 2025; 22:18. [PMID: 40361109 PMCID: PMC12070592 DOI: 10.1186/s12979-025-00513-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Accepted: 04/26/2025] [Indexed: 05/15/2025]
Abstract
BACKGROUND Recent studies have suggested that gestational diabetes mellitus (GDM) can accelerate cellular aging in multiple cell types in offspring, but its impact on immune senescence remains uncertain. Our prior study reveals GDM increased the secretion of inflammatory factors by monocytes in offspring. This study discovered the transcriptome characteristics of aging monocytes at the single-cell level and explore the impact of GDM on the progression of monocyte aging in offspring. METHOD Single-cell sequencing data from 56 healthy individuals (aged 0-100 years), comprising self-measured samples (n = 6) and publicly available datasets from the Gene Expression Omnibus (GEO, n = 50), were analyzed to characterize monocyte senescence. Linear mixed-effects modeling was used to screen for age-related genes. A random forest model was created to predict immune age in monocytes, allowing for quantitative assessment of aging. RESULTS We detected an increase in the number of inflammatory monocytes expressing IL1B and CXCL8 with age. Two age-related gene expression patterns were identified in monocytes. Analysis of offspring monocytes from mothers with GDM suggested that exposure to a GDM environment in the womb may lead to increased expression of aging-related genes, a hindered cell cycle, and increased immune age. The immune age of monocytes at birth is significantly linked to maternal weight gain, high fasting blood glucose levels, and cord blood C-peptide levels during pregnancy. CONCLUSIONS Exposure to GDM during pregnancy accelerates aging in offspring immune cells. Monitoring maternal weight and blood sugar during GDM can help prevent negative effects on the offspring immune system.
Collapse
Affiliation(s)
- Yan Zhang
- National Clinical Research Center for Metabolic Diseases, Key Laboratory of Diabetes Immunology, Ministry of Education, and Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Rui Guo
- National Clinical Research Center for Metabolic Diseases, Key Laboratory of Diabetes Immunology, Ministry of Education, and Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Min Yin
- National Clinical Research Center for Metabolic Diseases, Key Laboratory of Diabetes Immunology, Ministry of Education, and Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha, China
- Department of Nutritional, The Second Xiangya Hospital of Central South University, 139 Renmin Zhong Road, Changsha, 410011, Hunan, China
| | - Mei Shi
- National Clinical Research Center for Metabolic Diseases, Key Laboratory of Diabetes Immunology, Ministry of Education, and Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Ting Zhong
- National Clinical Research Center for Metabolic Diseases, Key Laboratory of Diabetes Immunology, Ministry of Education, and Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Shanshan Liu
- National Clinical Research Center for Metabolic Diseases, Key Laboratory of Diabetes Immunology, Ministry of Education, and Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Xia Li
- National Clinical Research Center for Metabolic Diseases, Key Laboratory of Diabetes Immunology, Ministry of Education, and Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha, China.
| |
Collapse
|
2
|
Newediuk L, Richardson ES, Bohart AM, Roberto-Charron A, Garroway CJ, Jones MJ. Designing Epigenetic Clocks for Wildlife Research. Mol Ecol Resour 2025:e14120. [PMID: 40326643 DOI: 10.1111/1755-0998.14120] [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: 02/12/2025] [Revised: 04/14/2025] [Accepted: 04/22/2025] [Indexed: 05/07/2025]
Abstract
The applications of epigenetic clocks - statistical models that predict an individual's age based on DNA methylation patterns - are expanding in wildlife conservation and management. This growing interest highlights the need for field-specific design best practices. Here, we provide recommendations for two main applications of wildlife epigenetic clocks: estimating the unknown ages of individuals and assessing their biological ageing rates. Epigenetic clocks were originally developed to measure biological ageing rates of human tissues, which presents challenges for their adoption in wildlife research. Most notably, the estimated chronological ages of sampled wildlife can be unreliable, and sampling restrictions limit the number and variety of tissues with which epigenetic clocks can be constructed, reducing their accuracy. To address these challenges, we present a detailed workflow for designing, validating applying accurate wildlife epigenetic clocks. Using simulations and analyses applied to an extensive polar bear dataset from across the Canadian Arctic, we demonstrate that accurate epigenetic clocks for wildlife can be constructed and validated using a limited number of samples, accommodating projects with small budgets and sampling constraints. The concerns we address are critical for clock design, whether researchers or third-party service providers perform the bioinformatics. With our workflow and examples, we hope to support the accessible and widespread use of epigenetic clocks in wildlife conservation and management.
Collapse
Affiliation(s)
- Levi Newediuk
- Department of Biological Sciences, University of Manitoba, Winnipeg, Manitoba, Canada
| | | | - Alyssa M Bohart
- Department of Environment, Government of Nunavut, Iqaluit, Nunavut, Canada
| | | | - Colin J Garroway
- Department of Biological Sciences, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Meaghan J Jones
- Department of Biochemistry and Medical Genetics, University of Manitoba Children's Hospital Research Institute of Manitoba, Winnipeg, Manitoba, Canada
| |
Collapse
|
3
|
Teschendorff AE, Horvath S. Epigenetic ageing clocks: statistical methods and emerging computational challenges. Nat Rev Genet 2025; 26:350-368. [PMID: 39806006 DOI: 10.1038/s41576-024-00807-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/20/2024] [Indexed: 01/16/2025]
Abstract
Over the past decade, epigenetic clocks have emerged as powerful machine learning tools, not only to estimate chronological and biological age but also to assess the efficacy of anti-ageing, cellular rejuvenation and disease-preventive interventions. However, many computational and statistical challenges remain that limit our understanding, interpretation and application of epigenetic clocks. Here, we review these computational challenges, focusing on interpretation, cell-type heterogeneity and emerging single-cell methods, aiming to provide guidelines for the rigorous construction of interpretable epigenetic clocks at cell-type and single-cell resolution.
Collapse
Affiliation(s)
- Andrew E Teschendorff
- CAS Key Laboratory of Computational Biology, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China.
| | | |
Collapse
|
4
|
Fries GR, Garza SDL, Zhao NO, Bass AW, Lima CNC, Kobori N, Barichello T, Turecki G, Schulz PE, Diniz BS, Soares JC. Association between epigenetic aging acceleration and amyloid biomarkers in bipolar disorder. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2025:2025.04.06.25325186. [PMID: 40297439 PMCID: PMC12036414 DOI: 10.1101/2025.04.06.25325186] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 04/30/2025]
Abstract
Objectives Bipolar disorder (BD) has been associated with an elevated risk of Alzheimer's Disease (AD). We assessed AD biomarkers in BD and tested whether epigenetic aging (EA) acceleration is a potential mechanism driving variability in these markers. Design Setting Participants Cross-sectional study of n=59 living individuals with BD and n=20 age- and sex-equated control participants, as well as analyses of postmortem brain samples (Brodmann area 9/46) from n=46 individuals with BD. Measurements Amyloid beta (Aβ)40, Aβ42, and total Tau levels were measured in plasma from individuals with BD and controls, and Aβ42 levels were measured in brains. EA and its acceleration (blood: GrimAge and DunedinPACE; brains: DNAmClockCortical) were estimated for all samples. Individuals with BD were split into quartiles with accelerated or slower EA if they were in the first or fourth quartiles for GrimAge acceleration (AgeAccelGrim), DunedinPACE, or DNAmClockCortical acceleration (DNAmClockCorticalAccel). Results Individuals with BD showed an increase in Aβ40 (p=.049) and a decrease in the Aβ42/40 ratio (p=.035) compared to controls. A decrease in the Aβ42/40 ratio was also found in individuals with BD with high versus low AgeAccelGrim (p=.028). Brain Aβ42 levels significantly correlated with DNAmClockCorticalAccel (r2=.270, p=.007), with those with high EA acceleration showing higher brain Aβ42 after controlling for confounders (p=.008). Conclusions Our results provide preliminary evidence that EA may explain the variability in AD risk in individuals with BD and could act as a target for preventing dementia and AD in BD.
Collapse
Affiliation(s)
- Gabriel R. Fries
- Translational Psychiatry Program, Faillace Department of Psychiatry and Behavioral Sciences, McGovern Medical School, The University of Texas Health Science Center at Houston, 1941 East Rd, Houston, Texas, USA
- Neuroscience Graduate Program, The University of Texas MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, 6767 Bertner Ave, Houston, Texas, USA
- Center of Excellence in Mood Disorders, Faillace Department of Psychiatry and Behavioral Sciences, McGovern Medical School, The University of Texas Health Science Center at Houston, 1941 East Rd, Houston, Texas, USA
| | - Steven De La Garza
- Translational Psychiatry Program, Faillace Department of Psychiatry and Behavioral Sciences, McGovern Medical School, The University of Texas Health Science Center at Houston, 1941 East Rd, Houston, Texas, USA
| | - Ning O. Zhao
- Translational Psychiatry Program, Faillace Department of Psychiatry and Behavioral Sciences, McGovern Medical School, The University of Texas Health Science Center at Houston, 1941 East Rd, Houston, Texas, USA
| | - Andres W. Bass
- Translational Psychiatry Program, Faillace Department of Psychiatry and Behavioral Sciences, McGovern Medical School, The University of Texas Health Science Center at Houston, 1941 East Rd, Houston, Texas, USA
| | - Camila N. C. Lima
- Translational Psychiatry Program, Faillace Department of Psychiatry and Behavioral Sciences, McGovern Medical School, The University of Texas Health Science Center at Houston, 1941 East Rd, Houston, Texas, USA
| | - Nobuhide Kobori
- Department of Neurobiology and Anatomy, McGovern Medical School, The University of Texas Health Science Center at Houston, 1941 East Rd, Houston, Texas, USA
| | - Tatiana Barichello
- Translational Psychiatry Program, Faillace Department of Psychiatry and Behavioral Sciences, McGovern Medical School, The University of Texas Health Science Center at Houston, 1941 East Rd, Houston, Texas, USA
- Neuroscience Graduate Program, The University of Texas MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, 6767 Bertner Ave, Houston, Texas, USA
| | - Gustavo Turecki
- Douglas Mental Health University Institute, Department of Psychiatry, McGill University, Montreal, Quebec, Canada
| | - Paul E. Schulz
- Department of Neurology, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA; Neurocognitive Disorders Center, McGovern Medical School, The University of Texas Health Science Center at Houston Neurosciences, Houston, Texas, USA
| | - Breno S. Diniz
- UConn Center on Aging & Department of Psychiatry, UConn School of Medicine, University of Connecticut Health Center, USA
| | - Jair C. Soares
- Translational Psychiatry Program, Faillace Department of Psychiatry and Behavioral Sciences, McGovern Medical School, The University of Texas Health Science Center at Houston, 1941 East Rd, Houston, Texas, USA
- Neuroscience Graduate Program, The University of Texas MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, 6767 Bertner Ave, Houston, Texas, USA
- Center of Excellence in Mood Disorders, Faillace Department of Psychiatry and Behavioral Sciences, McGovern Medical School, The University of Texas Health Science Center at Houston, 1941 East Rd, Houston, Texas, USA
| |
Collapse
|
5
|
Kusters CDJ, Horvath S. Quantification of Epigenetic Aging in Public Health. Annu Rev Public Health 2025; 46:91-110. [PMID: 39681336 DOI: 10.1146/annurev-publhealth-060222-015657] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2024]
Abstract
Estimators of biological age hold promise for use in preventive medicine, for early detection of chronic conditions, and for monitoring the effectiveness of interventions aimed at improving population health. Among the promising biomarkers in this field are DNA methylation-based biomarkers, commonly referred to as epigenetic clocks. This review provides a survey of these clocks, with an emphasis on second-generation clocks that predict human morbidity and mortality. It explores the validity of epigenetic clocks when considering factors such as race, sex differences, lifestyle, and environmental influences. Furthermore, the review addresses the current challenges and limitations in this research area.
Collapse
Affiliation(s)
- Cynthia D J Kusters
- Department of Epidemiology, Fielding School of Public Health, University of California, Los Angeles, California, USA;
| | - Steve Horvath
- Altos Labs, Cambridge, United Kingdom;
- Department of Biostatistics, Fielding School of Public Health, University of California, Los Angeles, California, USA
| |
Collapse
|
6
|
Gao Q, Wang J, Fang R, Sun H, Wang T. A doubly robust estimator for continuous treatments in high dimensions. BMC Med Res Methodol 2025; 25:35. [PMID: 39948447 PMCID: PMC11823051 DOI: 10.1186/s12874-025-02488-3] [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: 02/02/2024] [Accepted: 02/03/2025] [Indexed: 02/17/2025] Open
Abstract
BACKGROUND Generalized propensity score (GPS) methods have become popular for estimating causal relationships between a continuous treatment and an outcome in observational studies with rich covariate information. The presence of rich covariates enhances the plausibility of the unconfoundedness assumption. Nonetheless, it is also crucial to ensure the correct specification of both marginal and conditional treatment distributions, beyond the assumption of unconfoundedness. METHOD We address limitations in existing GPS methods by extending balance-based approaches to high dimensions and introducing the Generalized Outcome-Adaptive LASSO and Doubly Robust Estimate (GOALDeR). This novel approach integrates a balance-based method that is robust to the misspecification of distributions required for GPS methods, a doubly robust estimator that is robust to the misspecification of models, and a variable selection technique for causal inference that ensures an unbiased and statistically efficient estimation. RESULTS Simulation studies showed that GOALDeR was able to generate nearly unbiased estimates when either the GPS model or the outcome model was correctly specified. Notably, GOALDeR demonstrated greater precision and accuracy compared to existing methods and was slightly affected by the covariate correlation structure and ratio of sample size to covariate dimension. Real data analysis revealed no statistically significant dose-response relationship between epigenetic age acceleration and Alzheimer's disease. CONCLUSION In this study, we proposed GOALDeR as an advanced GPS method for causal inference in high dimensions, and empirically demonstrated that GOALDeR is doubly robust, with improved accuracy and precision compared to existing methods. The R package is available at https://github.com/QianGao-SXMU/GOALDeR .
Collapse
Affiliation(s)
- Qian Gao
- Department of Health Statistics, School of Public Health, MOE Key Laboratory of Coal Environmental Pathogenicity and Prevention, Shanxi Medical University, No.56 Xinjian South Road, Taiyuan, 030001, China
| | - Jiale Wang
- Department of Health Statistics, School of Public Health, MOE Key Laboratory of Coal Environmental Pathogenicity and Prevention, Shanxi Medical University, No.56 Xinjian South Road, Taiyuan, 030001, China
| | - Ruiling Fang
- Department of Health Statistics, School of Public Health, MOE Key Laboratory of Coal Environmental Pathogenicity and Prevention, Shanxi Medical University, No.56 Xinjian South Road, Taiyuan, 030001, China
| | - Hongwei Sun
- Department of Health Statistics, School of Public Health, Binzhou Medical University, Yantai, China
| | - Tong Wang
- Department of Health Statistics, School of Public Health, MOE Key Laboratory of Coal Environmental Pathogenicity and Prevention, Shanxi Medical University, No.56 Xinjian South Road, Taiyuan, 030001, China.
| |
Collapse
|
7
|
Gunter‐Rahman F, Adams CD, Raju RM, Zhang Y, Lee EA, Messerlian C. Multiomic profiling reveals timing of menopause predicts prefrontal cortex aging and cognitive function. Aging Cell 2025; 24:e14395. [PMID: 39501567 PMCID: PMC11822667 DOI: 10.1111/acel.14395] [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: 05/25/2024] [Revised: 08/30/2024] [Accepted: 10/15/2024] [Indexed: 02/14/2025] Open
Abstract
A new case of dementia is diagnosed every 3 s. Beyond age, risk prediction of dementia is challenging. There is growing evidence of underlying processes that connect aging across organ systems and may provide insight for early detection, and there is a need to identify early biomarkers at an age when action can be taken to mitigate cognitive decline. We hypothesized that timing of menopause, a marker of ovarian aging, predicts brain age decades later. We used 2086 subjects with multiple "omics" measurements from post-mortem brain samples. Age at menopause (AAM) is positively correlated with cognitive function and negatively correlated with pre-frontal cortex aging acceleration (calculated as estimated biological age from DNA methylation minus chronological age). Genetic correlations showed that at least part of these associations is derived from shared heritability. To dissect the mechanism linking AAM to cognitive decline, we turned to transcriptomic data which confirmed that later AAM was associated with gene expression in pre-frontal cortex consistent with better cognition, and among those who reached menopause naturally, decreased gene expression of pathways implicated in aging. Those with surgical menopause displayed different molecular changes, including perturbed nicotinamide adenine dinucleotide (NAD+) activity, validated by metabolomics. Bile acid metabolism was perturbed in both groups, although different bile acid ratios were associated with AAM in each. Together, our data suggest that AAM is predictive of brain aging and cognition, with potential mediation by the gut, although through different mechanisms depending on the type of menopause.
Collapse
Affiliation(s)
- Fatima Gunter‐Rahman
- Harvard‐MIT Program in Health Sciences and TechnologyMassachusetts Institute of TechnologyCambridgeMassachusettsUSA
- Division of Genetics and GenomicsBoston Children's HospitalBostonMassachusettsUSA
| | - Charleen D. Adams
- Department of Environmental HealthHarvard T.H. Chan School of Public HealthBostonMassachusettsUSA
| | - Ravikiran M. Raju
- Division of Newborn Medicine, Boston Children's HospitalHarvard Medical SchoolBostonMassachusettsUSA
- Picower Institute for Learning and MemoryMassachusetts Institute of TechnologyCambridgeMassachusettsUSA
| | - Yu Zhang
- Department of Environmental HealthHarvard T.H. Chan School of Public HealthBostonMassachusettsUSA
| | - Eunjung Alice Lee
- Division of Genetics and GenomicsBoston Children's HospitalBostonMassachusettsUSA
- Department of PediatricsHarvard Medical SchoolBostonMassachusettsUSA
- Broad Institute of MIT and HarvardCambridgeMassachusettsUSA
| | - Carmen Messerlian
- Department of Environmental HealthHarvard T.H. Chan School of Public HealthBostonMassachusettsUSA
- Department of EpidemiologyHarvard T.H. Chan School of Public HealthBostonMassachusettsUSA
- Department of Obstetrics and Gynecology, Vincent Center for Reproductive BiologyMassachusetts General Hospital Fertility CenterBostonMassachusettsUSA
| |
Collapse
|
8
|
Pandey KB. From bench to bedside: translational insights into aging research. FRONTIERS IN AGING 2025; 6:1492099. [PMID: 39926027 PMCID: PMC11802818 DOI: 10.3389/fragi.2025.1492099] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/06/2024] [Accepted: 01/09/2025] [Indexed: 02/11/2025]
Abstract
Aging research has rapidly advanced from fundamental discoveries at the molecular and cellular levels to promising clinical applications. This review discusses the critical translational insights that bridge the gap between bench research and bedside applications, highlighting key discoveries in the mechanisms of aging, biomarkers, and therapeutic interventions. It underscores the importance of interdisciplinary approaches and collaboration among scientists, clinicians, and policymakers to address the complexities of aging and improve health span.
Collapse
Affiliation(s)
- Kanti Bhooshan Pandey
- CSIR-Central Salt & Marine Chemicals Research Institute, Bhavnagar, Gujarat, India
- Faculty of Biological Sciences, Academy of Scientific and Innovative Research, Ghaziabad, Uttar Pradesh, India
| |
Collapse
|
9
|
Kiselev IS, Baulina NM, Favorova OO. Epigenetic Clock: DNA Methylation as a Marker of Biological Age and Age-Associated Diseases. BIOCHEMISTRY. BIOKHIMIIA 2025; 90:S356-S372. [PMID: 40164166 DOI: 10.1134/s0006297924602843] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Revised: 07/11/2024] [Accepted: 07/20/2024] [Indexed: 04/02/2025]
Abstract
Age is one of the key criteria of human health used in practical medicine to predict the risk of common chronic diseases. However, biological age, which reflects the state of an individual organism, functional capabilities, social well-being, and risk of premature death from various causes, often does not coincide with chronological age. To determine biological age of a particular individuals and the rate of their aging, specific panels of DNA methylation markers called "epigenetic clock" (EC) were proposed. This review summarizes the data about the main types of ECs developed to date and their key characteristics. We described the results of works studying individual aging rates in common age-associated diseases and outlined main directions, development of which could expand application of ECs in fundamental and practical medicine. There is no doubt that revealing complex mechanisms underlying interaction between the rate of epigenetic aging and the risk of age-associated diseases could play a key role for prediction and early diagnosis, as well as for the development of preventive measures that could delay onset of the disease.
Collapse
Affiliation(s)
- Ivan S Kiselev
- Chazov National Medical Research Center of Cardiology, Ministry of Health of the Russian Federation, Moscow, 121552, Russia.
