1
|
Scheffler F, Ipser J, Pancholi D, Murphy A, Cao Z, Ottino-González J, Thompson PM, Shoptaw S, Conrod P, Mackey S, Garavan H, Stein DJ. Mega-analysis of the brain-age gap in substance use disorder: An ENIGMA Addiction working group study. Addiction 2024; 119:1937-1946. [PMID: 39165145 DOI: 10.1111/add.16621] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Accepted: 06/19/2024] [Indexed: 08/22/2024]
Abstract
BACKGROUND AND AIMS The brain age gap (BAG), calculated as the difference between a machine learning model-based predicted brain age and chronological age, has been increasingly investigated in psychiatric disorders. Tobacco and alcohol use are associated with increased BAG; however, no studies have compared global and regional BAG across substances other than alcohol and tobacco. This study aimed to compare global and regional estimates of brain age in individuals with substance use disorders and healthy controls. DESIGN This was a cross-sectional study. SETTING This is an Enhancing Neuro Imaging through Meta-Analysis Consortium (ENIGMA) Addiction Working Group study including data from 38 global sites. PARTICIPANTS This study included 2606 participants, of whom 1725 were cases with a substance use disorder and 881 healthy controls. MEASUREMENTS This study used the Kaufmann brain age prediction algorithms to generate global and regional brain age estimates using T1 weighted magnetic resonance imaging (MRI) scans. We used linear mixed effects models to compare global and regional (FreeSurfer lobestrict output) BAG (i.e. predicted minus chronological age) between individuals with one of five primary substance use disorders as well as healthy controls. FINDINGS Alcohol use disorder (β = -5.49, t = -5.51, p < 0.001) was associated with higher global BAG, whereas amphetamine-type stimulant use disorder (β = 3.44, t = 2.42, p = 0.02) was associated with lower global BAG in the separate substance-specific models. CONCLUSIONS People with alcohol use disorder appear to have a higher brain-age gap than people without alcohol use disorder, which is consistent with other evidence of the negative impact of alcohol on the brain.
Collapse
Affiliation(s)
- Freda Scheffler
- Neuroscience Institute, University of Cape Town, Cape Town, South Africa
- Department of Psychiatry and Mental Health, University of Cape Town, Cape Town, South Africa
| | - Jonathan Ipser
- Neuroscience Institute, University of Cape Town, Cape Town, South Africa
- Department of Psychiatry and Mental Health, University of Cape Town, Cape Town, South Africa
| | - Devarshi Pancholi
- Department of Psychiatry, University of Vermont College of Medicine, Burlington, USA
| | - Alistair Murphy
- Department of Psychiatry, University of Vermont College of Medicine, Burlington, USA
| | - Zhipeng Cao
- Department of Psychiatry, University of Vermont College of Medicine, Burlington, USA
| | - Jonatan Ottino-González
- Department of Pediatrics, Division of Endocrinology, Diabetes, and Metabolism, Children's Hospital Los Angeles, Los Angeles, USA
| | - Paul M Thompson
- Imaging Genetics Center, Mark and Mary Stevens Institute for Neuroimaging and Informatics, Department of Neurology, Keck School of Medicine, University of Southern California, Marina del Rey, CA, USA
| | - Steve Shoptaw
- Department of Family Medicine, UCLA, Los Angeles, CA, USA
- University of Cape Town, Cape Town, South Africa
| | - Patricia Conrod
- Department of Psychiatry, Université de Montreal, CHU Ste Justine Hospital, Montreal, Canada
| | - Scott Mackey
- Department of Psychiatry, University of Vermont College of Medicine, Burlington, USA
| | - Hugh Garavan
- Department of Psychiatry, University of Vermont College of Medicine, Burlington, USA
| | - Dan J Stein
- Neuroscience Institute, University of Cape Town, Cape Town, South Africa