- Pirogov Russian National Research Medical University, Ministry of Health of the Russian Federation, Moscow, 117513, Russia
| | - Natalia M Baulina
- Chazov National Medical Research Center of Cardiology, Ministry of Health of the Russian Federation, Moscow, 121552, Russia
- Pirogov Russian National Research Medical University, Ministry of Health of the Russian Federation, Moscow, 117513, Russia
| | - Olga O Favorova
- Chazov National Medical Research Center of Cardiology, Ministry of Health of the Russian Federation, Moscow, 121552, Russia
- Pirogov Russian National Research Medical University, Ministry of Health of the Russian Federation, Moscow, 117513, Russia
| |
Collapse
|
10
|
Wang M, Yan H, Zhang Y, Zhou Q, Meng X, Lin J, Jiang Y, Pan Y, Wang Y. Accelerated biological aging increases the risk of short- and long-term stroke prognosis in patients with ischemic stroke or TIA. EBioMedicine 2025; 111:105494. [PMID: 39662178 PMCID: PMC11697706 DOI: 10.1016/j.ebiom.2024.105494] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2024] [Revised: 11/16/2024] [Accepted: 11/26/2024] [Indexed: 12/13/2024] Open
Abstract
BACKGROUND Biological age (BA), an integrated measure of physiological aging, has a clear link to stroke. There is a paucity of long-term longitudinal studies about the association between accelerated biological age and stroke prognosis in patients with previous strokes, and the differences in the predictive ability of various BA indicators calculated from clinical biochemistry biomarkers for future stroke outcomes are still unknown. To evaluate the role of three accelerated BA indicators for short- and long-term prognosis of patients with ischemic stroke or transient ischemic attack (TIA), and to identify the most appropriate predictor. METHODS This study included 7396 patients from the Third China National Stroke Registry (CNSR-III), a prospective national registry of patients with acute ischemic stroke or TIA between August 2015 and March 2018 in China. We constructed accelerated BA using three widely recognized algorithms: PhenoAge, Klemera-Doubal, and HD method. To ascertain the association of accelerated BA with the risk of short- and long-term stroke outcomes, a Cox or logistic regression model was conducted for the analysis. The net reclassification index and integrated discrimination improvement were used to evaluate the added model improvement ability of BA acceleration. FINDINGS Compared to those with the lowest of PhenoAge acceleration, patients with the highest were more likely to have a higher risk of stroke (HR 1.98, 95% CI 1.49-2.63, P < 0.001), ischemic stroke (HR 1.88, 95% CI 1.41-2.53, P < 0.001), composite vascular events (HR 2.03, 95% CI 1.53-2.68, P < 0.001), all-cause death (HR 7.02, 95% CI 3.41-14.47, P < 0.001) and the modified Rankin scale of 3-6 (OR 2.55, 95% CI 2.05-3.16, P < 0.001) at three months, and the association observed within one year and five years was similar to that within three months. The risk of all stroke outcomes for HDAge was consistent with PhenoAge acceleration, but KDMAge acceleration was the same, except for stroke within one year (HR 1.24, 95% CI 1.00-1.53, P = 0.053). PhenoAge acceleration provided a better improvement in the model's predictive ability for stroke prognosis, compared to BA determined by other algorithms. INTERPRETATION In this prospective cohort study, BA acceleration, particularly PhenoAge, may help identify stroke patients with risks of short- and long-term poor outcomes, potentially enabling subclinical prevention and early intervention. FUNDING This work was supported by grants from Chinese Academy of Medical Sciences Innovation Fund for Medical Sciences (2019-I2M-5-029), the National Natural Science Foundation of China (U20A20358), Beijing Hospitals Authority Clinical Medicine Development of special funding support (ZLRK202312), the National Key R&D Program of China (No. 2022YFC3602500, 2022YFC3602505), Outstanding Young Talents Project of Capital Medical University (A2105), and Beijing High-Level Public Health Technical Personnel Construction Project (Discipline leader -03-12).
Collapse
Affiliation(s)
- Mengxing Wang
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China; China National Clinical Research Centre for Neurological Diseases, Beijing, China
| | - Hongyi Yan
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China; China National Clinical Research Centre for Neurological Diseases, Beijing, China
| | - Yanli Zhang
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China; China National Clinical Research Centre for Neurological Diseases, Beijing, China
| | - Qi Zhou
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China; China National Clinical Research Centre for Neurological Diseases, Beijing, China
| | - Xia Meng
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China; China National Clinical Research Centre for Neurological Diseases, Beijing, China
| | - Jinxi Lin
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China; China National Clinical Research Centre for Neurological Diseases, Beijing, China
| | - Yong Jiang
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China; China National Clinical Research Centre for Neurological Diseases, Beijing, China
| | - Yuesong Pan
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China; China National Clinical Research Centre for Neurological Diseases, Beijing, China.
| | - Yongjun Wang
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China; China National Clinical Research Centre for Neurological Diseases, Beijing, China.
| |
Collapse
|
11
|
Tong H, Guo X, Jacques M, Luo Q, Eynon N, Teschendorff AE. Cell-type specific epigenetic clocks to quantify biological age at cell-type resolution. Aging (Albany NY) 2024; 16:13452-13504. [PMID: 39760516 PMCID: PMC11723652 DOI: 10.18632/aging.206184] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2024] [Accepted: 12/12/2024] [Indexed: 01/07/2025]
Abstract
The ability to accurately quantify biological age could help monitor and control healthy aging. Epigenetic clocks have emerged as promising tools for estimating biological age, yet they have been developed from heterogeneous bulk tissues, and are thus composites of two aging processes, one reflecting the change of cell-type composition with age and another reflecting the aging of individual cell-types. There is thus a need to dissect and quantify these two components of epigenetic clocks, and to develop epigenetic clocks that can yield biological age estimates at cell-type resolution. Here we demonstrate that in blood and brain, approximately 39% and 12% of an epigenetic clock's accuracy is driven by underlying shifts in lymphocyte and neuronal subsets, respectively. Using brain and liver tissue as prototypes, we build and validate neuron and hepatocyte specific DNA methylation clocks, and demonstrate that these cell-type specific clocks yield improved estimates of chronological age in the corresponding cell and tissue-types. We find that neuron and glia specific clocks display biological age acceleration in Alzheimer's Disease with the effect being strongest for glia in the temporal lobe. Moreover, CpGs from these clocks display a small but significant overlap with the causal DamAge-clock, mapping to key genes implicated in neurodegeneration. The hepatocyte clock is found accelerated in liver under various pathological conditions. In contrast, non-cell-type specific clocks do not display biological age-acceleration, or only do so marginally. In summary, this work highlights the importance of dissecting epigenetic clocks and quantifying biological age at cell-type resolution.
Collapse
Affiliation(s)
- Huige Tong
- CAS Key Laboratory of Computational Biology, CAS-MPG Partner Institute for Computational Biology, Shanghai Institute of Nutrition and Health, Shanghai Institute for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Xiaolong Guo
- CAS Key Laboratory of Computational Biology, CAS-MPG Partner Institute for Computational Biology, Shanghai Institute of Nutrition and Health, Shanghai Institute for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Macsue Jacques
- Australian Regenerative Medicine Institute (ARMI), Faculty of Medicine, Nursing and Health Sciences, Monash University, Clayton, Victoria 3800, Australia
| | - Qi Luo
- CAS Key Laboratory of Computational Biology, CAS-MPG Partner Institute for Computational Biology, Shanghai Institute of Nutrition and Health, Shanghai Institute for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Nir Eynon
- Australian Regenerative Medicine Institute (ARMI), Faculty of Medicine, Nursing and Health Sciences, Monash University, Clayton, Victoria 3800, Australia
| | - Andrew E. Teschendorff
- CAS Key Laboratory of Computational Biology, CAS-MPG Partner Institute for Computational Biology, Shanghai Institute of Nutrition and Health, Shanghai Institute for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| |
Collapse
|
12
|
Liang R, Tang Q, Chen J, Zhu L. Epigenetic Clocks: Beyond Biological Age, Using the Past to Predict the Present and Future. Aging Dis 2024:AD.2024.1495. [PMID: 39751861 DOI: 10.14336/ad.2024.1495] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2024] [Accepted: 12/13/2024] [Indexed: 01/04/2025] Open
Abstract
Predicting health trajectories and accurately measuring aging processes across the human lifespan remain profound scientific challenges. Assessing the effectiveness and impact of interventions targeting aging is even more elusive, largely due to the intricate, multidimensional nature of aging-a process that defies simple quantification. Traditional biomarkers offer only partial perspectives, capturing limited aspects of the aging landscape. Yet, over the past decade, groundbreaking advancements have emerged. Epigenetic clocks, derived from DNA methylation patterns, have established themselves as powerful aging biomarkers, capable of estimating biological age and assessing aging rates across diverse tissues with remarkable precision. These clocks provide predictive insights into mortality and age-related disease risks, effectively distinguishing biological age from chronological age and illuminating enduring questions in gerontology. Despite significant progress in epigenetic clock development, substantial challenges remain, underscoring the need for continued investigation to fully unlock their potential in the science of aging.
Collapse
Affiliation(s)
- Runyu Liang
- Heilongjiang University of Chinese Medicine, Harbin, China
| | - Qiang Tang
- Second Affiliated Hospital of Heilongjiang University of Chinese Medicine, Harbin, China
| | - Jia Chen
- Heilongjiang University of Chinese Medicine, Harbin, China
| | - Luwen Zhu
- Second Affiliated Hospital of Heilongjiang University of Chinese Medicine, Harbin, China
| |
Collapse
|
13
|
Jin S, Lu W, Zhang J, Zhang L, Tao F, Zhang Y, Hu X, Liu Q. The mechanisms, hallmarks, and therapies for brain aging and age-related dementia. Sci Bull (Beijing) 2024; 69:3756-3776. [PMID: 39332926 DOI: 10.1016/j.scib.2024.09.005] [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: 04/10/2024] [Revised: 06/14/2024] [Accepted: 09/02/2024] [Indexed: 09/29/2024]
Abstract
Age-related cognitive decline and dementia are significant manifestations of brain aging. As the elderly population grows rapidly, the health and socio-economic impacts of cognitive dysfunction have become increasingly significant. Although clinical treatment of dementia has faced considerable challenges over the past few decades, with limited breakthroughs in slowing its progression, there has been substantial progress in understanding the molecular mechanisms and hallmarks of age-related dementia (ARD). This progress brings new hope for the intervention and treatment of this disease. In this review, we categorize the latest findings in ARD biomarkers into four stages based on disease progression: Healthy brain, pre-clinical, mild cognitive impairment, and dementia. We then systematically summarize the most promising therapeutic approaches to prevent or slow ARD at four levels: Genome and epigenome, organelle, cell, and organ and organism. We emphasize the importance of early prevention and detection, along with the implementation of combined treatments as multimodal intervention strategies, to address brain aging and ARD in the future.
Collapse
Affiliation(s)
- Shiyun Jin
- Department of Neurology, The First Affiliated Hospital of USTC, Center for Advanced Interdisciplinary Science and Biomedicine of IHM, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230001, China; Anhui Province Key Laboratory of Biomedical Aging Research, University of Science and Technology of China, Hefei 230027, China; Department of Anesthesiology, the Second Affiliated Hospital of Anhui Medical University, Hefei 230601, China; Key Laboratory of Anesthesiology and Perioperative Medicine of Anhui Higher Education Institutes, Anhui Medical University, Hefei 230601, China
| | - Wenping Lu
- Department of Anesthesiology, the Second Affiliated Hospital of Anhui Medical University, Hefei 230601, China; Key Laboratory of Anesthesiology and Perioperative Medicine of Anhui Higher Education Institutes, Anhui Medical University, Hefei 230601, China
| | - Juan Zhang
- Department of Neurology, The First Affiliated Hospital of USTC, Center for Advanced Interdisciplinary Science and Biomedicine of IHM, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230001, China; Anhui Province Key Laboratory of Biomedical Aging Research, University of Science and Technology of China, Hefei 230027, China; Institute on Aging and Brain Disorders, The First Affiliated Hospital of USTC, University of Science and Technology of China, Hefei 230027, China
| | - Li Zhang
- Laboratory for Integrative Neuroscience, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD 20892, USA
| | - Fangbiao Tao
- MOE Key Laboratory of Population Health Across Life Cycle, Anhui Medical University, Hefei 230032, China.
| | - Ye Zhang
- Department of Anesthesiology, the Second Affiliated Hospital of Anhui Medical University, Hefei 230601, China; Key Laboratory of Anesthesiology and Perioperative Medicine of Anhui Higher Education Institutes, Anhui Medical University, Hefei 230601, China.
| | - Xianwen Hu
- Department of Anesthesiology, the Second Affiliated Hospital of Anhui Medical University, Hefei 230601, China; Key Laboratory of Anesthesiology and Perioperative Medicine of Anhui Higher Education Institutes, Anhui Medical University, Hefei 230601, China.
| | - Qiang Liu
- Department of Neurology, The First Affiliated Hospital of USTC, Center for Advanced Interdisciplinary Science and Biomedicine of IHM, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230001, China; Anhui Province Key Laboratory of Biomedical Aging Research, University of Science and Technology of China, Hefei 230027, China; Institute on Aging and Brain Disorders, The First Affiliated Hospital of USTC, University of Science and Technology of China, Hefei 230027, China.
| |
Collapse
|
14
|
Lopes CR, Cunha RA. Impact of coffee intake on human aging: Epidemiology and cellular mechanisms. Ageing Res Rev 2024; 102:102581. [PMID: 39557300 DOI: 10.1016/j.arr.2024.102581] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2024] [Revised: 11/09/2024] [Accepted: 11/12/2024] [Indexed: 11/20/2024]
Abstract
The conception of coffee consumption has undergone a profound modification, evolving from a noxious habit into a safe lifestyle actually preserving human health. The last 20 years also provided strikingly consistent epidemiological evidence showing that the regular consumption of moderate doses of coffee attenuates all-cause mortality, an effect observed in over 50 studies in different geographic regions and different ethnicities. Coffee intake attenuates the major causes of mortality, dampening cardiovascular-, cerebrovascular-, cancer- and respiratory diseases-associated mortality, as well as some of the major causes of functional deterioration in the elderly such as loss of memory, depression and frailty. The amplitude of the benefit seems discrete (17 % reduction) but nonetheless corresponds to an average increase in healthspan of 1.8 years of lifetime. This review explores evidence from studies in humans and human tissues supporting an ability of coffee and of its main components (caffeine and chlorogenic acids) to preserve the main biological mechanisms responsible for the aging process, namely genomic instability, macromolecular damage, metabolic and proteostatic impairments with particularly robust effects on the control of stress adaptation and inflammation and unclear effects on stem cells and regeneration. Further studies are required to detail these mechanistic benefits in aged individuals, which may offer new insights into understanding of the biology of aging and the development of new senostatic strategies. Additionally, the safety of this lifestyle factor in the elderly prompts a renewed attention to recommending the maintenance of coffee consumption throughout life as a healthy lifestyle and to further exploring who gets the greater benefit with what schedules of which particular types and doses of coffee.
Collapse
Affiliation(s)
- Cátia R Lopes
- CNC-Center for Neuroscience and Cell Biology, Portugal; Faculty of Medicine, Portugal
| | - Rodrigo A Cunha
- CNC-Center for Neuroscience and Cell Biology, Portugal; Faculty of Medicine, Portugal; MIA-Portugal, Multidisciplinary Institute of Aging, University of Coimbra, Portugal; Centro de Medicina Digital P5, Escola de Medicina da Universidade do Minho, Braga, Portugal.
| |
Collapse
|
15
|
Grodstein F, Lemos B, Yang J, de Paiva Lopes K, Vialle RA, Seyfried N, Wang Y, Shireby G, Hannon E, Thomas A, Brookes K, Mill J, De Jager PL, Bennett DA. Genetic architecture of epigenetic cortical clock age in brain tissue from older individuals: alterations in CD46 and other loci. Epigenetics 2024; 19:2392050. [PMID: 39169872 PMCID: PMC11346548 DOI: 10.1080/15592294.2024.2392050] [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: 03/07/2024] [Revised: 07/18/2024] [Accepted: 08/06/2024] [Indexed: 08/23/2024] Open
Abstract
The cortical epigenetic clock was developed in brain tissue as a biomarker of brain aging. As one way to identify mechanisms underlying aging, we conducted a GWAS of cortical age. We leveraged postmortem cortex tissue and genotyping array data from 694 participants of the Rush Memory and Aging Project and Religious Orders Study (ROSMAP; 11000,000 SNPs), and meta-analysed ROSMAP with 522 participants of Brains for Dementia Research (5,000,000 overlapping SNPs). We confirmed results using eQTL (cortical bulk and single nucleus gene expression), cortical protein levels (ROSMAP), and phenome-wide association studies (clinical/neuropathologic phenotypes, ROSMAP). In the meta-analysis, the strongest association was rs4244620 (p = 1.29 × 10-7), which also exhibited FDR-significant cis-eQTL effects for CD46 in bulk and single nucleus (microglia, astrocyte, oligodendrocyte, neuron) cortical gene expression. Additionally, rs4244620 was nominally associated with lower cognition, faster slopes of cognitive decline, and greater Parkinsonian signs (n ~ 1700 ROSMAP with SNP/phenotypic data; all p ≤ 0.04). In ROSMAP alone, the top SNP was rs4721030 (p = 8.64 × 10-8) annotated to TMEM106B and THSD7A. Further, in ROSMAP (n = 849), TMEM106B and THSD7A protein levels in cortex were related to many phenotypes, including greater AD pathology and lower cognition (all p ≤ 0.0007). Overall, we identified converging evidence of CD46 and possibly TMEM106B/THSD7A for potential roles in cortical epigenetic clock age.
Collapse
Affiliation(s)
- Francine Grodstein
- Rush Alzheimer’s Disease Center, Rush University Medical Center, Chicago, IL, USA
- Department of Internal Medicine, Rush University Medical Center, Chicago, IL, USA
| | - Bernardo Lemos
- Coit Center for Longevity and Neurotherapeutics, Department of Pharmacology and Toxicology, R. Ken Coit College of Pharmacy, The University of Arizona, Tucson, AZ, USA
| | - Jingyun Yang
- Rush Alzheimer’s Disease Center, Rush University Medical Center, Chicago, IL, USA
- Department of Neurological Sciences, Rush University Medical Center, Chicago, IL, USA
| | - Katia de Paiva Lopes
- Rush Alzheimer’s Disease Center, Rush University Medical Center, Chicago, IL, USA
- Department of Neurological Sciences, Rush University Medical Center, Chicago, IL, USA
| | - Ricardo A. Vialle
- Rush Alzheimer’s Disease Center, Rush University Medical Center, Chicago, IL, USA
- Department of Neurological Sciences, Rush University Medical Center, Chicago, IL, USA
| | - Nicholas Seyfried
- Department of Biochemistry, and Center for Neurodegenerative Diseases, Emory University, Atlanta, GA, USA
| | - Yanling Wang
- Rush Alzheimer’s Disease Center, Rush University Medical Center, Chicago, IL, USA
- Department of Neurological Sciences, Rush University Medical Center, Chicago, IL, USA
| | - Gemma Shireby
- Department of Clinical and Biomedical Sciences, University of Exeter Medical School, University of Exeter, Exeter, UK
| | - Eilis Hannon
- Department of Clinical and Biomedical Sciences, University of Exeter Medical School, University of Exeter, Exeter, UK
| | - Alan Thomas
- Translational and Clinical Research Institute, Newcastle University, Newcastle Upon Tyne, UK
| | - Keeley Brookes
- Biosciences, School of Science and Technology, Nottingham Trent University, Nottingham, UK
| | - Jonathan Mill
- Department of Clinical and Biomedical Sciences, University of Exeter Medical School, University of Exeter, Exeter, UK
| | - Philip L. De Jager
- Center for Translational and Computational Neuroimmunology, Department of Neurology, and Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, Columbia University Irving Medical Center, New York, NY, USA
| | - David A. Bennett
- Rush Alzheimer’s Disease Center, Rush University Medical Center, Chicago, IL, USA
- Department of Neurological Sciences, Rush University Medical Center, Chicago, IL, USA
| |
Collapse
|
16
|
Wang X, Liu Y, Qin G, Yu Y. Robust double machine learning model with application to omics data. BMC Bioinformatics 2024; 25:355. [PMID: 39543508 PMCID: PMC11566156 DOI: 10.1186/s12859-024-05975-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2024] [Accepted: 11/04/2024] [Indexed: 11/17/2024] Open
Abstract
BACKGROUND Recently, there has been a growing interest in combining causal inference with machine learning algorithms. Double machine learning model (DML), as an implementation of this combination, has received widespread attention for their expertise in estimating causal effects within high-dimensional complex data. However, the DML model is sensitive to the presence of outliers and heavy-tailed noise in the outcome variable. In this paper, we propose the robust double machine learning (RDML) model to achieve a robust estimation of causal effects when the distribution of the outcome is contaminated by outliers or exhibits symmetrically heavy-tailed characteristics. RESULTS In the modelling of RDML model, we employed median machine learning algorithms to achieve robust predictions for the treatment and outcome variables. Subsequently, we established a median regression model for the prediction residuals. These two steps ensure robust causal effect estimation. Simulation study show that the RDML model is comparable to the existing DML model when the data follow normal distribution, while the RDML model has obvious superiority when the data follow mixed normal distribution and t-distribution, which is manifested by having a smaller RMSE. Meanwhile, we also apply the RDML model to the deoxyribonucleic acid methylation dataset from the Alzheimer's disease (AD) neuroimaging initiative database with the aim of investigating the impact of Cerebrospinal Fluid Amyloid β 42 (CSF A β 42) on AD severity. CONCLUSION These findings illustrate that the RDML model is capable of robustly estimating causal effect, even when the outcome distribution is affected by outliers or displays symmetrically heavy-tailed properties.
Collapse
Affiliation(s)
- Xuqing Wang
- Department of Biostatistics, Key Laboratory of Public Health Safety of Ministry of Education, Key Laboratory for Health Technology Assessment, National Commission of Health, School of Public Health, Fudan University, Shanghai, China
| | - Yahang Liu
- Department of Biostatistics, Key Laboratory of Public Health Safety of Ministry of Education, Key Laboratory for Health Technology Assessment, National Commission of Health, School of Public Health, Fudan University, Shanghai, China
| | - Guoyou Qin
- Department of Biostatistics, Key Laboratory of Public Health Safety of Ministry of Education, Key Laboratory for Health Technology Assessment, National Commission of Health, School of Public Health, Fudan University, Shanghai, China.
- Shanghai Institute of Infectious Disease and Biosecurity, Shanghai, China.
| | - Yongfu Yu
- Department of Biostatistics, Key Laboratory of Public Health Safety of Ministry of Education, Key Laboratory for Health Technology Assessment, National Commission of Health, School of Public Health, Fudan University, Shanghai, China.
- Shanghai Institute of Infectious Disease and Biosecurity, Shanghai, China.
| |
Collapse
|
17
|
Goldberg D, Wadhwani AR, Dehghani N, Sreepada LP, Fu H, De Jager PL, Bennett DA, Wolk DA, Lee EB, Farrell K, Crary JF, Zhou W, McMillan CT. Epigenetic signatures of regional tau pathology and cognition in the aging and pathological brain. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.11.07.24316933. [PMID: 39606399 PMCID: PMC11601699 DOI: 10.1101/2024.11.07.24316933] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/29/2024]
Abstract
Primary age-related tauopathy (PART) and Alzheimer's disease (AD) share hippocampal phospho-tau (p-tau) pathology but differ in p-tau extent and amyloid presence. As a result, PART uniquely enables investigation of amyloid-independent p-tau mechanisms during brain aging. We conducted the first epigenome-wide association (EWAS) study of PART, which yielded 13 new and robust p-tau/methylation associations. We then jointly analyzed PART and AD epigenomes to develop "TauAge", novel epigenetic clocks that predict p-tau severity in region-specific, age-, and amyloid-independent manners. Integrative transcriptomic analyses revealed that genes involved in synaptic transmission are related to hippocampal p-tau severity in both PART and AD, while neuroinflammatory genes are related to frontal cortex p-tau severity in AD only. Further, a machine learning classifier based on PART-vs-AD epigenetic differences discriminates neuropathological diagnoses and stratifies indeterminate cases into subgroups with disparity in cognitive impairment. Together, these findings demonstrate the brain epigenome's substantial role in linking tau pathology to cognitive outcomes in aging and AD.
Collapse
|
18
|
Shastri GG, Sudre G, Ahn K, Jung B, Kolachana B, Auluck PK, Elnitski L, Shaw P. Examining epigenetic aging in the post-mortem brain in attention deficit hyperactivity disorder. Front Genet 2024; 15:1480761. [PMID: 39440240 PMCID: PMC11493619 DOI: 10.3389/fgene.2024.1480761] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2024] [Accepted: 09/24/2024] [Indexed: 10/25/2024] Open
Abstract
Mathematical algorithms known as "epigenetic clocks" use methylation values at a set of CpG sites to estimate the biological age of an individual in a tissue-specific manner. These clocks have demonstrated both acceleration and delays in epigenetic aging in multiple neuropsychiatric conditions, including schizophrenia and neurodevelopmental disorders such as autism spectrum disorder. However, no study to date has examined epigenetic aging in ADHD despite its status as one of the most prevalent neurodevelopmental conditions, with 1 in 9 children having ever received an ADHD diagnosis in the US. Only a handful of studies have examined epigenetic age in brain tissue from neurodevelopmental conditions, with none focused on ADHD, despite the obvious relevance to pathogenesis. Thus, here we asked if post-mortem brain tissue in those with lifetime histories of ADHD would show accelerated or delayed epigenetic age, as has been found for other neurodevelopmental conditions. We applied four different epigenetic clocks to estimate epigenetic age in individuals with ADHD and unaffected controls from cortical (anterior cingulate cortex, N = 55) and striatal (caudate, N = 56) post-mortem brain tissue, as well as peripheral blood (N = 84) and saliva (N = 112). After determining which epigenetic clock performed best in each tissue, we asked if ADHD was associated with altered biological aging in corticostriatal brain and peripheral tissues. We found that a range of epigenetic clocks accurately predicted chronological age in all tissues. We also found that a diagnosis of ADHD was not significantly associated with differential epigenetic aging, neither for the postmortem ACC or caudate, nor for peripheral tissues. These findings held when accounting for comorbid psychiatric diagnoses, substance use, and stimulant medication. Thus, in this study of epigenetic clocks in ADHD, we find no evidence of altered epigenetic aging in corticostriatal brain regions nor in peripheral tissue. We consider reasons for this unexpected finding, including the limited sampling of brain regions, the age range of individuals studied, and the possibility that processes that accelerate epigenetic age may be counteracted by the developmental delay posited in some models of ADHD.
Collapse
Affiliation(s)
- Gauri G. Shastri
- Social and Behavioral Research Branch, National Human Genome Research Institute, NIH, Bethesda, MD, United States
- Zucker Hillside Hospital, Northwell Health, New York, United States
| | - Gustavo Sudre
- Social and Behavioral Research Branch, National Human Genome Research Institute, NIH, Bethesda, MD, United States
| | - Kwangmi Ahn
- Social and Behavioral Research Branch, National Human Genome Research Institute, NIH, Bethesda, MD, United States
| | - Benjamin Jung
- Social and Behavioral Research Branch, National Human Genome Research Institute, NIH, Bethesda, MD, United States
| | - Bhaskar Kolachana
- Human Brain Collection Core, National Institute of Mental Health, NIH, Bethesda, MD, United States
| | - Pavan K. Auluck
- Human Brain Collection Core, National Institute of Mental Health, NIH, Bethesda, MD, United States
| | - Laura Elnitski
- Translational and Functional Genomics Branch, National Human Genome Research Institute, NIH, Bethesda, MD, United States
| | - Philip Shaw
- Social and Behavioral Research Branch, National Human Genome Research Institute, NIH, Bethesda, MD, United States
- Pears Maudsley Center for Children and Young People, King’s College, Institute of Psychiatry, Psychology and Neuroscience, London, United Kingdom
| |
Collapse
|
19
|
Fröhlich AS, Gerstner N, Gagliardi M, Ködel M, Yusupov N, Matosin N, Czamara D, Sauer S, Roeh S, Murek V, Chatzinakos C, Daskalakis NP, Knauer-Arloth J, Ziller MJ, Binder EB. Single-nucleus transcriptomic profiling of human orbitofrontal cortex reveals convergent effects of aging and psychiatric disease. Nat Neurosci 2024; 27:2021-2032. [PMID: 39227716 PMCID: PMC11452345 DOI: 10.1038/s41593-024-01742-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2023] [Accepted: 07/30/2024] [Indexed: 09/05/2024]
Abstract
Aging is a complex biological process and represents the largest risk factor for neurodegenerative disorders. The risk for neurodegenerative disorders is also increased in individuals with psychiatric disorders. Here, we characterized age-related transcriptomic changes in the brain by profiling ~800,000 nuclei from the orbitofrontal cortex from 87 individuals with and without psychiatric diagnoses and replicated findings in an independent cohort with 32 individuals. Aging affects all cell types, with LAMP5+LHX6+ interneurons, a cell-type abundant in primates, by far the most affected. Disrupted synaptic transmission emerged as a convergently affected pathway in aged tissue. Age-related transcriptomic changes overlapped with changes observed in Alzheimer's disease across multiple cell types. We find evidence for accelerated transcriptomic aging in individuals with psychiatric disorders and demonstrate a converging signature of aging and psychopathology across multiple cell types. Our findings shed light on cell-type-specific effects and biological pathways underlying age-related changes and their convergence with effects driven by psychiatric diagnosis.
Collapse
Affiliation(s)
- Anna S Fröhlich
- Department of Genes and Environment, Max Planck Institute of Psychiatry, Munich, Germany.
- International Max Planck Research School for Translational Psychiatry, Munich, Germany.
| | - Nathalie Gerstner
- Department of Genes and Environment, Max Planck Institute of Psychiatry, Munich, Germany
- International Max Planck Research School for Translational Psychiatry, Munich, Germany
- Institute of Computational Biology, Helmholtz Zentrum München, Neuherberg, Germany
| | - Miriam Gagliardi
- Department of Psychiatry, University of Münster, Münster, Germany
| | - Maik Ködel
- Department of Genes and Environment, Max Planck Institute of Psychiatry, Munich, Germany
| | - Natan Yusupov
- Department of Genes and Environment, Max Planck Institute of Psychiatry, Munich, Germany
- International Max Planck Research School for Translational Psychiatry, Munich, Germany
| | - Natalie Matosin
- School of Medical Sciences, Faculty of Medicine and Health, University of Sydney, Camperdown, New South Wales, Australia
- Charles Perkins Centre, University of Sydney, Camperdown, New South Wales, Australia
| | - Darina Czamara
- Department of Genes and Environment, Max Planck Institute of Psychiatry, Munich, Germany
| | - Susann Sauer
- Department of Genes and Environment, Max Planck Institute of Psychiatry, Munich, Germany
| | - Simone Roeh
- Department of Genes and Environment, Max Planck Institute of Psychiatry, Munich, Germany
| | - Vanessa Murek
- Department of Psychiatry, University of Münster, Münster, Germany
| | - Chris Chatzinakos
- Department of Psychiatry, McLean Hospital, Harvard Medical School, Belmont, MA, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Psychiatry and Behavioral Sciences, Institute for Genomics in Health, SUNY Downstate Health Sciences University, Brooklyn, NY, USA
| | - Nikolaos P Daskalakis
- Department of Psychiatry, McLean Hospital, Harvard Medical School, Belmont, MA, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Janine Knauer-Arloth
- Department of Genes and Environment, Max Planck Institute of Psychiatry, Munich, Germany
- Institute of Computational Biology, Helmholtz Zentrum München, Neuherberg, Germany
| | - Michael J Ziller
- Department of Psychiatry, University of Münster, Münster, Germany
| | - Elisabeth B Binder
- Department of Genes and Environment, Max Planck Institute of Psychiatry, Munich, Germany.
- Department of Psychiatry and Behavioral Sciences, Emory University School of Medicine, Atlanta, GA, USA.
| |
Collapse
|
20
|
Martínez-Magaña JJ, Hurtado-Soriano J, Rivero-Segura NA, Montalvo-Ortiz JL, Garcia-delaTorre P, Becerril-Rojas K, Gomez-Verjan JC. Towards a Novel Frontier in the Use of Epigenetic Clocks in Epidemiology. Arch Med Res 2024; 55:103033. [PMID: 38955096 DOI: 10.1016/j.arcmed.2024.103033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Revised: 05/10/2024] [Accepted: 06/17/2024] [Indexed: 07/04/2024]
Abstract
Health problems associated with aging are a major public health concern for the future. Aging is a complex process with wide intervariability among individuals. Therefore, there is a need for innovative public health strategies that target factors associated with aging and the development of tools to assess the effectiveness of these strategies accurately. Novel approaches to measure biological age, such as epigenetic clocks, have become relevant. These clocks use non-sequential variable information from the genome and employ mathematical algorithms to estimate biological age based on DNA methylation levels. Therefore, in the present study, we comprehensively review the current status of the epigenetic clocks and their associations across the human phenome. We emphasize the potential utility of these tools in an epidemiological context, particularly in evaluating the impact of public health interventions focused on promoting healthy aging. Our review describes associations between epigenetic clocks and multiple traits across the life and health span. Additionally, we highlighted the evolution of studies beyond mere associations to establish causal mechanisms between epigenetic age and disease. We explored the application of epigenetic clocks to measure the efficacy of interventions focusing on rejuvenation.
Collapse
Affiliation(s)
- José Jaime Martínez-Magaña
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA; U.S. Department of Veterans Affairs National Center for Post-Traumatic Stress Disorder, Clinical Neuroscience Division, West Haven, CT, USA; VA Connecticut Healthcare System, West Haven, CT, USA
| | | | | | - Janitza L Montalvo-Ortiz
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA; U.S. Department of Veterans Affairs National Center for Post-Traumatic Stress Disorder, Clinical Neuroscience Division, West Haven, CT, USA; VA Connecticut Healthcare System, West Haven, CT, USA
| | - Paola Garcia-delaTorre
- Unidad de Investigación Epidemiológica y en Servicios de Salud, Área de Envejecimiento, Centro Médico Nacional, Siglo XXI, Instituto Mexicano del Seguro Social, Mexico City, Mexico
| | | | | |
Collapse
|
21
|
Villa C, Combi R. Epigenetics in Alzheimer's Disease: A Critical Overview. Int J Mol Sci 2024; 25:5970. [PMID: 38892155 PMCID: PMC11173284 DOI: 10.3390/ijms25115970] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Revised: 05/27/2024] [Accepted: 05/28/2024] [Indexed: 06/21/2024] Open
Abstract
Epigenetic modifications have been implicated in a number of complex diseases as well as being a hallmark of organismal aging. Several reports have indicated an involvement of these changes in Alzheimer's disease (AD) risk and progression, most likely contributing to the dysregulation of AD-related gene expression measured by DNA methylation studies. Given that DNA methylation is tissue-specific and that AD is a brain disorder, the limitation of these studies is the ability to identify clinically useful biomarkers in a proxy tissue, reflective of the tissue of interest, that would be less invasive, more cost-effective, and easily obtainable. The age-related DNA methylation changes have also been used to develop different generations of epigenetic clocks devoted to measuring the aging in different tissues that sometimes suggests an age acceleration in AD patients. This review critically discusses epigenetic changes and aging measures as potential biomarkers for AD detection, prognosis, and progression. Given that epigenetic alterations are chemically reversible, treatments aiming at reversing these modifications will be also discussed as promising therapeutic strategies for AD.
Collapse
Affiliation(s)
| | - Romina Combi
- School of Medicine and Surgery, University of Milano-Bicocca, 20900 Monza, Italy;
| |
Collapse
|
22
|
Ravaioli F, Stagni F, Guidi S, Pirazzini C, Garagnani P, Silvani A, Zoccoli G, Bartesaghi R, Bacalini MG. Increased hippocampal epigenetic age in the Ts65Dn mouse model of Down Syndrome. Front Aging Neurosci 2024; 16:1401109. [PMID: 38836050 PMCID: PMC11148439 DOI: 10.3389/fnagi.2024.1401109] [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: 03/14/2024] [Accepted: 05/06/2024] [Indexed: 06/06/2024] Open
Abstract
Down syndrome (DS) is a segmental progeroid genetic disorder associated with multi-systemic precocious aging phenotypes, which are particularly evident in the immune and nervous systems. Accordingly, people with DS show an increased biological age as measured by epigenetic clocks. The Ts65Dn trisomic mouse, which harbors extra-numerary copies of chromosome 21 (Hsa21)-syntenic regions, was shown to recapitulate several progeroid features of DS, but no biomarkers of age have been applied to it so far. In this pilot study, we used a mouse-specific epigenetic clock to measure the epigenetic age of hippocampi from Ts65Dn and euploid mice at 20 weeks. Ts65Dn mice showed an increased epigenetic age in comparison with controls, and the observed changes in DNA methylation partially recapitulated those observed in hippocampi from people with DS. Collectively, our results support the use of the Ts65Dn model to decipher the molecular mechanisms underlying the progeroid DS phenotypes.
Collapse
Affiliation(s)
| | - Fiorenza Stagni
- Department for Life Quality Studies, University of Bologna, Rimini, Italy
| | - Sandra Guidi
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| | - Chiara Pirazzini
- Department of Medical and Surgical Sciences (DIMEC), University of Bologna, Bologna, Italy
| | - Paolo Garagnani
- Department of Medical and Surgical Sciences (DIMEC), University of Bologna, Bologna, Italy
- IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
| | - Alessandro Silvani
- PRISM Lab, Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| | - Giovanna Zoccoli
- PRISM Lab, Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| | - Renata Bartesaghi
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| | | |
Collapse
|
23
|
Cai Y, Han Z, Cheng H, Li H, Wang K, Chen J, Liu ZX, Xie Y, Lin Y, Zhou S, Wang S, Zhou X, Jin S. The impact of ageing mechanisms on musculoskeletal system diseases in the elderly. Front Immunol 2024; 15:1405621. [PMID: 38774874 PMCID: PMC11106385 DOI: 10.3389/fimmu.2024.1405621] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2024] [Accepted: 04/22/2024] [Indexed: 05/24/2024] Open
Abstract
Ageing is an inevitable process that affects various tissues and organs of the human body, leading to a series of physiological and pathological changes. Mechanisms such as telomere depletion, stem cell depletion, macrophage dysfunction, and cellular senescence gradually manifest in the body, significantly increasing the incidence of diseases in elderly individuals. These mechanisms interact with each other, profoundly impacting the quality of life of older adults. As the ageing population continues to grow, the burden on the public health system is expected to intensify. Globally, the prevalence of musculoskeletal system diseases in elderly individuals is increasing, resulting in reduced limb mobility and prolonged suffering. This review aims to elucidate the mechanisms of ageing and their interplay while exploring their impact on diseases such as osteoarthritis, osteoporosis, and sarcopenia. By delving into the mechanisms of ageing, further research can be conducted to prevent and mitigate its effects, with the ultimate goal of alleviating the suffering of elderly patients in the future.
Collapse
Affiliation(s)
- Yijin Cai
- School of Medical and Life Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Zhongyu Han
- School of Medical and Life Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Hong Cheng
- School of Automation Engineering, University of Electronic Science and Technology, Chengdu, China
| | - Hongpeng Li
- School of Medical and Life Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Ke Wang
- Eye School of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Jia Chen
- School of Health Preservation and Rehabilitation, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Zhi-Xiang Liu
- School of Health Preservation and Rehabilitation, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yulong Xie
- School of Health Preservation and Rehabilitation, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yumeng Lin
- Eye School of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Shuwei Zhou
- Jiangsu Key Laboratory of Molecular and Functional Imaging, Department of Radiology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, China
| | - Siyu Wang
- Department of Gastroenterology, The First Hospital of Hunan University of Chinese Medicine, Changsha, China
| | - Xiao Zhou
- Second Clinical Medical College, Heilongjiang University of Chinese Medicine, Heilongjiang, China
| | - Song Jin
- Department of Rehabilitation, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| |
Collapse
|
24
|
Zhao J, Li H, Qu J, Zong X, Liu Y, Kuang Z, Wang H. A multi-organization epigenetic age prediction based on a channel attention perceptron networks. Front Genet 2024; 15:1393856. [PMID: 38725481 PMCID: PMC11080615 DOI: 10.3389/fgene.2024.1393856] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2024] [Accepted: 04/09/2024] [Indexed: 05/12/2024] Open
Abstract
DNA methylation indicates the individual's aging, so-called Epigenetic clocks, which will improve the research and diagnosis of aging diseases by investigating the correlation between methylation loci and human aging. Although this discovery has inspired many researchers to develop traditional computational methods to quantify the correlation and predict the chronological age, the performance bottleneck delayed access to the practical application. Since artificial intelligence technology brought great opportunities in research, we proposed a perceptron model integrating a channel attention mechanism named PerSEClock. The model was trained on 24,516 CpG loci that can utilize the samples from all types of methylation identification platforms and tested on 15 independent datasets against seven methylation-based age prediction methods. PerSEClock demonstrated the ability to assign varying weights to different CpG loci. This feature allows the model to enhance the weight of age-related loci while reducing the weight of irrelevant loci. The method is free to use for academics at www.dnamclock.com/#/original.