- Department of Psychiatry and Mental Health, University of Cape Town, Cape Town, South Africa
- South African Medical Research Council (SAMRC) Unit on Risk and Resilience in Mental Disorders, University of Cape Town, Cape Town, South Africa
| |
Collapse
|
2
|
Antoniades M, Srinivasan D, Wen J, Erus G, Abdulkadir A, Mamourian E, Melhem R, Hwang G, Cui Y, Govindarajan ST, Chen AA, Zhou Z, Yang Z, Chen J, Pomponio R, Sotardi S, An Y, Bilgel M, LaMontagne P, Singh A, Benzinger T, Beason-Held L, Marcus DS, Yaffe K, Launer L, Morris JC, Tosun D, Ferrucci L, Bryan RN, Resnick SM, Habes M, Wolk D, Fan Y, Nasrallah IM, Shou H, Davatzikos C. Relationship between MRI brain-age heterogeneity, cognition, genetics and Alzheimer's disease neuropathology. EBioMedicine 2024; 109:105399. [PMID: 39437659 PMCID: PMC11536027 DOI: 10.1016/j.ebiom.2024.105399] [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: 11/13/2023] [Revised: 09/24/2024] [Accepted: 09/30/2024] [Indexed: 10/25/2024] Open
Abstract
BACKGROUND Brain ageing is highly heterogeneous, as it is driven by a variety of normal and neuropathological processes. These processes may differentially affect structural and functional brain ageing across individuals, with more pronounced ageing (older brain age) during midlife being indicative of later development of dementia. Here, we examined whether brain-ageing heterogeneity in unimpaired older adults related to neurodegeneration, different cognitive trajectories, genetic and amyloid-beta (Aβ) profiles, and to predicted progression to Alzheimer's disease (AD). METHODS Functional and structural brain age measures were obtained for resting-state functional MRI and structural MRI, respectively, in 3460 cognitively normal individuals across an age range spanning 42-85 years. Participants were categorised into four groups based on the difference between their chronological and predicted age in each modality: advanced age in both (n = 291), resilient in both (n = 260) or advanced in one/resilient in the other (n = 163/153). With the resilient group as the reference, brain-age groups were compared across neuroimaging features of neuropathology (white matter hyperintensity volume, neuronal loss measured with Neurite Orientation Dispersion and Density Imaging, AD-specific atrophy patterns measured with the Spatial Patterns of Abnormality for Recognition of Early Alzheimer's Disease index, amyloid burden using amyloid positron emission tomography (PET), progression to mild cognitive impairment and baseline and longitudinal cognitive measures (trail making task, mini mental state examination, digit symbol substitution task). FINDINGS Individuals with advanced structural and functional brain-ages had more features indicative of neurodegeneration and they had poor cognition. Individuals with a resilient brain-age in both modalities had a genetic variant that has been shown to be associated with age of onset of AD. Mixed brain-age was associated with selective cognitive deficits. INTERPRETATION The advanced group displayed evidence of increased atrophy across all neuroimaging features that was not found in either of the mixed groups. This is in line with biomarkers of preclinical AD and cerebrovascular disease. These findings suggest that the variation in structural and functional brain ageing across individuals reflects the degree of underlying neuropathological processes and may indicate the propensity to develop dementia in later life. FUNDING The National Institute on Aging, the National Institutes of Health, the Swiss National Science Foundation, the Kaiser Foundation Research Institute and the National Heart, Lung, and Blood Institute.
Collapse
Affiliation(s)
- Mathilde Antoniades
- AI(2)D, Center for AI and Data Science for Integrated Diagnostics, and Center for Biomedical Image Computing and Analytics, University of Pennsylvania, Philadelphia, PA, USA.