Collapse
Affiliation(s)
- Jian Zhao
- School of Computer Science and Technology, Changchun University, Changchun, China
| | - Haixia Li
- School of Computer Science and Technology, Changchun University, Changchun, China
| | - Jing Qu
- School of Computer Science and Technology, Jilin University, Changchun, China
- School of Information Science and Technology, Institute of Computational Biology, Northeast Normal University, Changchun, China
| | - Xizeng Zong
- Clinical Research Centre, Guangzhou First People’s Hospital, School of Medicine, South China University of Technology, Guangzhou, Guangdong, China
- School of Computer Science and Engineering, Changchun University of Technology, Changchun, China
| | - Yuchen Liu
- Department of Medicine, Boston University School of Medicine, Boston, MA, United States
| | - Zhejun Kuang
- School of Computer Science and Technology, Changchun University, Changchun, China
| | - Han Wang
- School of Information Science and Technology, Institute of Computational Biology, Northeast Normal University, Changchun, China
| |
Collapse
|
25
|
Whitman ET, Ryan CP, Abraham WC, Addae A, Corcoran DL, Elliott ML, Hogan S, Ireland D, Keenan R, Knodt AR, Melzer TR, Poulton R, Ramrakha S, Sugden K, Williams BS, Zhou J, Hariri AR, Belsky DW, Moffitt TE, Caspi A. A blood biomarker of the pace of aging is associated with brain structure: replication across three cohorts. Neurobiol Aging 2024; 136:23-33. [PMID: 38301452 PMCID: PMC11017787 DOI: 10.1016/j.neurobiolaging.2024.01.008] [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: 09/06/2023] [Revised: 01/16/2024] [Accepted: 01/19/2024] [Indexed: 02/03/2024]
Abstract
Biological aging is the correlated decline of multi-organ system integrity central to the etiology of many age-related diseases. A novel epigenetic measure of biological aging, DunedinPACE, is associated with cognitive dysfunction, incident dementia, and mortality. Here, we tested for associations between DunedinPACE and structural MRI phenotypes in three datasets spanning midlife to advanced age: the Dunedin Study (age=45 years), the Framingham Heart Study Offspring Cohort (mean age=63 years), and the Alzheimer's Disease Neuroimaging Initiative (mean age=75 years). We also tested four additional epigenetic measures of aging: the Horvath clock, the Hannum clock, PhenoAge, and GrimAge. Across all datasets (total N observations=3380; total N individuals=2322), faster DunedinPACE was associated with lower total brain volume, lower hippocampal volume, greater burden of white matter microlesions, and thinner cortex. Across all measures, DunedinPACE and GrimAge had the strongest and most consistent associations with brain phenotypes. Our findings suggest that single timepoint measures of multi-organ decline such as DunedinPACE could be useful for gauging nervous system health.
Collapse
Affiliation(s)
- Ethan T Whitman
- Department of Psychology and Neuroscience, Duke University, Durham, NC, USA.
| | - Calen P Ryan
- Butler Columbia Aging Center, Columbia University Mailman School of Public Health, New York, USA
| | | | - Angela Addae
- Department of Psychology and Neuroscience, Duke University, Durham, NC, USA
| | - David L Corcoran
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Maxwell L Elliott
- Department of Psychology, Center for Brain Science, Harvard University, Cambridge, MA, USA
| | - Sean Hogan
- Dunedin Multidisciplinary Health and Development Research Unit, Department of Psychology, University of Otago, Dunedin, New Zealand
| | - David Ireland
- Dunedin Multidisciplinary Health and Development Research Unit, Department of Psychology, University of Otago, Dunedin, New Zealand
| | - Ross Keenan
- Brain Research New Zealand-Rangahau Roro Aotearoa, Centre of Research Excellence, Universities of Auckland and Otago, New Zealand; Christchurch Radiology Group, Christchurch, New Zealand
| | - Annchen R Knodt
- Department of Psychology and Neuroscience, Duke University, Durham, NC, USA
| | - Tracy R Melzer
- Brain Research New Zealand-Rangahau Roro Aotearoa, Centre of Research Excellence, Universities of Auckland and Otago, New Zealand; Department of Medicine, University of Otago, Christchurch, New Zealand
| | - Richie Poulton
- Dunedin Multidisciplinary Health and Development Research Unit, Department of Psychology, University of Otago, Dunedin, New Zealand
| | - Sandhya Ramrakha
- Dunedin Multidisciplinary Health and Development Research Unit, Department of Psychology, University of Otago, Dunedin, New Zealand
| | - Karen Sugden
- Department of Psychology and Neuroscience, Duke University, Durham, NC, USA
| | | | - Jiayi Zhou
- Butler Columbia Aging Center, Columbia University Mailman School of Public Health, New York, USA
| | - Ahmad R Hariri
- Department of Psychology and Neuroscience, Duke University, Durham, NC, USA
| | - Daniel W Belsky
- Butler Columbia Aging Center, Columbia University Mailman School of Public Health, New York, USA; Department of Epidemiology, Columbia University Mailman School of Public Health, New York, USA
| | - Terrie E Moffitt
- Department of Psychology and Neuroscience, Duke University, Durham, NC, USA; Center for Genomic and Computational Biology, Duke University, Durham, NC, USA; King's College London, Social, Genetic, and Developmental Psychiatry Centre, Institute of Psychiatry, Psychology, & Neuroscience, London, UK; PROMENTA, Department of Psychology, University of Oslo, Norway; Department of Psychiatry and Behavioral Sciences, Duke University, Durham, NC, USA
| | - Avshalom Caspi
- Department of Psychology and Neuroscience, Duke University, Durham, NC, USA; Center for Genomic and Computational Biology, Duke University, Durham, NC, USA; King's College London, Social, Genetic, and Developmental Psychiatry Centre, Institute of Psychiatry, Psychology, & Neuroscience, London, UK; PROMENTA, Department of Psychology, University of Oslo, Norway; Department of Psychiatry and Behavioral Sciences, Duke University, Durham, NC, USA
| |
Collapse
|
26
|
Goyne CE, Fair AE, Sumowski PE, Graves JS. The Impact of Aging on Multiple Sclerosis. Curr Neurol Neurosci Rep 2024; 24:83-93. [PMID: 38416310 DOI: 10.1007/s11910-024-01333-2] [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] [Accepted: 01/17/2024] [Indexed: 02/29/2024]
Abstract
PURPOSE OF REVIEW Multiple sclerosis (MS) is a chronic, immune-mediated demyelinating disorder of the central nervous system. Age is one of the most important factors in determining MS phenotype. This review provides an overview of how age influences MS clinical characteristics, pathology, and treatment. RECENT FINDINGS New methods for measuring aging have improved our understanding of the aging process in MS. New studies have characterized the molecular and cellular composition of chronic active or smoldering plaques in MS. These lesions are important contributors to disability progression in MS. These studies highlight the important role of immunosenescence and the innate immune system in sustaining chronic inflammation. Given these changes in immune function, several studies have assessed optimal treatment strategies in aging individuals with MS. MS phenotype is intimately linked with chronologic age and immunosenescence. While there are many unanswered questions, there has been much progress in understanding this relationship which may lead to more effective treatments for progressive disease.
Collapse
Affiliation(s)
- Christopher E Goyne
- Department of Neurosciences, University of California San Diego, 9452 Medical Center Drive, Ste 4W-222, La Jolla, San Diego, CA, 92037, USA
| | - Ashley E Fair
- Department of Neurosciences, University of California San Diego, 9452 Medical Center Drive, Ste 4W-222, La Jolla, San Diego, CA, 92037, USA
| | - Paige E Sumowski
- Department of Neurosciences, University of California San Diego, 9452 Medical Center Drive, Ste 4W-222, La Jolla, San Diego, CA, 92037, USA
| | - Jennifer S Graves
- Department of Neurosciences, University of California San Diego, 9452 Medical Center Drive, Ste 4W-222, La Jolla, San Diego, CA, 92037, USA.
| |
Collapse
|
27
|
Dohm-Hansen S, English JA, Lavelle A, Fitzsimons CP, Lucassen PJ, Nolan YM. The 'middle-aging' brain. Trends Neurosci 2024; 47:259-272. [PMID: 38508906 DOI: 10.1016/j.tins.2024.02.001] [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: 10/16/2023] [Revised: 01/09/2024] [Accepted: 02/05/2024] [Indexed: 03/22/2024]
Abstract
Middle age has historically been an understudied period of life compared to older age, when cognitive and brain health decline are most pronounced, but the scope for intervention may be limited. However, recent research suggests that middle age could mark a shift in brain aging. We review emerging evidence on multiple levels of analysis indicating that midlife is a period defined by unique central and peripheral processes that shape future cognitive trajectories and brain health. Informed by recent developments in aging research and lifespan studies in humans and animal models, we highlight the utility of modeling non-linear changes in study samples with wide subject age ranges to distinguish life stage-specific processes from those acting linearly throughout the lifespan.
Collapse
Affiliation(s)
- Sebastian Dohm-Hansen
- Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland; INFANT Research Centre, University College Cork, Cork, Ireland; APC Microbiome Ireland, University College Cork, Cork, Ireland
| | - Jane A English
- Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland; INFANT Research Centre, University College Cork, Cork, Ireland
| | - Aonghus Lavelle
- Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland; APC Microbiome Ireland, University College Cork, Cork, Ireland
| | - Carlos P Fitzsimons
- Swammerdam Institute for Life Sciences, Brain Plasticity Group, University of Amsterdam, Amsterdam, The Netherlands
| | - Paul J Lucassen
- Swammerdam Institute for Life Sciences, Brain Plasticity Group, University of Amsterdam, Amsterdam, The Netherlands
| | - Yvonne M Nolan
- Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland; APC Microbiome Ireland, University College Cork, Cork, Ireland.
| |
Collapse
|
28
|
Sala C, Di Lena P, Fernandes Durso D, Faria do Valle I, Bacalini MG, Dall’Olio D, Franceschi C, Castellani G, Garagnani P, Nardini C. Where are we in the implementation of tissue-specific epigenetic clocks? FRONTIERS IN BIOINFORMATICS 2024; 4:1306244. [PMID: 38501111 PMCID: PMC10944965 DOI: 10.3389/fbinf.2024.1306244] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2023] [Accepted: 02/14/2024] [Indexed: 03/20/2024] Open
Abstract
Introduction: DNA methylation clocks presents advantageous characteristics with respect to the ambitious goal of identifying very early markers of disease, based on the concept that accelerated ageing is a reliable predictor in this sense. Methods: Such tools, being epigenomic based, are expected to be conditioned by sex and tissue specificities, and this work is about quantifying this dependency as well as that from the regression model and the size of the training set. Results: Our quantitative results indicate that elastic-net penalization is the best performing strategy, and better so when-unsurprisingly-the data set is bigger; sex does not appear to condition clocks performances and tissue specific clocks appear to perform better than generic blood clocks. Finally, when considering all trained clocks, we identified a subset of genes that, to the best of our knowledge, have not been presented yet and might deserve further investigation: CPT1A, MMP15, SHROOM3, SLIT3, and SYNGR. Conclusion: These factual starting points can be useful for the future medical translation of clocks and in particular in the debate between multi-tissue clocks, generally trained on a large majority of blood samples, and tissue-specific clocks.
Collapse
Affiliation(s)
- Claudia Sala
- Department of Medical and Surgical Sciences, University of Bologna, Bologna, Italy
| | - Pietro Di Lena
- Department of Computer Science and Engineering, University of Bologna, Bologna, Italy
| | - Danielle Fernandes Durso
- National Counsel of Technological and Scientific Development (CNPq), Ministry of Science Technology and Innovation (MCTI), Brasília, Brazil
| | | | | | - Daniele Dall’Olio
- IRCCS Istituto delle Scienze Neurologiche di Bologna, Bologna, Italy
| | - Claudio Franceschi
- Institute of Information Technologies, Mathematics and Mechanics, Lobachevsky University, Nizhny Novgorod, Russia
| | - Gastone Castellani
- Department of Medical and Surgical Sciences, University of Bologna, Bologna, Italy
| | - Paolo Garagnani
- Department of Medical and Surgical Sciences, University of Bologna, Bologna, Italy
| | - Christine Nardini
- Istituto per le Applicazioni del Calcolo “Mauro Picone”, Consiglio Nazionale delle Ricerche, Roma, Italy
| |
Collapse
|
29
|
Zhang Z, Reynolds SR, Stolrow HG, Chen J, Christensen BC, Salas LA. Deciphering the role of immune cell composition in epigenetic age acceleration: Insights from cell-type deconvolution applied to human blood epigenetic clocks. Aging Cell 2024; 23:e14071. [PMID: 38146185 PMCID: PMC10928575 DOI: 10.1111/acel.14071] [Citation(s) in RCA: 22] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2023] [Revised: 12/04/2023] [Accepted: 12/05/2023] [Indexed: 12/27/2023] Open
Abstract
Aging is a significant risk factor for various human disorders, and DNA methylation clocks have emerged as powerful tools for estimating biological age and predicting health-related outcomes. Methylation data from blood DNA has been a focus of more recently developed DNA methylation clocks. However, the impact of immune cell composition on epigenetic age acceleration (EAA) remains unclear as only some clocks incorporate partial cell type composition information when analyzing EAA. We investigated associations of 12 immune cell types measured by cell-type deconvolution with EAA predicted by six widely-used DNA methylation clocks in data from >10,000 blood samples. We observed significant associations of immune cell composition with EAA for all six clocks tested. Across the clocks, nine or more of the 12 cell types tested exhibited significant associations with EAA. Higher memory lymphocyte subtype proportions were associated with increased EAA, and naïve lymphocyte subtypes were associated with decreased EAA. To demonstrate the potential confounding of EAA by immune cell composition, we applied EAA in rheumatoid arthritis. Our research maps immune cell type contributions to EAA in human blood and offers opportunities to adjust for immune cell composition in EAA studies to a significantly more granular level. Understanding associations of EAA with immune profiles has implications for the interpretation of epigenetic age and its relevance in aging and disease research. Our detailed map of immune cell type contributions serves as a resource for studies utilizing epigenetic clocks across diverse research fields, including aging-related diseases, precision medicine, and therapeutic interventions.
Collapse
Affiliation(s)
- Ze Zhang
- Department of EpidemiologyGeisel School of Medicine at DartmouthLebanonNew HampshireUSA
- Dartmouth Cancer CenterDartmouth‐Hitchcock Medical CenterLebanonNew HampshireUSA
- Quantitative Biomedical Sciences ProgramGuarini School of Graduate and Advanced StudiesHanoverNew HampshireUSA
| | - Samuel R. Reynolds
- Department of EpidemiologyGeisel School of Medicine at DartmouthLebanonNew HampshireUSA
| | - Hannah G. Stolrow
- Department of EpidemiologyGeisel School of Medicine at DartmouthLebanonNew HampshireUSA
- Dartmouth Cancer CenterDartmouth‐Hitchcock Medical CenterLebanonNew HampshireUSA
| | - Ji‐Qing Chen
- Department of EpidemiologyGeisel School of Medicine at DartmouthLebanonNew HampshireUSA
- Molecular and Cellular Biology ProgramGuarini School of Graduate and Advanced StudiesHanoverNew HampshireUSA
| | - Brock C. Christensen
- Department of EpidemiologyGeisel School of Medicine at DartmouthLebanonNew HampshireUSA
- Dartmouth Cancer CenterDartmouth‐Hitchcock Medical CenterLebanonNew HampshireUSA
- Quantitative Biomedical Sciences ProgramGuarini School of Graduate and Advanced StudiesHanoverNew HampshireUSA
- Molecular and Cellular Biology ProgramGuarini School of Graduate and Advanced StudiesHanoverNew HampshireUSA
| | - Lucas A. Salas
- Department of EpidemiologyGeisel School of Medicine at DartmouthLebanonNew HampshireUSA
- Dartmouth Cancer CenterDartmouth‐Hitchcock Medical CenterLebanonNew HampshireUSA
- Quantitative Biomedical Sciences ProgramGuarini School of Graduate and Advanced StudiesHanoverNew HampshireUSA
- Molecular and Cellular Biology ProgramGuarini School of Graduate and Advanced StudiesHanoverNew HampshireUSA
| |
Collapse
|
30
|
Wang Y, Grant OA, Zhai X, Mcdonald-Maier KD, Schalkwyk LC. Insights into ageing rates comparison across tissues from recalibrating cerebellum DNA methylation clock. GeroScience 2024; 46:39-56. [PMID: 37597113 PMCID: PMC10828477 DOI: 10.1007/s11357-023-00871-w] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Accepted: 07/07/2023] [Indexed: 08/21/2023] Open
Abstract
DNA methylation (DNAm)-based age clocks have been studied extensively as a biomarker of human ageing and a risk factor for age-related diseases. Despite different tissues having vastly different rates of proliferation, it is still largely unknown whether they age at different rates. It was previously reported that the cerebellum ages slowly; however, this claim was drawn from a single clock using a relatively small sample size and so warrants further investigation. We collected the largest cerebellum DNAm dataset (N = 752) to date. We found the respective epigenetic ages are all severely underestimated by six representative DNAm age clocks, with the underestimation effects more pronounced in the four clocks whose training datasets do not include brain-related tissues. We identified 613 age-associated CpGs in the cerebellum, which accounts for only 14.5% of the number found in the middle temporal gyrus from the same population (N = 404). From the 613 cerebellum age-associated CpGs, we built a highly accurate age prediction model for the cerebellum named CerebellumClockspecific (Pearson correlation=0.941, MAD=3.18 years). Ageing rate comparisons based on the two tissue-specific clocks constructed on the 201 overlapping age-associated CpGs support the cerebellum has younger DNAm age. Nevertheless, we built BrainCortexClock to prove a single DNAm clock is able to unbiasedly estimate DNAm ages of both cerebellum and cerebral cortex, when they are adequately and equally represented in the training dataset. Comparing ageing rates across tissues using DNA methylation multi-tissue clocks is flawed. The large underestimation of age prediction for cerebellums by previous clocks mainly reflects the improper usage of these age clocks. There exist strong and consistent ageing effects on the cerebellar methylome, and we suggest the smaller number of age-associated CpG sites in cerebellum is largely attributed to its extremely low average cell replication rates.
Collapse
Affiliation(s)
- Yucheng Wang
- School of Computer Science and Electronic Engineering, University of Essex, Colchester, CO4 3SQ, UK
- School of Life Sciences, University of Essex, Colchester, CO4 3SQ, UK
| | - Olivia A Grant
- School of Life Sciences, University of Essex, Colchester, CO4 3SQ, UK
- Institute of Social and Economic Research, University of Essex, Colchester, CO4 3SQ, UK
- Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, E1 2AT, UK
| | - Xiaojun Zhai
- School of Computer Science and Electronic Engineering, University of Essex, Colchester, CO4 3SQ, UK.
| | - Klaus D Mcdonald-Maier
- School of Computer Science and Electronic Engineering, University of Essex, Colchester, CO4 3SQ, UK
| | | |
Collapse
|
31
|
Zhang J, Sun X, Jia X, Sun B, Xu S, Zhang W, Liu Z. Integrative multi-omics analysis reveals the critical role of the PBXIP1 gene in Alzheimer's disease. Aging Cell 2024; 23:e14044. [PMID: 37984333 PMCID: PMC10861197 DOI: 10.1111/acel.14044] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2023] [Revised: 10/29/2023] [Accepted: 11/01/2023] [Indexed: 11/22/2023] Open
Abstract
Alzheimer's disease (AD) is a neurodegenerative disorder, and its strongest risk factor is aging. A few studies have explored the relationship between aging and AD, while the underlying mechanism remains unclear. We assembled data across multi-omics (i.e., epigenetics, transcriptomics, and proteomics, based on frozen tissues from the dorsolateral prefrontal cortex) and neuropathological and clinical traits from the Religious Orders Study and Rush Memory and Aging Project (ROSMAP). Aging was assessed using six DNA methylation clocks (including the Horvath clock, Hannum clock, Levine clock, HorvathSkin clock, Lin clock, and Cortical clock) that capture mortality risk in literature. After accounting for age, we first identified a gene module (including 263 genes) that was related to the integrated aging measure of six clocks, as well as three neuropathological traits of AD (i.e., β-amyloid, Tau tangles, and tangle density). Interestingly, among 20 key genes with top intramodular connectivity of the module, PBXIP1 was the only one that was significantly associated with all three neuropathological traits of AD at the protein level after Bonferroni correction. Furthermore, PBXIP1 was associated with the clinical diagnosis of AD in both ROSMAP and three independent datasets. Moreover, PBXIP1 may be related to AD through its role in astrocytes and hippocampal neurons, and the mTOR pathway. The results suggest the critical role of PBXIP1 in AD and support the potential and feasibility of using multi-omics data to investigate mechanisms of complex diseases. However, more validations in different populations and experiments in vitro and in vivo are required in the future.