| | - Dhivya Srinivasan
- AI(2)D, Center for AI and Data Science for Integrated Diagnostics, and Center for Biomedical Image Computing and Analytics, University of Pennsylvania, Philadelphia, PA, USA
| | - Junhao Wen
- AI(2)D, Center for AI and Data Science for Integrated Diagnostics, and Center for Biomedical Image Computing and Analytics, University of Pennsylvania, Philadelphia, PA, USA; Laboratory of AI and Biomedical Science (LABS), University of Southern California, Los Angeles, CA, USA
| | - Guray Erus
- AI(2)D, Center for AI and Data Science for Integrated Diagnostics, and Center for Biomedical Image Computing and Analytics, University of Pennsylvania, Philadelphia, PA, USA
| | - Ahmed Abdulkadir
- AI(2)D, Center for AI and Data Science for Integrated Diagnostics, and Center for Biomedical Image Computing and Analytics, University of Pennsylvania, Philadelphia, PA, USA; Department of Clinical Neuroscience, Center for Research in Neuroscience, Lausanne University Hospital, Lausanne, Switzerland
| | - Elizabeth Mamourian
- AI(2)D, Center for AI and Data Science for Integrated Diagnostics, and Center for Biomedical Image Computing and Analytics, University of Pennsylvania, Philadelphia, PA, USA
| | - Randa Melhem
- AI(2)D, Center for AI and Data Science for Integrated Diagnostics, and Center for Biomedical Image Computing and Analytics, University of Pennsylvania, Philadelphia, PA, USA
| | - Gyujoon Hwang
- AI(2)D, Center for AI and Data Science for Integrated Diagnostics, and Center for Biomedical Image Computing and Analytics, University of Pennsylvania, Philadelphia, PA, USA; Department of Psychiatry and Behavioral Medicine, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Yuhan Cui
- AI(2)D, Center for AI and Data Science for Integrated Diagnostics, and Center for Biomedical Image Computing and Analytics, University of Pennsylvania, Philadelphia, PA, USA
| | - Sindhuja Tirumalai Govindarajan
- AI(2)D, Center for AI and Data Science for Integrated Diagnostics, and Center for Biomedical Image Computing and Analytics, University of Pennsylvania, Philadelphia, PA, USA
| | - Andrew A Chen
- Penn Statistics in Imaging and Visualization Center, Department of Biostatistics, Epidemiology, and Informatics, University of Pennsylvania, Philadelphia, PA 19104, United States
| | - Zhen Zhou
- AI(2)D, Center for AI and Data Science for Integrated Diagnostics, and Center for Biomedical Image Computing and Analytics, University of Pennsylvania, Philadelphia, PA, USA
| | - Zhijian Yang
- AI(2)D, Center for AI and Data Science for Integrated Diagnostics, and Center for Biomedical Image Computing and Analytics, University of Pennsylvania, Philadelphia, PA, USA
| | - Jiong Chen
- AI(2)D, Center for AI and Data Science for Integrated Diagnostics, and Center for Biomedical Image Computing and Analytics, University of Pennsylvania, Philadelphia, PA, USA
| | - Raymond Pomponio
- Department of Biostatistics, Colorado School of Public Health, Aurora, CO 80045, USA
| | - Susan Sotardi
- Department of Radiology, Children's Hospital of Philadelphia, Perelman School of Medicine at the University of Pennsylvania, USA
| | - Yang An
- Laboratory of Behavioral Neuroscience, National Institute on Aging, National Institutes of Health, Baltimore, MD, USA
| | - Murat Bilgel
- Laboratory of Behavioral Neuroscience, National Institute on Aging, National Institutes of Health, Baltimore, MD, USA
| | - Pamela LaMontagne
- Department of Radiology, Washington University School of Medicine, St. Louis, MO, USA
| | - Ashish Singh
- Department of Biostatistics, Epidemiology and Informatics, University of Pennsylvania, USA
| | - Tammie Benzinger
- Department of Radiology, Washington University School of Medicine, St. Louis, MO, USA
| | - Lori Beason-Held
- Laboratory of Behavioral Neuroscience, National Institute on Aging, National Institutes of Health, Baltimore, MD, USA
| | - Daniel S Marcus
- Department of Radiology, Washington University School of Medicine, St. Louis, MO, USA
| | | | - Lenore Launer
- Neuroepidemiology Section, Intramural Research Program, National Institute on Aging, Bethesda, MD, USA
| | - John C Morris
- Knight Alzheimer Disease Research Center, Washington University in St. Louis, St. Louis, MO, USA
| | - Duygu Tosun
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, San Francisco, CA, USA
| | - Luigi Ferrucci
- National Institute on Aging, National Institute of Health, Baltimore, MD 21224, USA
| | - R Nick Bryan
- Department of Radiology, University of Pennsylvania, Philadelphia, PA, USA
| | - Susan M Resnick
- Laboratory of Behavioral Neuroscience, National Institute on Aging, National Institutes of Health, Baltimore, MD, USA
| | - Mohamad Habes
- AI(2)D, Center for AI and Data Science for Integrated Diagnostics, and Center for Biomedical Image Computing and Analytics, University of Pennsylvania, Philadelphia, PA, USA; Neuroimage Analytics Laboratory (NAL) and the Biggs Institute Neuroimaging Core (BINC), Glenn Biggs Institute for Alzheimer's and Neurodegenerative Diseases, University of Texas Health Science Center San Antonio, San Antonio, TX, USA