Collapse
Affiliation(s)
- Jingyun Zhang
- Center for Clinical Big Data and Analytics of the Second Affiliated Hospital, and Department of Big Data in Health Science School of Public Health, the Key Laboratory of Intelligent Preventive Medicine of Zhejiang ProvinceZhejiang University School of MedicineHangzhouZhejiangChina
| | - Xiaoyi Sun
- Center for Clinical Big Data and Analytics of the Second Affiliated Hospital, and Department of Big Data in Health Science School of Public Health, the Key Laboratory of Intelligent Preventive Medicine of Zhejiang ProvinceZhejiang University School of MedicineHangzhouZhejiangChina
| | - 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, the Key Laboratory of Intelligent Preventive Medicine of Zhejiang ProvinceZhejiang University School of MedicineHangzhouZhejiangChina
| | - Binggui Sun
- Department of Neurobiology, School of Basic Medical Sciences, Key Laboratory of Medical Neurobiology (Ministry of Health of China), Key Laboratory of Neurobiology of Zhejiang ProvinceZhejiang University School of MedicineHangzhouZhejiangChina
| | - Shijun Xu
- Institute of Material Medica Integration and Transformation for Brain Disorders, and School of PharmacyChengdu University of Traditional Chinese MedicineChengduSichuanChina
| | - Weiping Zhang
- Department of Pharmacology, Institute of Neuroscience, Key Laboratory of Medical Neurobiology of the Ministry of Health of China, Zhejiang Province Key Laboratory of Mental Disorder's ManagementZhejiang University School of MedicineHangzhouZhejiangChina
| | - 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, the Key Laboratory of Intelligent Preventive Medicine of Zhejiang ProvinceZhejiang University School of MedicineHangzhouZhejiangChina
| |
Collapse
|
32
|
Liu Y, Gao Q, Wei K, Huang C, Wang C, Yu Y, Qin G, Wang T. High-dimensional generalized median adaptive lasso with application to omics data. Brief Bioinform 2024; 25:bbae059. [PMID: 38436558 PMCID: PMC10939310 DOI: 10.1093/bib/bbae059] [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: 09/03/2023] [Revised: 01/03/2024] [Indexed: 03/05/2024] Open
Abstract
Recently, there has been a growing interest in variable selection for causal inference within the context of high-dimensional data. However, when the outcome exhibits a skewed distribution, ensuring the accuracy of variable selection and causal effect estimation might be challenging. Here, we introduce the generalized median adaptive lasso (GMAL) for covariate selection to achieve an accurate estimation of causal effect even when the outcome follows skewed distributions. A distinctive feature of our proposed method is that we utilize a linear median regression model for constructing penalty weights, thereby maintaining the accuracy of variable selection and causal effect estimation even when the outcome presents extremely skewed distributions. Simulation results showed that our proposed method performs comparably to existing methods in variable selection when the outcome follows a symmetric distribution. Besides, the proposed method exhibited obvious superiority over the existing methods when the outcome follows a skewed distribution. Meanwhile, our proposed method consistently outperformed the existing methods in causal estimation, as indicated by smaller root-mean-square error. We also utilized the GMAL method on a deoxyribonucleic acid methylation dataset from the Alzheimer's disease (AD) neuroimaging initiative database to investigate the association between cerebrospinal fluid tau protein levels and the severity of AD.
Collapse
Affiliation(s)
- Yahang Liu
- Department of Biostatistics, School of Public Health, Fudan University, Shanghai, China
| | - Qian Gao
- Department of Health Statistics, School of Public Health, Shanxi Medical University, Taiyuan, China
- Key Laboratory of Coal Environmental Pathogenicity and Prevention (Shanxi Medical University), Ministry of Education, China
| | - Kecheng Wei
- Department of Biostatistics, School of Public Health, Fudan University, Shanghai, China
| | - Chen Huang
- Department of Biostatistics, School of Public Health, Fudan University, Shanghai, China
| | - Ce Wang
- Department of Biostatistics, School of Public Health, Fudan University, Shanghai, China
| | - Yongfu Yu
- Department of Biostatistics, School of Public Health, Fudan University, Shanghai, China
- Shanghai Institute of Infectious Disease and Biosecurity, Shanghai, China
- Key Laboratory of Public Health Safety of Ministry of Education, Key Laboratory for Health Technology Assessment, National Commission of Health, Fudan University, Shanghai, China
| | - Guoyou Qin
- Department of Biostatistics, School of Public Health, Fudan University, Shanghai, China
- Shanghai Institute of Infectious Disease and Biosecurity, Shanghai, China
- Key Laboratory of Public Health Safety of Ministry of Education, Key Laboratory for Health Technology Assessment, National Commission of Health, Fudan University, Shanghai, China
| | - Tong Wang
- Department of Health Statistics, School of Public Health, Shanxi Medical University, Taiyuan, China
- Key Laboratory of Coal Environmental Pathogenicity and Prevention (Shanxi Medical University), Ministry of Education, China
| |
Collapse
|
33
|
Zhao Y, Ai W, Zheng J, Hu X, Zhang L. A bibliometric and visual analysis of epigenetic research publications for Alzheimer's disease (2013-2023). Front Aging Neurosci 2024; 16:1332845. [PMID: 38292341 PMCID: PMC10824959 DOI: 10.3389/fnagi.2024.1332845] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Accepted: 01/02/2024] [Indexed: 02/01/2024] Open
Abstract
Background Currently, the prevalence of Alzheimer's disease (AD) is progressively rising, particularly in developed nations. There is an escalating focus on the onset and progression of AD. A mounting body of research indicates that epigenetics significantly contributes to AD and holds substantial promise as a novel therapeutic target for its treatment. Objective The objective of this article is to present the AD areas of research interest, comprehend the contextual framework of the subject research, and investigate the prospective direction for future research development. Methods ln Web of Science Core Collection (WOSCC), we searched documents by specific subject terms and their corresponding free words. VOSviewer, CiteSpace and Scimago Graphica were used to perform statistical analysis on measurement metrics such as the number of published papers, national cooperative networks, publishing countries, institutions, authors, co-cited journals, keywords, and visualize networks of related content elements. Results We selected 1,530 articles from WOSCC from January 2013 to June 2023 about epigenetics of AD. Based on visual analysis, we could get that China and United States were the countries with the most research in this field. Bennett DA was the most contributed and prestigious scientist. The top 3 cited journals were Journal of Alzheimer's Disease, Neurobiology of Aging and Molecular Neurobiology. According to the analysis of keywords and the frequency of citations, ncRNAs, transcription factor, genome, histone modification, blood DNA methylation, acetylation, biomarkers were hot research directions in AD today. Conclusion According to bibliometric analysis, epigenetic research in AD was a promising research direction, and epigenetics had the potential to be used as AD biomarkers and therapeutic targets.
Collapse
Affiliation(s)
- YaPing Zhao
- School of Clinical Medicine, Chengdu Medical College, Chengdu, China
| | - WenJing Ai
- School of Clinical Medicine, Chengdu Medical College, Chengdu, China
| | - JingFeng Zheng
- School of Clinical Medicine, Chengdu Medical College, Chengdu, China
| | - XianLiang Hu
- Chengdu Eighth People’s Hospital, Geriatric Hospital of Chengdu Medical College, Chengdu, China
| | - LuShun Zhang
- Sichuan Key Laboratory of Development and Regeneration, Department of Neurobiology, Chengdu Medical College, Chengdu, China
- Department of Pathology and Pathophysiology, Chengdu Medical College, Chengdu, China
| |
Collapse
|
34
|
Marriott H, Kabiljo R, Hunt GP, Khleifat AA, Jones A, Troakes C, Pfaff AL, Quinn JP, Koks S, Dobson RJ, Schwab P, Al-Chalabi A, Iacoangeli A. Unsupervised machine learning identifies distinct ALS molecular subtypes in post-mortem motor cortex and blood expression data. Acta Neuropathol Commun 2023; 11:208. [PMID: 38129934 PMCID: PMC10734072 DOI: 10.1186/s40478-023-01686-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2023] [Accepted: 11/10/2023] [Indexed: 12/23/2023] Open
Abstract
Amyotrophic lateral sclerosis (ALS) displays considerable clinical and genetic heterogeneity. Machine learning approaches have previously been utilised for patient stratification in ALS as they can disentangle complex disease landscapes. However, lack of independent validation in different populations and tissue samples have greatly limited their use in clinical and research settings. We overcame these issues by performing hierarchical clustering on the 5000 most variably expressed autosomal genes from motor cortex expression data of people with sporadic ALS from the KCL BrainBank (N = 112). Three molecular phenotypes linked to ALS pathogenesis were identified: synaptic and neuropeptide signalling, oxidative stress and apoptosis, and neuroinflammation. Cluster validation was achieved by applying linear discriminant analysis models to cases from TargetALS US motor cortex (N = 93), as well as Italian (N = 15) and Dutch (N = 397) blood expression datasets, for which there was a high assignment probability (80-90%) for each molecular subtype. The ALS and motor cortex specificity of the expression signatures were tested by mapping KCL BrainBank controls (N = 59), and occipital cortex (N = 45) and cerebellum (N = 123) samples from TargetALS to each cluster, before constructing case-control and motor cortex-region logistic regression classifiers. We found that the signatures were not only able to distinguish people with ALS from controls (AUC 0.88 ± 0.10), but also reflect the motor cortex-based disease process, as there was perfect discrimination between motor cortex and the other brain regions. Cell types known to be involved in the biological processes of each molecular phenotype were found in higher proportions, reinforcing their biological interpretation. Phenotype analysis revealed distinct cluster-related outcomes in both motor cortex datasets, relating to disease onset and progression-related measures. Our results support the hypothesis that different mechanisms underpin ALS pathogenesis in subgroups of patients and demonstrate potential for the development of personalised treatment approaches. Our method is available for the scientific and clinical community at https://alsgeclustering.er.kcl.ac.uk .
Collapse
Affiliation(s)
- Heather Marriott
- Department of Basic and Clinical Neuroscience, Maurice Wohl Clinical Neuroscience Institute, Institute of Psychiatry, Psychology and Neuroscience, King?s College London, London, SE5 9NU, UK
- Department of Biostatistics and Health Informatics, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| | - Renata Kabiljo
- Department of Biostatistics and Health Informatics, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| | - Guy P Hunt
- Department of Basic and Clinical Neuroscience, Maurice Wohl Clinical Neuroscience Institute, Institute of Psychiatry, Psychology and Neuroscience, King?s College London, London, SE5 9NU, UK
- Department of Biostatistics and Health Informatics, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
- Perron Institute for Neurological and Translational Science, Nedlands, WA, 6009, Australia
- Centre for Molecular Medicine and Innovative Therapeutics, Murdoch University, Murdoch, WA, 6150, Australia
| | - Ahmad Al Khleifat
- Department of Basic and Clinical Neuroscience, Maurice Wohl Clinical Neuroscience Institute, Institute of Psychiatry, Psychology and Neuroscience, King?s College London, London, SE5 9NU, UK
| | - Ashley Jones
- Department of Basic and Clinical Neuroscience, Maurice Wohl Clinical Neuroscience Institute, Institute of Psychiatry, Psychology and Neuroscience, King?s College London, London, SE5 9NU, UK
| | - Claire Troakes
- Department of Basic and Clinical Neuroscience, Maurice Wohl Clinical Neuroscience Institute, Institute of Psychiatry, Psychology and Neuroscience, King?s College London, London, SE5 9NU, UK
- MRC London Neurodegenerative Diseases Brain Bank, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| | - Abigail L Pfaff
- Perron Institute for Neurological and Translational Science, Nedlands, WA, 6009, Australia
- Centre for Molecular Medicine and Innovative Therapeutics, Murdoch University, Murdoch, WA, 6150, Australia
| | - John P Quinn
- Department of Pharmacology and Therapeutics, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool, L69 3BX, UK
| | - Sulev Koks
- Perron Institute for Neurological and Translational Science, Nedlands, WA, 6009, Australia
- Centre for Molecular Medicine and Innovative Therapeutics, Murdoch University, Murdoch, WA, 6150, Australia
| | - Richard J Dobson
- Department of Biostatistics and Health Informatics, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
- NIHR Maudsley Biomedical Research Centre (BRC), South London and Maudsley NHS Foundation Trust and King's College London, London, UK
- Institute of Health Informatics, University College London, London, UK
- NIHR Biomedical Research Centre, University College London Hospitals NHS Foundation Trust, London, UK
| | - Patrick Schwab
- GlaxoSmithKline, Artificial Intelligence and Machine Learning, Durham, NC, USA
| | - Ammar Al-Chalabi
- Department of Basic and Clinical Neuroscience, Maurice Wohl Clinical Neuroscience Institute, Institute of Psychiatry, Psychology and Neuroscience, King?s College London, London, SE5 9NU, UK
- King's College Hospital, London, SE5 9RS, UK
| | - Alfredo Iacoangeli
- Department of Basic and Clinical Neuroscience, Maurice Wohl Clinical Neuroscience Institute, Institute of Psychiatry, Psychology and Neuroscience, King?s College London, London, SE5 9NU, UK.
- Department of Biostatistics and Health Informatics, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK.
- NIHR Maudsley Biomedical Research Centre (BRC), South London and Maudsley NHS Foundation Trust and King's College London, London, UK.
| |
Collapse
|
35
|
Yang T, Xiao Y, Cheng Y, Huang J, Wei Q, Li C, Shang H. Epigenetic clocks in neurodegenerative diseases: a systematic review. J Neurol Neurosurg Psychiatry 2023; 94:1064-1070. [PMID: 36963821 DOI: 10.1136/jnnp-2022-330931] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/11/2022] [Accepted: 03/03/2023] [Indexed: 03/26/2023]
Abstract
BACKGROUND Biological ageing is one of the principal risk factors for neurodegenerative diseases. It is becoming increasingly clear that acceleration of DNA methylation age, as measured by the epigenetic clock, is closely associated with many age-related diseases. METHODS We searched the PubMed and Web of Science databases to identify eligible studies reporting epigenetic clocks in several neurodegenerative diseases, including Alzheimer's disease (AD), Parkinson's disease (PD), amyotrophic lateral sclerosis (ALS) and Huntington's disease (HD). RESULTS Twenty-three studies (12 for AD, 4 for PD, 5 for ALS, and 2 for HD) were included. We systematically summarised the clinical utility of 11 epigenetic clocks (based on blood and brain tissues) in assessing the risk factors, age of onset, diagnosis, progression, prognosis and pathology of AD, PD, ALS and HD. We also critically described our current understandings to these evidences, and further discussed key challenges, potential mechanisms and future perspectives of epigenetic ageing in neurodegenerative diseases. CONCLUSIONS Epigenetic clocks hold great potential in neurodegenerative diseases. Further research is encouraged to evaluate the clinical utility and promote the application. PROSPERO REGISTRATION NUMBER CRD42022365233.
Collapse
Affiliation(s)
- Tianmi Yang
- Department of Neurology, Sichuan University, Chengdu, Sichuan, China
| | - Yi Xiao
- Department of Neurology, Sichuan University, Chengdu, Sichuan, China
| | - Yangfan Cheng
- Department of Neurology, Sichuan University, Chengdu, Sichuan, China
| | - Jingxuan Huang
- Department of Neurology, Sichuan University, Chengdu, Sichuan, China
| | - Qianqian Wei
- Department of Neurology, Sichuan University, Chengdu, Sichuan, China
| | - Chunyu Li
- Department of Neurology, Sichuan University, Chengdu, Sichuan, China
| | - Huifang Shang
- Department of Neurology, Sichuan University, Chengdu, Sichuan, China
| |
Collapse
|
36
|
Marx GA, Kauffman J, McKenzie AT, Koenigsberg DG, McMillan CT, Morgello S, Karlovich E, Insausti R, Richardson TE, Walker JM, White CL, Babrowicz BM, Shen L, McKee AC, Stein TD, Farrell K, Crary JF. Histopathologic brain age estimation via multiple instance learning. Acta Neuropathol 2023; 146:785-802. [PMID: 37815677 PMCID: PMC10627911 DOI: 10.1007/s00401-023-02636-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Revised: 09/14/2023] [Accepted: 09/18/2023] [Indexed: 10/11/2023]
Abstract
Understanding age acceleration, the discordance between biological and chronological age, in the brain can reveal mechanistic insights into normal physiology as well as elucidate pathological determinants of age-related functional decline and identify early disease changes in the context of Alzheimer's and other disorders. Histopathological whole slide images provide a wealth of pathologic data on the cellular level that can be leveraged to build deep learning models to assess age acceleration. Here, we used a collection of digitized human post-mortem hippocampal sections to develop a histological brain age estimation model. Our model predicted brain age within a mean absolute error of 5.45 ± 0.22 years, with attention weights corresponding to neuroanatomical regions vulnerable to age-related changes. We found that histopathologic brain age acceleration had significant associations with clinical and pathologic outcomes that were not found with epigenetic based measures. Our results indicate that histopathologic brain age is a powerful, independent metric for understanding factors that contribute to brain aging.
Collapse
Affiliation(s)
- Gabriel A Marx
- Department of Pathology, Icahn School of Medicine at Mount Sinai, Friedman Brain Institute, 1 Gustave L. Levy Place, Box 1194, New York, NY, 10029, USA
- Department of Artificial Intelligence and Human Health, Nash Family Department of Neuroscience, Ronald M. Loeb Center for Alzheimer's Disease, Friedman Brain Institute, Neuropathology Brain Bank and Research CoRE, Icahn School of Medicine at Mount Sinai, 1 Gustave L. Levy Place, Box 1194, New York, NY, 10029, USA
| | - Justin Kauffman
- Department of Pathology, Icahn School of Medicine at Mount Sinai, Friedman Brain Institute, 1 Gustave L. Levy Place, Box 1194, New York, NY, 10029, USA
- Department of Artificial Intelligence and Human Health, Nash Family Department of Neuroscience, Ronald M. Loeb Center for Alzheimer's Disease, Friedman Brain Institute, Neuropathology Brain Bank and Research CoRE, Icahn School of Medicine at Mount Sinai, 1 Gustave L. Levy Place, Box 1194, New York, NY, 10029, USA
| | - Andrew T McKenzie
- Department of Pathology, Icahn School of Medicine at Mount Sinai, Friedman Brain Institute, 1 Gustave L. Levy Place, Box 1194, New York, NY, 10029, USA
- Department of Artificial Intelligence and Human Health, Nash Family Department of Neuroscience, Ronald M. Loeb Center for Alzheimer's Disease, Friedman Brain Institute, Neuropathology Brain Bank and Research CoRE, Icahn School of Medicine at Mount Sinai, 1 Gustave L. Levy Place, Box 1194, New York, NY, 10029, USA
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Daniel G Koenigsberg
- Department of Pathology, Icahn School of Medicine at Mount Sinai, Friedman Brain Institute, 1 Gustave L. Levy Place, Box 1194, New York, NY, 10029, USA
- Department of Artificial Intelligence and Human Health, Nash Family Department of Neuroscience, Ronald M. Loeb Center for Alzheimer's Disease, Friedman Brain Institute, Neuropathology Brain Bank and Research CoRE, Icahn School of Medicine at Mount Sinai, 1 Gustave L. Levy Place, Box 1194, New York, NY, 10029, USA
| | - Cory T McMillan
- Frontotemporal Degeneration Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Susan Morgello
- Department of Pathology, Icahn School of Medicine at Mount Sinai, Friedman Brain Institute, 1 Gustave L. Levy Place, Box 1194, New York, NY, 10029, USA
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, Friedman Brain Institute, New York, NY, USA
| | - Esma Karlovich
- Department of Pathology, Icahn School of Medicine at Mount Sinai, Friedman Brain Institute, 1 Gustave L. Levy Place, Box 1194, New York, NY, 10029, USA
| | - Ricardo Insausti
- Human Neuroanatomy Laboratory, School of Medicine, University of Castilla-La Mancha, Albacete, Spain
| | - Timothy E Richardson
- Department of Pathology, Icahn School of Medicine at Mount Sinai, Friedman Brain Institute, 1 Gustave L. Levy Place, Box 1194, New York, NY, 10029, USA
| | - Jamie M Walker
- Department of Pathology, Icahn School of Medicine at Mount Sinai, Friedman Brain Institute, 1 Gustave L. Levy Place, Box 1194, New York, NY, 10029, USA
| | - Charles L White
- Department of Pathology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Bergan M Babrowicz
- Department of Pathology, Icahn School of Medicine at Mount Sinai, Friedman Brain Institute, 1 Gustave L. Levy Place, Box 1194, New York, NY, 10029, USA
| | - Li Shen
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, Friedman Brain Institute, New York, NY, USA
| | - Ann C McKee
- Department of Pathology, Alzheimer's Disease and CTE Center, Boston University School of Medicine, Boston, MA, USA
- Department of Veterans Affairs Medical Center, Bedford, MA, USA
- VA Boston Healthcare System, Boston, MA, USA
| | - Thor D Stein
- Department of Pathology, Alzheimer's Disease and CTE Center, Boston University School of Medicine, Boston, MA, USA
- Department of Veterans Affairs Medical Center, Bedford, MA, USA
- VA Boston Healthcare System, Boston, MA, USA
| | - Kurt Farrell
- Department of Pathology, Icahn School of Medicine at Mount Sinai, Friedman Brain Institute, 1 Gustave L. Levy Place, Box 1194, New York, NY, 10029, USA.
- Department of Artificial Intelligence and Human Health, Nash Family Department of Neuroscience, Ronald M. Loeb Center for Alzheimer's Disease, Friedman Brain Institute, Neuropathology Brain Bank and Research CoRE, Icahn School of Medicine at Mount Sinai, 1 Gustave L. Levy Place, Box 1194, New York, NY, 10029, USA.
| | - John F Crary
- Department of Pathology, Icahn School of Medicine at Mount Sinai, Friedman Brain Institute, 1 Gustave L. Levy Place, Box 1194, New York, NY, 10029, USA.
- Department of Artificial Intelligence and Human Health, Nash Family Department of Neuroscience, Ronald M. Loeb Center for Alzheimer's Disease, Friedman Brain Institute, Neuropathology Brain Bank and Research CoRE, Icahn School of Medicine at Mount Sinai, 1 Gustave L. Levy Place, Box 1194, New York, NY, 10029, USA.
| |
Collapse
|
37
|
Shacfe G, Turko R, Syed HH, Masoud I, Tahmaz Y, Samhan LM, Alkattan K, Shafqat A, Yaqinuddin A. A DNA Methylation Perspective on Infertility. Genes (Basel) 2023; 14:2132. [PMID: 38136954 PMCID: PMC10743303 DOI: 10.3390/genes14122132] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2023] [Revised: 11/24/2023] [Accepted: 11/25/2023] [Indexed: 12/24/2023] Open
Abstract
Infertility affects a significant number of couples worldwide and its incidence is increasing. While assisted reproductive technologies (ART) have revolutionized the treatment landscape of infertility, a significant number of couples present with an idiopathic cause for their infertility, hindering effective management. Profiling the genome and transcriptome of infertile men and women has revealed abnormal gene expression. Epigenetic modifications, which comprise dynamic processes that can transduce environmental signals into gene expression changes, may explain these findings. Indeed, aberrant DNA methylation has been widely characterized as a cause of abnormal sperm and oocyte gene expression with potentially deleterious consequences on fertilization and pregnancy outcomes. This review aims to provide a concise overview of male and female infertility through the lens of DNA methylation alterations.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Areez Shafqat
- College of Medicine, Alfaisal University, Riyadh 11533, Saudi Arabia; (G.S.); (R.T.); (H.H.S.); (I.M.); (Y.T.); (L.M.S.); (K.A.); (A.Y.)
| | | |
Collapse
|
38
|
Shafqat A, Khan S, Omer MH, Niaz M, Albalkhi I, AlKattan K, Yaqinuddin A, Tchkonia T, Kirkland JL, Hashmi SK. Cellular senescence in brain aging and cognitive decline. Front Aging Neurosci 2023; 15:1281581. [PMID: 38076538 PMCID: PMC10702235 DOI: 10.3389/fnagi.2023.1281581] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2023] [Accepted: 11/01/2023] [Indexed: 10/16/2024] Open
Abstract
Cellular senescence is a biological aging hallmark that plays a key role in the development of neurodegenerative diseases. Clinical trials are currently underway to evaluate the effectiveness of senotherapies for these diseases. However, the impact of senescence on brain aging and cognitive decline in the absence of neurodegeneration remains uncertain. Moreover, patient populations like cancer survivors, traumatic brain injury survivors, obese individuals, obstructive sleep apnea patients, and chronic kidney disease patients can suffer age-related brain changes like cognitive decline prematurely, suggesting that they may suffer accelerated senescence in the brain. Understanding the role of senescence in neurocognitive deficits linked to these conditions is crucial, especially considering the rapidly evolving field of senotherapeutics. Such treatments could help alleviate early brain aging in these patients, significantly reducing patient morbidity and healthcare costs. This review provides a translational perspective on how cellular senescence plays a role in brain aging and age-related cognitive decline. We also discuss important caveats surrounding mainstream senotherapies like senolytics and senomorphics, and present emerging evidence of hyperbaric oxygen therapy and immune-directed therapies as viable modalities for reducing senescent cell burden.
Collapse
Affiliation(s)
- Areez Shafqat
- College of Medicine, Alfaisal University, Riyadh, Saudi Arabia
| | | | - Mohamed H. Omer
- School of Medicine, Cardiff University, Cardiff, United Kingdom
| | - Mahnoor Niaz
- Medical College, Aga Khan University, Karachi, Pakistan
| | | | - Khaled AlKattan
- College of Medicine, Alfaisal University, Riyadh, Saudi Arabia
| | | | - Tamara Tchkonia
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, MN, United States
| | - James L. Kirkland
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, MN, United States
| | - Shahrukh K. Hashmi
- Department of Internal Medicine, Mayo Clinic, Rochester, MN, United States
- Clinical Affairs, Khalifa University, Abu Dhabi, United Arab Emirates
- Department of Medicine, SSMC, Abu Dhabi, United Arab Emirates
| |
Collapse
|
39
|
Harvanek ZM, Boks MP, Vinkers CH, Higgins-Chen AT. The Cutting Edge of Epigenetic Clocks: In Search of Mechanisms Linking Aging and Mental Health. Biol Psychiatry 2023; 94:694-705. [PMID: 36764569 PMCID: PMC10409884 DOI: 10.1016/j.biopsych.2023.02.001] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/31/2022] [Revised: 01/31/2023] [Accepted: 02/01/2023] [Indexed: 02/11/2023]
Abstract
Individuals with psychiatric disorders are at increased risk of age-related diseases and early mortality. Recent studies demonstrate that this link between mental health and aging is reflected in epigenetic clocks, aging biomarkers based on DNA methylation. The reported relationships between epigenetic clocks and mental health are mostly correlational, and the mechanisms are poorly understood. Here, we review recent progress concerning the molecular and cellular processes underlying epigenetic clocks as well as novel technologies enabling further studies of the causes and consequences of epigenetic aging. We then review the current literature on how epigenetic clocks relate to specific aspects of mental health, such as stress, medications, substance use, health behaviors, and symptom clusters. We propose an integrated framework where mental health and epigenetic aging are each broken down into multiple distinct processes, which are then linked to each other, using stress and schizophrenia as examples. This framework incorporates the heterogeneity and complexity of both mental health conditions and aging, may help reconcile conflicting results, and provides a basis for further hypothesis-driven research in humans and model systems to investigate potentially causal mechanisms linking aging and mental health.
Collapse
Affiliation(s)
- Zachary M Harvanek
- Department of Psychiatry, Yale University School of Medicine, New Haven, Connecticut
| | - Marco P Boks
- Department of Psychiatry, University Medical Center Utrecht Brain Center, University of Utrecht, Utrecht, the Netherlands
| | - Christiaan H Vinkers
- Department of Psychiatry, Amsterdam University Medical Center, location Vrije Universiteit Amsterdam, Amsterdam, the Netherlands; Mood, Anxiety, Psychosis, Sleep & Stress program, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
| | - Albert T Higgins-Chen
- Department of Psychiatry, Yale University School of Medicine, New Haven, Connecticut; Department of Pathology, Yale University School of Medicine, New Haven, Connecticut.
| |
Collapse
|
40
|
Signal B, Pérez Suárez TG, Taberlay PC, Woodhouse A. Cellular specificity is key to deciphering epigenetic changes underlying Alzheimer's disease. Neurobiol Dis 2023; 186:106284. [PMID: 37683959 DOI: 10.1016/j.nbd.2023.106284] [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: 05/01/2023] [Revised: 08/23/2023] [Accepted: 09/05/2023] [Indexed: 09/10/2023] Open
Abstract
Different cell types in the brain play distinct roles in Alzheimer's disease (AD) progression. Late onset AD (LOAD) is a complex disease, with a large genetic component, but many risk loci fall in non-coding genome regions. Epigenetics implicates the non-coding genome with control of gene expression. The epigenome is highly cell-type specific and dynamically responds to the environment. Therefore, epigenetic mechanisms are well placed to explain genetic and environmental factors that are associated with AD. However, given this cellular specificity, purified cell populations or single cells need to be profiled to avoid effect masking. Here we review the current state of cell-type specific genome-wide profiling in LOAD, covering DNA methylation (CpG, CpH, and hydroxymethylation), histone modifications, and chromatin changes. To date, these data reveal that distinct cell types contribute and react differently to AD progression through epigenetic alterations. This review addresses the current gap in prior bulk-tissue derived work by spotlighting cell-specific changes that govern the complex interplay of cells throughout disease progression and are critical in understanding and developing effective treatments for AD.
Collapse
Affiliation(s)
- Brandon Signal
- School of Medicine, College of Health and Medicine, University of Tasmania, Hobart, TAS, Australia.
| | | | - Phillippa C Taberlay
- School of Medicine, College of Health and Medicine, University of Tasmania, Hobart, TAS, Australia
| | - Adele Woodhouse
- Wicking Dementia Research and Education Centre, University of Tasmania, Hobart, TAS, Australia
| |
Collapse
|
41
|
Lynch MT, Taub MA, Farfel JM, Yang J, Abadir P, De Jager PL, Grodstein F, Bennett DA, Mathias RA. Evaluating genomic signatures of aging in brain tissue as it relates to Alzheimer's disease. Sci Rep 2023; 13:14747. [PMID: 37679407 PMCID: PMC10484923 DOI: 10.1038/s41598-023-41400-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Accepted: 08/25/2023] [Indexed: 09/09/2023] Open
Abstract
Telomere length (TL) attrition, epigenetic age acceleration, and mitochondrial DNA copy number (mtDNAcn) decline are established hallmarks of aging. Each has been individually associated with Alzheimer's dementia, cognitive function, and pathologic Alzheimer's disease (AD). Epigenetic age and mtDNAcn have been studied in brain tissue directly but prior work on TL in brain is limited to small sample sizes and most studies have examined leukocyte TL. Importantly, TL, epigenetic age clocks, and mtDNAcn have not been studied jointly in brain tissue from an AD cohort. We examined dorsolateral prefrontal cortex (DLPFC) tissue from N = 367 participants of the Religious Orders Study (ROS) or the Rush Memory and Aging Project (MAP). TL and mtDNAcn were estimated from whole genome sequencing (WGS) data and cortical clock age was computed on 347 CpG sites. We examined dementia, MCI, and level of and change in cognition, pathologic AD, and three quantitative AD traits, as well as measures of other neurodegenerative diseases and cerebrovascular diseases (CVD). We previously showed that mtDNAcn from DLPFC brain tissue was associated with clinical and pathologic features of AD. Here, we show that those associations are independent of TL. We found TL to be associated with β-amyloid levels (beta = - 0.15, p = 0.023), hippocampal sclerosis (OR = 0.56, p = 0.0015) and cerebral atherosclerosis (OR = 1.44, p = 0.0007). We found strong associations between mtDNAcn and clinical measures of AD. The strongest associations with pathologic measures of AD were with cortical clock and there were associations of mtDNAcn with global AD pathology and tau tangles. Of the other pathologic traits, mtDNAcn was associated with hippocampal sclerosis, macroscopic infarctions and CAA and cortical clock was associated with Lewy bodies. Multi-modal age acceleration, accelerated aging on both mtDNAcn and cortical clock, had greater effect size than a single measure alone. These findings highlight for the first time that age acceleration determined on multiple genomic measures, mtDNAcn and cortical clock may have a larger effect on AD/AD related disorders (ADRD) pathogenesis than single measures.
Collapse
Affiliation(s)
- Megan T Lynch
- Department of Medicine, School of Medicine, Johns Hopkins University, Baltimore, MD, USA
| | - Margaret A Taub
- Department of Biostatistics, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Jose M Farfel
- Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, IL, USA
| | - Jingyun Yang
- Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, IL, USA
| | - Peter Abadir
- Department of Medicine, School of Medicine, Johns Hopkins University, Baltimore, MD, USA
| | - Philip L De Jager
- Center for Translational and Computational Neuroimmunology, Department of Neurology, Columbia University Irving Medical Center, New York, NY, USA
| | - Francine Grodstein
- Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, IL, USA
| | - David A Bennett
- Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, IL, USA
| | - Rasika A Mathias
- Department of Medicine, School of Medicine, Johns Hopkins University, Baltimore, MD, USA.
| |
Collapse
|
42
|
Jeremic D, Jiménez-Díaz L, Navarro-López JD. Targeting epigenetics: A novel promise for Alzheimer's disease treatment. Ageing Res Rev 2023; 90:102003. [PMID: 37422087 DOI: 10.1016/j.arr.2023.102003] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Revised: 05/30/2023] [Accepted: 07/03/2023] [Indexed: 07/10/2023]
Abstract
So far, the search for a cure for Alzheimer Disease (AD) has been unsuccessful. The only approved drugs attenuate some symptoms, but do not halt the progress of this disease, which affects 50 million people worldwide and will increase its incidence in the coming decades. Such scenario demands new therapeutic approaches to fight against this devastating dementia. In recent years, multi-omics research and the analysis of differential epigenetic marks in AD subjects have contributed to our understanding of AD; however, the impact of epigenetic research is yet to be seen. This review integrates the most recent data on pathological processes and epigenetic changes relevant for aging and AD, as well as current therapies targeting epigenetic machinery in clinical trials. Evidence shows that epigenetic modifications play a key role in gene expression, which could provide multi-target preventative and therapeutic approaches in AD. Both novel and repurposed drugs are employed in AD clinical trials due to their epigenetic effects, as well as increasing number of natural compounds. Given the reversible nature of epigenetic modifications and the complexity of gene-environment interactions, the combination of epigenetic-based therapies with environmental strategies and drugs with multiple targets might be needed to properly help AD patients.
Collapse
Affiliation(s)
- Danko Jeremic
- University of Castilla-La Mancha, NeuroPhysiology & Behavior Lab, Biomedical Research Center (CRIB), School of Medicine of Ciudad Real, Spain
| | - Lydia Jiménez-Díaz
- University of Castilla-La Mancha, NeuroPhysiology & Behavior Lab, Biomedical Research Center (CRIB), School of Medicine of Ciudad Real, Spain.
| | - Juan D Navarro-López
- University of Castilla-La Mancha, NeuroPhysiology & Behavior Lab, Biomedical Research Center (CRIB), School of Medicine of Ciudad Real, Spain.
| |
Collapse
|
43
|
Harrer P, Mirza-Schreiber N, Mandel V, Roeber S, Stefani A, Naher S, Wagner M, Gieger C, Waldenberger M, Peters A, Högl B, Herms J, Schormair B, Zhao C, Winkelmann J, Oexle K. Epigenetic Association Analyses and Risk Prediction of RLS. Mov Disord 2023; 38:1410-1418. [PMID: 37212434 DOI: 10.1002/mds.29440] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2022] [Revised: 04/12/2023] [Accepted: 04/26/2023] [Indexed: 05/23/2023] Open
Abstract
BACKGROUND As opposed to other neurobehavioral disorders, epigenetic analyses and biomarkers are largely missing in the case of idiopathic restless legs syndrome (RLS). OBJECTIVES Our aims were to develop a biomarker for RLS based on DNA methylation in blood and to examine DNA methylation in brain tissues for dissecting RLS pathophysiology. METHODS Methylation of blood DNA from three independent cohorts (n = 2283) and post-mortem brain DNA from two cohorts (n = 61) was assessed by Infinium EPIC 850 K BeadChip. Epigenome-wide association study (EWAS) results of individual cohorts were combined by random-effect meta-analysis. A three-stage selection procedure (discovery, n = 884; testing, n = 520; validation, n = 879) established an epigenetic risk score including 30 CpG sites. Epigenetic age was assessed by Horvath's multi-tissue clock and Shireby's cortical clock. RESULTS EWAS meta-analysis revealed 149 CpG sites linked to 136 genes (P < 0.05 after Bonferroni correction) in blood and 23 CpG linked to 18 genes in brain (false discovery rate [FDR] < 5%). Gene-set analyses of blood EWAS results suggested enrichments in brain tissue types and in subunits of the kainate-selective glutamate receptor complex. Individual candidate genes of the brain EWAS could be assigned to neurodevelopmental or metabolic traits. The blood epigenetic risk score achieved an area under the curve (AUC) of 0.70 (0.67-0.73) in the validation set, comparable to analogous scores in other neurobehavioral disorders. A significant difference in biological age in blood or brain of RLS patients was not detectable. CONCLUSIONS DNA methylation supports the notion of altered neurodevelopment in RLS. Epigenetic risk scores are reliably associated with RLS but require even higher accuracy to be useful as biomarkers. © 2023 The Authors. Movement Disorders published by Wiley Periodicals LLC on behalf of International Parkinson and Movement Disorder Society.
Collapse
Affiliation(s)
- Philip Harrer
- Institute of Neurogenomics, Helmholtz Zentrum München, German Research Centre for Environmental Health, Munich, Germany
- Institute of Human Genetics, School of Medicine, Technical University of Munich, Munich, Germany
| | - Nazanin Mirza-Schreiber
- Institute of Neurogenomics, Helmholtz Zentrum München, German Research Centre for Environmental Health, Munich, Germany
- Neurogenetic Systems Analysis Group, Institute of Neurogenomics, Helmholtz Zentrum München, German Research Center for Environmental Health, Munich, Germany
| | - Vanessa Mandel
- Institute of Neurogenomics, Helmholtz Zentrum München, German Research Centre for Environmental Health, Munich, Germany
- Institute of Human Genetics, School of Medicine, Technical University of Munich, Munich, Germany
- Neurogenetic Systems Analysis Group, Institute of Neurogenomics, Helmholtz Zentrum München, German Research Center for Environmental Health, Munich, Germany
| | - Sigrun Roeber
- Center for Neuropathology and Prion Research, Ludwig-Maximilians-Universität, Munich, Germany
| | - Ambra Stefani
- Department of Neurology, Medical University of Innsbruck, Innsbruck, Austria
| | - Shamsun Naher
- Institute of Neurogenomics, Helmholtz Zentrum München, German Research Centre for Environmental Health, Munich, Germany
- Neurogenetic Systems Analysis Group, Institute of Neurogenomics, Helmholtz Zentrum München, German Research Center for Environmental Health, Munich, Germany
| | - Matias Wagner
- Institute of Neurogenomics, Helmholtz Zentrum München, German Research Centre for Environmental Health, Munich, Germany
- Institute of Human Genetics, School of Medicine, Technical University of Munich, Munich, Germany
| | - Christian Gieger
- Research Unit of Molecular Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
- Institute of Epidemiology, Helmholtz Zentrum München, German Research Centre for Environmental Health, Munich, Germany
| | - Melanie Waldenberger
- Research Unit of Molecular Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
- Institute of Epidemiology, Helmholtz Zentrum München, German Research Centre for Environmental Health, Munich, Germany
| | - Annette Peters
- Institute of Epidemiology, Helmholtz Zentrum München, German Research Centre for Environmental Health, Munich, Germany
- German Center for Diabetes Research (DZD), Neuherberg, Germany
- Chair of Epidemiology, Institute for Medical Information Processing, Biometry and Epidemiology, Medical Faculty, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Birgit Högl
- Department of Neurology, Medical University of Innsbruck, Innsbruck, Austria
| | - Jochen Herms
- Center for Neuropathology and Prion Research, Ludwig-Maximilians-Universität, Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Barbara Schormair
- Institute of Neurogenomics, Helmholtz Zentrum München, German Research Centre for Environmental Health, Munich, Germany
- Institute of Human Genetics, School of Medicine, Technical University of Munich, Munich, Germany
| | - Chen Zhao
- Institute of Neurogenomics, Helmholtz Zentrum München, German Research Centre for Environmental Health, Munich, Germany
- Neurogenetic Systems Analysis Group, Institute of Neurogenomics, Helmholtz Zentrum München, German Research Center for Environmental Health, Munich, Germany
| | - Juliane Winkelmann
- Institute of Neurogenomics, Helmholtz Zentrum München, German Research Centre for Environmental Health, Munich, Germany
- Institute of Human Genetics, School of Medicine, Technical University of Munich, Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
- Chair of Neurogenetics, School of Medicine, Technical University of Munich, Munich, Germany
| | - Konrad Oexle
- Institute of Neurogenomics, Helmholtz Zentrum München, German Research Centre for Environmental Health, Munich, Germany
- Institute of Human Genetics, School of Medicine, Technical University of Munich, Munich, Germany
- Neurogenetic Systems Analysis Group, Institute of Neurogenomics, Helmholtz Zentrum München, German Research Center for Environmental Health, Munich, Germany
| |
Collapse
|
44
|
Murthy M, Rizzu P, Heutink P, Mill J, Lashley T, Bettencourt C. Epigenetic Age Acceleration in Frontotemporal Lobar Degeneration: A Comprehensive Analysis in the Blood and Brain. Cells 2023; 12:1922. [PMID: 37508584 PMCID: PMC10378390 DOI: 10.3390/cells12141922] [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: 05/18/2023] [Revised: 06/22/2023] [Accepted: 07/17/2023] [Indexed: 07/30/2023] Open
Abstract
Frontotemporal lobar degeneration (FTLD) includes a heterogeneous group of disorders pathologically characterized by the degeneration of the frontal and temporal lobes. In addition to major genetic contributors of FTLD such as mutations in MAPT, GRN, and C9orf72, recent work has identified several epigenetic modifications including significant differential DNA methylation in DLX1, and OTUD4 loci. As aging remains one of the major risk factors for FTLD, we investigated the presence of accelerated epigenetic aging in FTLD compared to controls. We calculated epigenetic age in both peripheral blood and brain tissues of multiple FTLD subtypes using several DNA methylation clocks, i.e., DNAmClockMulti, DNAmClockHannum, DNAmClockCortical, GrimAge, and PhenoAge, and determined age acceleration and its association with different cellular proportions and clinical traits. Significant epigenetic age acceleration was observed in the peripheral blood of both frontotemporal dementia (FTD) and progressive supranuclear palsy (PSP) patients compared to controls with DNAmClockHannum, even after accounting for confounding factors. A similar trend was observed with both DNAmClockMulti and DNAmClockCortical in post-mortem frontal cortex tissue of PSP patients and in FTLD cases harboring GRN mutations. Our findings support that increased epigenetic age acceleration in the peripheral blood could be an indicator for PSP and to a smaller extent, FTD.
Collapse
Affiliation(s)
- Megha Murthy
- Queen Square Brain Bank for Neurological Disorders, UCL Queen Square Institute of Neurology, London WC1N 1PJ, UK (T.L.)
- Department of Clinical and Movement Neurosciences, UCL Queen Square Institute of Neurology, London WC1N 1PJ, UK
| | - Patrizia Rizzu
- German Center for Neurodegenerative Diseases (DZNE), 72076 Tübingen, Germany
| | - Peter Heutink
- German Center for Neurodegenerative Diseases (DZNE), 72076 Tübingen, Germany
- Alector, Inc., South San Francisco, CA 94080, USA
| | - Jonathan Mill
- Department of Clinical and Biomedical Sciences, Faculty of Health and Life Sciences, University of Exeter, Exeter EX4 5DW, UK
| | - Tammaryn Lashley
- Queen Square Brain Bank for Neurological Disorders, UCL Queen Square Institute of Neurology, London WC1N 1PJ, UK (T.L.)
- Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, London WC1N 1PJ, UK
| | - Conceição Bettencourt
- Queen Square Brain Bank for Neurological Disorders, UCL Queen Square Institute of Neurology, London WC1N 1PJ, UK (T.L.)
- Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, London WC1N 1PJ, UK
| |
Collapse
|
45
|
Abstract
The process of aging manifests from a highly interconnected network of biological cascades resulting in the degradation and breakdown of every living organism over time. This natural development increases risk for numerous diseases and can be debilitating. Academic and industrial investigators have long sought to impede, or potentially reverse, aging in the hopes of alleviating clinical burden, restoring functionality, and promoting longevity. Despite widespread investigation, identifying impactful therapeutics has been hindered by narrow experimental validation and the lack of rigorous study design. In this review, we explore the current understanding of the biological mechanisms of aging and how this understanding both informs and limits interpreting data from experimental models based on these mechanisms. We also discuss select therapeutic strategies that have yielded promising data in these model systems with potential clinical translation. Lastly, we propose a unifying approach needed to rigorously vet current and future therapeutics and guide evaluation toward efficacious therapies.
Collapse
Affiliation(s)
- Robert S Rosen
- Department of Biomedical Engineering, Rutgers, The State University of New Jersey, Piscataway, New Jersey, USA;
| | - Martin L Yarmush
- Department of Biomedical Engineering, Rutgers, The State University of New Jersey, Piscataway, New Jersey, USA;
| |
Collapse
|
46
|
Milicic L, Porter T, Vacher M, Laws SM. Utility of DNA Methylation as a Biomarker in Aging and Alzheimer's Disease. J Alzheimers Dis Rep 2023; 7:475-503. [PMID: 37313495 PMCID: PMC10259073 DOI: 10.3233/adr-220109] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Accepted: 04/23/2023] [Indexed: 06/15/2023] Open
Abstract
Epigenetic mechanisms such as DNA methylation have been implicated in a number of diseases including cancer, heart disease, autoimmune disorders, and neurodegenerative diseases. While it is recognized that DNA methylation is tissue-specific, a limitation for many studies is the ability to sample the tissue of interest, which is why there is a need for a proxy tissue such as blood, that is reflective of the methylation state of the target tissue. In the last decade, DNA methylation has been utilized in the design of epigenetic clocks, which aim to predict an individual's biological age based on an algorithmically defined set of CpGs. A number of studies have found associations between disease and/or disease risk with increased biological age, adding weight to the theory of increased biological age being linked with disease processes. Hence, this review takes a closer look at the utility of DNA methylation as a biomarker in aging and disease, with a particular focus on Alzheimer's disease.
Collapse
Affiliation(s)
- Lidija Milicic
- Centre for Precision Health, Edith Cowan University, Joondalup, Western Australia, Australia
- Collaborative Genomics and Translation Group, Edith Cowan University, Joondalup, Western Australia, Australia
- School of Medical and Health Sciences, Edith Cowan University, Joondalup, Western Australia, Australia
| | - Tenielle Porter
- Centre for Precision Health, Edith Cowan University, Joondalup, Western Australia, Australia
- Collaborative Genomics and Translation Group, Edith Cowan University, Joondalup, Western Australia, Australia
- School of Medical and Health Sciences, Edith Cowan University, Joondalup, Western Australia, Australia
- Curtin Medical School, Curtin University, Bentley, Western Australia, Australia
| | - Michael Vacher
- Centre for Precision Health, Edith Cowan University, Joondalup, Western Australia, Australia
- CSIRO Health and Biosecurity, Australian e-Health Research Centre, Floreat, Western Australia
| | - Simon M. Laws
- Centre for Precision Health, Edith Cowan University, Joondalup, Western Australia, Australia
- Collaborative Genomics and Translation Group, Edith Cowan University, Joondalup, Western Australia, Australia
- School of Medical and Health Sciences, Edith Cowan University, Joondalup, Western Australia, Australia
- Curtin Medical School, Curtin University, Bentley, Western Australia, Australia
| |
Collapse
|
47
|
Sommerer Y, Dobricic V, Schilling M, Ohlei O, Sabet SS, Wesse T, Fuß J, Franzenburg S, Franke A, Parkkinen L, Lill CM, Bertram L. Entorhinal cortex epigenome-wide association study highlights four novel loci showing differential methylation in Alzheimer's disease. Alzheimers Res Ther 2023; 15:92. [PMID: 37149695 PMCID: PMC10163801 DOI: 10.1186/s13195-023-01232-7] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Accepted: 04/15/2023] [Indexed: 05/08/2023]
Abstract
BACKGROUND Studies on DNA methylation (DNAm) in Alzheimer's disease (AD) have recently highlighted several genomic loci showing association with disease onset and progression. METHODS Here, we conducted an epigenome-wide association study (EWAS) using DNAm profiles in entorhinal cortex (EC) from 149 AD patients and control brains and combined these with two previously published EC datasets by meta-analysis (total n = 337). RESULTS We identified 12 cytosine-phosphate-guanine (CpG) sites showing epigenome-wide significant association with either case-control status or Braak's tau-staging. Four of these CpGs, located in proximity to CNFN/LIPE, TENT5A, PALD1/PRF1, and DIRAS1, represent novel findings. Integrating DNAm levels with RNA sequencing-based mRNA expression data generated in the same individuals showed significant DNAm-mRNA correlations for 6 of the 12 significant CpGs. Lastly, by calculating rates of epigenetic age acceleration using two recently proposed "epigenetic clock" estimators we found a significant association with accelerated epigenetic aging in the brains of AD patients vs. controls. CONCLUSION In summary, our study represents the hitherto most comprehensive EWAS in AD using EC and highlights several novel differentially methylated loci with potential effects on gene expression.
Collapse
Affiliation(s)
- Yasmine Sommerer
- Lübeck Interdisciplinary Platform for Genome Analytics (LIGA), University of Lübeck, Ratzeburger Allee 160, Haus V50, 1St Floor, Room 319, 23562, Lübeck, Germany
| | - Valerija Dobricic
- Lübeck Interdisciplinary Platform for Genome Analytics (LIGA), University of Lübeck, Ratzeburger Allee 160, Haus V50, 1St Floor, Room 319, 23562, Lübeck, Germany
| | - Marcel Schilling
- Lübeck Interdisciplinary Platform for Genome Analytics (LIGA), University of Lübeck, Ratzeburger Allee 160, Haus V50, 1St Floor, Room 319, 23562, Lübeck, Germany
| | - Olena Ohlei
- Lübeck Interdisciplinary Platform for Genome Analytics (LIGA), University of Lübeck, Ratzeburger Allee 160, Haus V50, 1St Floor, Room 319, 23562, Lübeck, Germany
| | - Sanaz Sedghpour Sabet
- Institute of Clinical Molecular Biology, Christian-Albrechts-University of Kiel, Kiel, Germany
| | - Tanja Wesse
- Institute of Clinical Molecular Biology, Christian-Albrechts-University of Kiel, Kiel, Germany
| | - Janina Fuß
- Institute of Clinical Molecular Biology, Christian-Albrechts-University of Kiel, Kiel, Germany
| | - Sören Franzenburg
- Institute of Clinical Molecular Biology, Christian-Albrechts-University of Kiel, Kiel, Germany
| | - Andre Franke
- Institute of Clinical Molecular Biology, Christian-Albrechts-University of Kiel, Kiel, Germany
| | - Laura Parkkinen
- Nuffield Department of Clinical Neurosciences, Oxford Parkinson's Disease Centre, University of Oxford, Oxford, UK
| | - Christina M Lill
- Lübeck Interdisciplinary Platform for Genome Analytics (LIGA), University of Lübeck, Ratzeburger Allee 160, Haus V50, 1St Floor, Room 319, 23562, Lübeck, Germany
- Ageing Epidemiology Unit (AGE), School of Public Health, Imperial College London, London, UK
- Institute of Epidemiology and Social Medicine, University of Münster, Münster, Germany
| | - Lars Bertram
- Lübeck Interdisciplinary Platform for Genome Analytics (LIGA), University of Lübeck, Ratzeburger Allee 160, Haus V50, 1St Floor, Room 319, 23562, Lübeck, Germany.
- Center for Lifespan Changes in Brain and Cognition (LCBC), Department of Psychology, University of Oslo, Oslo, Norway.
| |
Collapse
|
48
|
Bao H, Cao J, Chen M, Chen M, Chen W, Chen X, Chen Y, Chen Y, Chen Y, Chen Z, Chhetri JK, Ding Y, Feng J, Guo J, Guo M, He C, Jia Y, Jiang H, Jing Y, Li D, Li J, Li J, Liang Q, Liang R, Liu F, Liu X, Liu Z, Luo OJ, Lv J, Ma J, Mao K, Nie J, Qiao X, Sun X, Tang X, Wang J, Wang Q, Wang S, Wang X, Wang Y, Wang Y, Wu R, Xia K, Xiao FH, Xu L, Xu Y, Yan H, Yang L, Yang R, Yang Y, Ying Y, Zhang L, Zhang W, Zhang W, Zhang X, Zhang Z, Zhou M, Zhou R, Zhu Q, Zhu Z, Cao F, Cao Z, Chan P, Chen C, Chen G, Chen HZ, Chen J, Ci W, Ding BS, Ding Q, Gao F, Han JDJ, Huang K, Ju Z, Kong QP, Li J, Li J, Li X, Liu B, Liu F, Liu L, Liu Q, Liu Q, Liu X, Liu Y, Luo X, Ma S, Ma X, Mao Z, Nie J, Peng Y, Qu J, Ren J, Ren R, Song M, Songyang Z, Sun YE, Sun Y, Tian M, Wang S, et alBao H, Cao J, Chen M, Chen M, Chen W, Chen X, Chen Y, Chen Y, Chen Y, Chen Z, Chhetri JK, Ding Y, Feng J, Guo J, Guo M, He C, Jia Y, Jiang H, Jing Y, Li D, Li J, Li J, Liang Q, Liang R, Liu F, Liu X, Liu Z, Luo OJ, Lv J, Ma J, Mao K, Nie J, Qiao X, Sun X, Tang X, Wang J, Wang Q, Wang S, Wang X, Wang Y, Wang Y, Wu R, Xia K, Xiao FH, Xu L, Xu Y, Yan H, Yang L, Yang R, Yang Y, Ying Y, Zhang L, Zhang W, Zhang W, Zhang X, Zhang Z, Zhou M, Zhou R, Zhu Q, Zhu Z, Cao F, Cao Z, Chan P, Chen C, Chen G, Chen HZ, Chen J, Ci W, Ding BS, Ding Q, Gao F, Han JDJ, Huang K, Ju Z, Kong QP, Li J, Li J, Li X, Liu B, Liu F, Liu L, Liu Q, Liu Q, Liu X, Liu Y, Luo X, Ma S, Ma X, Mao Z, Nie J, Peng Y, Qu J, Ren J, Ren R, Song M, Songyang Z, Sun YE, Sun Y, Tian M, Wang S, Wang S, Wang X, Wang X, Wang YJ, Wang Y, Wong CCL, Xiang AP, Xiao Y, Xie Z, Xu D, Ye J, Yue R, Zhang C, Zhang H, Zhang L, Zhang W, Zhang Y, Zhang YW, Zhang Z, Zhao T, Zhao Y, Zhu D, Zou W, Pei G, Liu GH. Biomarkers of aging. SCIENCE CHINA. LIFE SCIENCES 2023; 66:893-1066. [PMID: 37076725 PMCID: PMC10115486 DOI: 10.1007/s11427-023-2305-0] [Show More Authors] [Citation(s) in RCA: 163] [Impact Index Per Article: 81.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 01/19/2023] [Accepted: 02/27/2023] [Indexed: 04/21/2023]
Abstract
Aging biomarkers are a combination of biological parameters to (i) assess age-related changes, (ii) track the physiological aging process, and (iii) predict the transition into a pathological status. Although a broad spectrum of aging biomarkers has been developed, their potential uses and limitations remain poorly characterized. An immediate goal of biomarkers is to help us answer the following three fundamental questions in aging research: How old are we? Why do we get old? And how can we age slower? This review aims to address this need. Here, we summarize our current knowledge of biomarkers developed for cellular, organ, and organismal levels of aging, comprising six pillars: physiological characteristics, medical imaging, histological features, cellular alterations, molecular changes, and secretory factors. To fulfill all these requisites, we propose that aging biomarkers should qualify for being specific, systemic, and clinically relevant.
Collapse
Affiliation(s)
- Hainan Bao
- 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
| | - Jiani Cao
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China
| | - Mengting Chen
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, 410008, China
- Hunan Key Laboratory of Aging Biology, Xiangya Hospital, Central South University, Changsha, 410008, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Min Chen
- Clinic Center of Human Gene Research, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Clinical Research Center of Metabolic and Cardiovascular Disease, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Key Laboratory of Metabolic Abnormalities and Vascular Aging, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Wei Chen
- Stem Cell Translational Research Center, Tongji Hospital, Tongji University School of Medicine, Shanghai, 200065, China
| | - Xiao Chen
- Department of Nuclear Medicine, Daping Hospital, Third Military Medical University, Chongqing, 400042, China
| | - Yanhao Chen
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Yu Chen
- Shanghai Key Laboratory of Maternal Fetal Medicine, Clinical and Translational Research Center of Shanghai First Maternity and Infant Hospital, Frontier Science Center for Stem Cell Research, Shanghai Key Laboratory of Signaling and Disease Research, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China
| | - Yutian Chen
- The Department of Endovascular Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Zhiyang Chen
- Key Laboratory of Regenerative Medicine of Ministry of Education, Institute of Ageing and Regenerative Medicine, Jinan University, Guangzhou, 510632, China
| | - Jagadish K Chhetri
- National Clinical Research Center for Geriatric Diseases, Xuanwu Hospital, Capital Medical University, Beijing, 100053, China
| | - Yingjie Ding
- 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
| | - Junlin Feng
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Jun Guo
- The Key Laboratory of Geriatrics, Beijing Institute of Geriatrics, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing Hospital/National Center of Gerontology of National Health Commission, Beijing, 100730, China
| | - Mengmeng Guo
- School of Pharmaceutical Sciences, Tsinghua University, Beijing, 100084, China
| | - Chuting He
- University of Chinese Academy of Sciences, Beijing, 100049, China
- State Key Laboratory of Membrane Biology, 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
| | - Yujuan Jia
- Department of Neurology, First Affiliated Hospital, Shanxi Medical University, Taiyuan, 030001, China
| | - Haiping Jiang
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China
- University of Chinese Academy of Sciences, Beijing, 100049, 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
| | - Ying Jing
- Beijing Municipal Geriatric Medical Research Center, 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
- Advanced Innovation Center for Human Brain Protection, and National Clinical Research Center for Geriatric Disorders, Xuanwu Hospital Capital Medical University, Beijing, 100053, China
| | - Dingfeng Li
- Department of Neurology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230036, 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
| | - Jingyi Li
- University of Chinese Academy of Sciences, Beijing, 100049, China
- State Key Laboratory of Membrane Biology, 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
| | - Qinhao Liang
- College of Life Sciences, TaiKang Center for Life and Medical Sciences, Wuhan University, Wuhan, 430072, China
| | - Rui Liang
- Research Institute of Transplant Medicine, Organ Transplant Center, NHC Key Laboratory for Critical Care Medicine, Tianjin First Central Hospital, Nankai University, Tianjin, 300384, China
| | - Feng Liu
- MOE Key Laboratory of Gene Function and Regulation, Guangzhou Key Laboratory of Healthy Aging Research, School of Life Sciences, Institute of Healthy Aging Research, Sun Yat-sen University, Guangzhou, 510275, China
| | - Xiaoqian Liu
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China
- University of Chinese Academy of Sciences, Beijing, 100049, 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
| | - Zuojun Liu
- School of Life Sciences, Hainan University, Haikou, 570228, China
| | - Oscar Junhong Luo
- Department of Systems Biomedical Sciences, School of Medicine, Jinan University, Guangzhou, 510632, China
| | - Jianwei Lv
- School of Life Sciences, Xiamen University, Xiamen, 361102, China
| | - Jingyi Ma
- The State Key Laboratory of Organ Failure Research, National Clinical Research Center of Kidney Disease, Division of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Kehang Mao
- Peking-Tsinghua Center for Life Sciences, Academy for Advanced Interdisciplinary Studies, Center for Quantitative Biology (CQB), Peking University, Beijing, 100871, China
| | - Jiawei Nie
- Shanghai Institute of Hematology, State Key Laboratory for Medical Genomics, National Research Center for Translational Medicine (Shanghai), International Center for Aging and Cancer, Collaborative Innovation Center of Hematology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Xinhua Qiao
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China
| | - Xinpei Sun
- Peking University International Cancer Institute, Health Science Center, Peking University, Beijing, 100101, China
| | - Xiaoqiang Tang
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, State Key Laboratory of Biotherapy, West China Second University Hospital, Sichuan University, Chengdu, 610041, China
| | - Jianfang Wang
- Institute for Regenerative Medicine, Shanghai East Hospital, Frontier Science Center for Stem Cell Research, Shanghai Key Laboratory of Signaling and Disease Research, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China
| | - Qiaoran 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
| | - Siyuan Wang
- Clinical Research Institute, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science & Peking Union Medical College, Beijing, 100730, China
| | - Xuan Wang
- Hepatobiliary and Pancreatic Center, Medical Research Center, Beijing Tsinghua Changgung Hospital, Beijing, 102218, China
| | - Yaning Wang
- Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080, China
- Advanced Medical Technology Center, The First Affiliated Hospital, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080, China
| | - Yuhan Wang
- University of Chinese Academy of Sciences, Beijing, 100049, China
- State Key Laboratory of Membrane Biology, 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
| | - Rimo Wu
- Bioland Laboratory (Guangzhou Regenerative Medicine and Health Guangdong Laboratory), Guangzhou, 510005, China
| | - Kai Xia
- Center for Stem Cell Biologyand Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-sen University, Guangzhou, 510080, China
- National-Local Joint Engineering Research Center for Stem Cells and Regenerative Medicine, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080, China
| | - Fu-Hui Xiao
- CAS Center for Excellence in Animal Evolution and Genetics, Chinese Academy of Sciences, Kunming, 650223, China
- State Key Laboratory of Genetic Resources and Evolution, Key Laboratory of Healthy Aging Research of Yunnan Province, Kunming Key Laboratory of Healthy Aging Study, KIZ/CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, 650223, China
| | - Lingyan Xu
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, 200241, China
| | - Yingying Xu
- 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
| | - Haoteng Yan
- Beijing Municipal Geriatric Medical Research Center, 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
- Advanced Innovation Center for Human Brain Protection, and National Clinical Research Center for Geriatric Disorders, Xuanwu Hospital Capital Medical University, Beijing, 100053, China
| | - Liang Yang
- CAS Key Laboratory of Regenerative Biology, Joint School of Life Sciences, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou Medical University, Guangzhou, 510530, China
| | - Ruici Yang
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, 200031, China
| | - Yuanxin Yang
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai, 201210, China
| | - Yilin Ying
- Department of Geriatrics, Medical Center on Aging of Shanghai Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
- International Laboratory in Hematology and Cancer, Shanghai Jiao Tong University School of Medicine/Ruijin Hospital, Shanghai, 200025, China
| | - Le Zhang
- Gerontology Center of Hubei Province, Wuhan, 430000, China
- Institute of Gerontology, Department of Geriatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Weiwei Zhang
- Department of Cardiology, The Second Medical Centre, Chinese PLA General Hospital, National Clinical Research Center for Geriatric Diseases, Beijing, 100853, China
| | - Wenwan Zhang
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Xing Zhang
- Key Laboratory of Ministry of Education, School of Aerospace Medicine, Fourth Military Medical University, Xi'an, 710032, China
| | - Zhuo Zhang
- Optogenetics & Synthetic Biology Interdisciplinary Research Center, State Key Laboratory of Bioreactor Engineering, Shanghai Frontiers Science Center of Optogenetic Techniques for Cell Metabolism, School of Pharmacy, East China University of Science and Technology, Shanghai, 200237, China
- Research Unit of New Techniques for Live-cell Metabolic Imaging, Chinese Academy of Medical Sciences, Beijing, 100730, China
| | - Min Zhou
- Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital of Central South University, Changsha, 410008, China
| | - Rui Zhou
- Department of Nuclear Medicine and PET Center, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, 310009, China
| | - Qingchen Zhu
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Zhengmao Zhu
- Department of Genetics and Cell Biology, College of Life Science, Nankai University, Tianjin, 300071, China
- Haihe Laboratory of Cell Ecosystem, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300020, China
| | - Feng Cao
- Department of Cardiology, The Second Medical Centre, Chinese PLA General Hospital, National Clinical Research Center for Geriatric Diseases, Beijing, 100853, China.
| | - Zhongwei Cao
- State Key Laboratory of Biotherapy, West China Second University Hospital, Sichuan University, Chengdu, 610041, China.
| | - Piu Chan
- National Clinical Research Center for Geriatric Diseases, Xuanwu Hospital, Capital Medical University, Beijing, 100053, China.
| | - Chang Chen
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China.
| | - Guobing Chen
- Department of Microbiology and Immunology, School of Medicine, Jinan University, Guangzhou, 510632, China.
- Guangdong-Hong Kong-Macau Great Bay Area Geroscience Joint Laboratory, Guangzhou, 510000, China.
| | - Hou-Zao Chen
- Department of Biochemistryand Molecular Biology, State Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100005, China.
| | - Jun Chen
- Peking University Research Center on Aging, Beijing Key Laboratory of Protein Posttranslational Modifications and Cell Function, Department of Biochemistry and Molecular Biology, Department of Integration of Chinese and Western Medicine, School of Basic Medical Science, Peking University, Beijing, 100191, China.
| | - Weimin Ci
- 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.
| | - Bi-Sen Ding
- State Key Laboratory of Biotherapy, West China Second University Hospital, Sichuan University, Chengdu, 610041, China.
| | - Qiurong Ding
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, 200031, China.
| | - Feng Gao
- Key Laboratory of Ministry of Education, School of Aerospace Medicine, Fourth Military Medical University, Xi'an, 710032, China.
| | - Jing-Dong J Han
- Peking-Tsinghua Center for Life Sciences, Academy for Advanced Interdisciplinary Studies, Center for Quantitative Biology (CQB), Peking University, Beijing, 100871, China.
| | - Kai Huang
- Clinic Center of Human Gene Research, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
- Hubei Clinical Research Center of Metabolic and Cardiovascular Disease, Huazhong University of Science and Technology, Wuhan, 430022, China.
- Hubei Key Laboratory of Metabolic Abnormalities and Vascular Aging, Huazhong University of Science and Technology, Wuhan, 430022, China.
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
| | - Zhenyu Ju
- Key Laboratory of Regenerative Medicine of Ministry of Education, Institute of Ageing and Regenerative Medicine, Jinan University, Guangzhou, 510632, China.
| | - Qing-Peng Kong
- CAS Center for Excellence in Animal Evolution and Genetics, Chinese Academy of Sciences, Kunming, 650223, China.
- State Key Laboratory of Genetic Resources and Evolution, Key Laboratory of Healthy Aging Research of Yunnan Province, Kunming Key Laboratory of Healthy Aging Study, KIZ/CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, 650223, China.
| | - Ji Li
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, 410008, China.
- Hunan Key Laboratory of Aging Biology, Xiangya Hospital, Central South University, Changsha, 410008, China.
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, China.
| | - Jian Li
- The Key Laboratory of Geriatrics, Beijing Institute of Geriatrics, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing Hospital/National Center of Gerontology of National Health Commission, Beijing, 100730, China.
| | - Xin Li
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China.
- University of Chinese Academy of Sciences, Beijing, 100049, 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.
| | - Baohua Liu
- School of Basic Medical Sciences, Shenzhen University Medical School, Shenzhen, 518060, China.
| | - Feng Liu
- Metabolic Syndrome Research Center, The Second Xiangya Hospital, Central South Unversity, Changsha, 410011, China.
| | - Lin Liu
- Department of Genetics and Cell Biology, College of Life Science, Nankai University, Tianjin, 300071, China.
- Haihe Laboratory of Cell Ecosystem, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300020, China.
- Institute of Translational Medicine, Tianjin Union Medical Center, Nankai University, Tianjin, 300000, China.
- State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin, 300350, China.
| | - Qiang Liu
- Department of Neurology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230036, China.
| | - Qiang Liu
- Department of Neurology, Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin, 300052, China.
- Tianjin Institute of Immunology, Tianjin Medical University, Tianjin, 300070, China.
| | - Xingguo Liu
- CAS Key Laboratory of Regenerative Biology, Joint School of Life Sciences, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou Medical University, Guangzhou, 510530, China.
| | - Yong Liu
- College of Life Sciences, TaiKang Center for Life and Medical Sciences, Wuhan University, Wuhan, 430072, China.
| | - Xianghang Luo
- Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital of Central South University, Changsha, 410008, China.
| | - Shuai Ma
- University of Chinese Academy of Sciences, Beijing, 100049, China.
- State Key Laboratory of Membrane Biology, 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.
| | - Xinran Ma
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, 200241, China.
| | - Zhiyong Mao
- Shanghai Key Laboratory of Maternal Fetal Medicine, Clinical and Translational Research Center of Shanghai First Maternity and Infant Hospital, Frontier Science Center for Stem Cell Research, Shanghai Key Laboratory of Signaling and Disease Research, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China.
| | - Jing Nie
- The State Key Laboratory of Organ Failure Research, National Clinical Research Center of Kidney Disease, Division of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China.
| | - Yaojin Peng
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China.
- University of Chinese Academy of Sciences, Beijing, 100049, China.
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, 100101, China.
| | - Jing Qu
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China.
- University of Chinese Academy of Sciences, Beijing, 100049, 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.
| | - Jie Ren
- 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.
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, 100101, China.
| | - Ruibao Ren
- Shanghai Institute of Hematology, State Key Laboratory for Medical Genomics, National Research Center for Translational Medicine (Shanghai), International Center for Aging and Cancer, Collaborative Innovation Center of Hematology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
- International Center for Aging and Cancer, Hainan Medical University, Haikou, 571199, China.
| | - Moshi Song
- University of Chinese Academy of Sciences, Beijing, 100049, China.
- State Key Laboratory of Membrane Biology, 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.
| | - Zhou Songyang
- MOE Key Laboratory of Gene Function and Regulation, Guangzhou Key Laboratory of Healthy Aging Research, School of Life Sciences, Institute of Healthy Aging Research, Sun Yat-sen University, Guangzhou, 510275, China.
- Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, China.
| | - Yi Eve Sun
- Stem Cell Translational Research Center, Tongji Hospital, Tongji University School of Medicine, Shanghai, 200065, China.
| | - Yu Sun
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Chinese Academy of Sciences, Shanghai, 200031, China.
- Department of Medicine and VAPSHCS, University of Washington, Seattle, WA, 98195, USA.
| | - Mei Tian
- Human Phenome Institute, Fudan University, Shanghai, 201203, China.
| | - Shusen Wang
- Research Institute of Transplant Medicine, Organ Transplant Center, NHC Key Laboratory for Critical Care Medicine, Tianjin First Central Hospital, Nankai University, Tianjin, 300384, China.
| | - Si Wang
- Beijing Municipal Geriatric Medical Research Center, 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.
- Advanced Innovation Center for Human Brain Protection, and National Clinical Research Center for Geriatric Disorders, Xuanwu Hospital Capital Medical University, Beijing, 100053, China.
| | - Xia Wang
- School of Pharmaceutical Sciences, Tsinghua University, Beijing, 100084, China.
| | - Xiaoning Wang
- Institute of Geriatrics, The second Medical Center, Beijing Key Laboratory of Aging and Geriatrics, National Clinical Research Center for Geriatric Diseases, Chinese PLA General Hospital, Beijing, 100853, China.
| | - Yan-Jiang Wang
- Department of Neurology and Center for Clinical Neuroscience, Daping Hospital, Third Military Medical University, Chongqing, 400042, China.
| | - Yunfang Wang
- Hepatobiliary and Pancreatic Center, Medical Research Center, Beijing Tsinghua Changgung Hospital, Beijing, 102218, China.
| | - Catherine C L Wong
- Clinical Research Institute, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science & Peking Union Medical College, Beijing, 100730, China.
| | - Andy Peng Xiang
- Center for Stem Cell Biologyand Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-sen University, Guangzhou, 510080, China.
- National-Local Joint Engineering Research Center for Stem Cells and Regenerative Medicine, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080, China.
| | - Yichuan Xiao
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Chinese Academy of Sciences, Shanghai, 200031, China.
| | - Zhengwei Xie
- Peking University International Cancer Institute, Health Science Center, Peking University, Beijing, 100101, China.
- Beijing & Qingdao Langu Pharmaceutical R&D Platform, Beijing Gigaceuticals Tech. Co. Ltd., Beijing, 100101, China.
| | - Daichao Xu
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai, 201210, China.
| | - Jing Ye
- Department of Geriatrics, Medical Center on Aging of Shanghai Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
- International Laboratory in Hematology and Cancer, Shanghai Jiao Tong University School of Medicine/Ruijin Hospital, Shanghai, 200025, China.
| | - Rui Yue
- Institute for Regenerative Medicine, Shanghai East Hospital, Frontier Science Center for Stem Cell Research, Shanghai Key Laboratory of Signaling and Disease Research, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China.
| | - Cuntai Zhang
- Gerontology Center of Hubei Province, Wuhan, 430000, China.
- Institute of Gerontology, Department of Geriatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
| | - Hongbo Zhang
- Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080, China.
- Advanced Medical Technology Center, The First Affiliated Hospital, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080, China.
| | - Liang Zhang
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Chinese Academy of Sciences, Shanghai, 200031, China.
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, 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.
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, 100101, China.
| | - Yong Zhang
- Bioland Laboratory (Guangzhou Regenerative Medicine and Health Guangdong Laboratory), Guangzhou, 510005, China.
- The State Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and School of Basic Medicine, Peking Union Medical College, Beijing, 100005, China.
| | - Yun-Wu Zhang
- Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, Institute of Neuroscience, School of Medicine, Xiamen University, Xiamen, 361102, China.
| | - Zhuohua Zhang
- Key Laboratory of Molecular Precision Medicine of Hunan Province and Center for Medical Genetics, Institute of Molecular Precision Medicine, Xiangya Hospital, Central South University, Changsha, 410078, China.
- Department of Neurosciences, Hengyang Medical School, University of South China, Hengyang, 421001, China.
| | - Tongbiao Zhao
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China.
- University of Chinese Academy of Sciences, Beijing, 100049, 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.
| | - Yuzheng Zhao
- Optogenetics & Synthetic Biology Interdisciplinary Research Center, State Key Laboratory of Bioreactor Engineering, Shanghai Frontiers Science Center of Optogenetic Techniques for Cell Metabolism, School of Pharmacy, East China University of Science and Technology, Shanghai, 200237, China.
- Research Unit of New Techniques for Live-cell Metabolic Imaging, Chinese Academy of Medical Sciences, Beijing, 100730, China.
| | - Dahai Zhu
- Bioland Laboratory (Guangzhou Regenerative Medicine and Health Guangdong Laboratory), Guangzhou, 510005, China.
- The State Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and School of Basic Medicine, Peking Union Medical College, Beijing, 100005, China.
| | - Weiguo Zou
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, 200031, China.
| | - Gang Pei
- Shanghai Key Laboratory of Signaling and Disease Research, Laboratory of Receptor-Based Biomedicine, The Collaborative Innovation Center for Brain Science, School of Life Sciences and Technology, Tongji University, Shanghai, 200070, China.
| | - Guang-Hui Liu
- University of Chinese Academy of Sciences, Beijing, 100049, China.
- State Key Laboratory of Membrane Biology, 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.
- Advanced Innovation Center for Human Brain Protection, and National Clinical Research Center for Geriatric Disorders, Xuanwu Hospital Capital Medical University, Beijing, 100053, China.
| |
Collapse
|
49
|
Martínez-Magaña JJ, Krystal JH, Girgenti MJ, Núnez-Ríos DL, Nagamatsu ST, Andrade-Brito DE, Traumatic Stress Brain Research Group, Montalvo-Ortiz JL. Decoding the role of transcriptomic clocks in the human prefrontal cortex. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2023:2023.04.19.23288765. [PMID: 37163025 PMCID: PMC10168432 DOI: 10.1101/2023.04.19.23288765] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/11/2023]
Abstract
Aging is a complex process with interindividual variability, which can be measured by aging biological clocks. Aging clocks are machine-learning algorithms guided by biological information and associated with mortality risk and a wide range of health outcomes. One of these aging clocks are transcriptomic clocks, which uses gene expression data to predict biological age; however, their functional role is unknown. Here, we profiled two transcriptomic clocks (RNAAgeCalc and knowledge-based deep neural network clock) in a large dataset of human postmortem prefrontal cortex (PFC) samples. We identified that deep-learning transcriptomic clock outperforms RNAAgeCalc to predict transcriptomic age in the human PFC. We identified associations of transcriptomic clocks with psychiatric-related traits. Further, we applied system biology algorithms to identify common gene networks among both clocks and performed pathways enrichment analyses to assess its functionality and prioritize genes involved in the aging processes. Identified gene networks showed enrichment for diseases of signal transduction by growth factor receptors and second messenger pathways. We also observed enrichment of genome-wide signals of mental and physical health outcomes and identified genes previously associated with human brain aging. Our findings suggest a link between transcriptomic aging and health disorders, including psychiatric traits. Further, it reveals functional genes within the human PFC that may play an important role in aging and health risk.
Collapse
Affiliation(s)
- José J. Martínez-Magaña
- Division of Human Genetics, Department of Psychiatry, Yale University School of Medicine, New Haven
- National Center for PTSD, US Department of Veterans Affairs, West Haven, CT, USA
| | - John H. Krystal
- Division of Human Genetics, Department of Psychiatry, Yale University School of Medicine, New Haven
- National Center for PTSD, US Department of Veterans Affairs, West Haven, CT, USA
- Psychiatry Service, VA Connecticut Health Care System, West Haven, CT, USA
| | - Matthew J. Girgenti
- Division of Human Genetics, Department of Psychiatry, Yale University School of Medicine, New Haven
- National Center for PTSD, US Department of Veterans Affairs, West Haven, CT, USA
| | - Diana L. Núnez-Ríos
- Division of Human Genetics, Department of Psychiatry, Yale University School of Medicine, New Haven
- National Center for PTSD, US Department of Veterans Affairs, West Haven, CT, USA
| | - Sheila T. Nagamatsu
- Division of Human Genetics, Department of Psychiatry, Yale University School of Medicine, New Haven
- National Center for PTSD, US Department of Veterans Affairs, West Haven, CT, USA
| | - Diego E. Andrade-Brito
- Division of Human Genetics, Department of Psychiatry, Yale University School of Medicine, New Haven
- National Center for PTSD, US Department of Veterans Affairs, West Haven, CT, USA
| | | | - Janitza L. Montalvo-Ortiz
- Division of Human Genetics, Department of Psychiatry, Yale University School of Medicine, New Haven
- National Center for PTSD, US Department of Veterans Affairs, West Haven, CT, USA
- Psychiatry Service, VA Connecticut Health Care System, West Haven, CT, USA
| |
Collapse
|
50
|
Simons RL, Ong ML, Lei MK, Beach SRH, Zhang Y, Philibert R, Mielke MM. Changes in Loneliness, BDNF, and Biological Aging Predict Trajectories in a Blood-Based Epigenetic Measure of Cortical Aging: A Study of Older Black Americans. Genes (Basel) 2023; 14:842. [PMID: 37107599 PMCID: PMC10138024 DOI: 10.3390/genes14040842] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2023] [Revised: 03/17/2023] [Accepted: 03/29/2023] [Indexed: 04/03/2023] Open
Abstract
A recent epigenetic measure of aging has developed based on human cortex tissue. This cortical clock (CC) dramatically outperformed extant blood-based epigenetic clocks in predicting brain age and neurological degeneration. Unfortunately, measures that require brain tissue are of limited utility to investigators striving to identify everyday risk factors for dementia. The present study investigated the utility of using the CpG sites included in the CC to formulate a peripheral blood-based cortical measure of brain age (CC-Bd). To establish the utility of CC-Bd, we used growth curves with individually varying time points and longitudinal data from a sample of 694 aging African Americans. We examined whether three risk factors that have been linked to cognitive decline-loneliness, depression, and BDNFm-predicted CC-Bd after controlling for several factors, including three new-generation epigenetic clocks. Our findings showed that two clocks-DunedinPACE and PoAm-predicted CC-BD, but that increases in loneliness and BDNFm continued to be robust predictors of accelerated CC-Bd even after taking these effects into account. This suggests that CC-Bd is assessing something more than the pan-tissue epigenetic clocks but that, at least in part, brain health is also associated with the general aging of the organism.
Collapse
Affiliation(s)
- Ronald L. Simons
- Department of Sociology, University of Georgia, Athens, GA 30602, USA
| | - Mei Ling Ong
- Center for Family Research, University of Georgia, Athens, GA 30602, USA
| | - Man-Kit Lei
- Department of Sociology, University of Georgia, Athens, GA 30602, USA
| | | | - Yue Zhang
- Department of Sociology, University of Georgia, Athens, GA 30602, USA
| | - Robert Philibert
- Department of Psychiatry, University of Iowa School of Medicine, Iowa City, IA 52242, USA
| | - Michelle M. Mielke
- Department of Epidemiology and Prevention, Wake Forest University School of Medicine, Winston-Salem, NC 27101, USA
| |
Collapse
|