| | - David Wolk
- Department of Neurology, University of Pennsylvania, Philadelphia, PA, USA
| | - Yong Fan
- Department of Radiology, University of Pennsylvania, Philadelphia, PA, USA
| | - Ilya M Nasrallah
- Department of Radiology, University of Pennsylvania, Philadelphia, PA, USA
| | - Haochang Shou
- Department of Biostatistics, Epidemiology and Informatics, University of Pennsylvania, USA
| | - Christos Davatzikos
- AI(2)D, Center for AI and Data Science for Integrated Diagnostics, and Center for Biomedical Image Computing and Analytics, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
3
|
Leech KA, Kettlety SA, Mack WJ, Kreder KJ, Schrepf A, Kutch JJ. Brain predicted age in chronic pelvic pain: a study by the Multidisciplinary Approach to the Study of Chronic Pelvic Pain Research Network. Pain 2024:00006396-990000000-00744. [PMID: 39432808 DOI: 10.1097/j.pain.0000000000003424] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2023] [Accepted: 08/29/2024] [Indexed: 10/23/2024]
Abstract
ABSTRACT The effect of chronic pain on brain-predicted age is unclear. We performed secondary analyses of a large cross-sectional and 3-year longitudinal data set from the Multidisciplinary Approach to the Study of Chronic Pelvic Pain Research Network to test the hypothesis that chronic pelvic pain accelerates brain aging and brain aging rate. Brain-predicted ages of 492 chronic pelvic pain patients and 72 controls were determined from T1-weighted MRI scans and used to calculate the brain-predicted age gap estimation (brainAGE; brain-predicted - chronological age). Separate regression models determined whether the presence of chronic pelvic pain could explain brainAGE and brain aging rate when accounting for covariates. We performed secondary analyses to understand whether brainAGE was associated with factors that subtype chronic pelvic pain patients (inflammation, widespread pain, and psychological comorbidities). We found a significant association between chronic pelvic pain and brainAGE that differed by sex. Women with chronic pelvic pain had higher brainAGE than female controls, whereas men with chronic pelvic pain exhibited lower brainAGE than male controls on average-however, the effect was not statistically significant in men or women when considered independently. Secondary analyses demonstrated preliminary evidence of an association between inflammatory load and brainAGE. Further studies of brainAGE and inflammatory load are warranted.
Collapse
Affiliation(s)
- Kristan A Leech
- Division of Biokinesiology and Physical Therapy, University of Southern California, Los Angeles, CA, United States
| | - Sarah A Kettlety
- Division of Biokinesiology and Physical Therapy, University of Southern California, Los Angeles, CA, United States
| | - Wendy J Mack
- Population and Public Health Sciences, University of Southern California, Los Angeles, CA, United States
| | - Karl J Kreder
- Department of Urology, University of Iowa, Iowa City, IA, United States
| | - Andrew Schrepf
- Departments of Anesthesiology, Obstetrics & Gynecology, University of Michigan, Michigan Medicine, Ann Arbor, MI, United States
| | - Jason J Kutch
- Division of Biokinesiology and Physical Therapy, University of Southern California, Los Angeles, CA, United States
| |
Collapse
|
4
|
Constantinides C, Caramaschi D, Zammit S, Freeman TP, Walton E. Exploring associations between psychotic experiences and structural brain age: a population-based study in late adolescence. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.10.07.24314890. [PMID: 39417107 PMCID: PMC11482991 DOI: 10.1101/2024.10.07.24314890] [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: 10/19/2024]
Abstract
Neuroimaging studies show advanced structural "brain age" in schizophrenia and related psychotic disorders, potentially reflecting aberrant brain ageing or maturation. The extent to which altered brain age is associated with subthreshold psychotic experiences (PE) in youth remains unclear. We investigated the association between PE and brain-predicted age difference (brain-PAD) in late adolescence using a population-based sample of 117 participants with PE and 115 without PE (aged 19-21 years) from the Avon Longitudinal Study of Parents and Children. Brain-PAD was estimated using a publicly available machine learning model previously trained on a combination of region-wise T1-weighted grey-matter measures. We found little evidence for an association between PEs and brain-PAD after adjusting for age and sex (Cohen's d = -0.21 [95% CI -0.47, 0.05], p = 0.11). While there was some evidence for lower brain-PAD in those with PEs relative to those without PEs after additionally adjusting for parental social class (Cohen's d = -0.31 [95% CI -0.58, -0.03], p = 0.031) or birth weight (Cohen's d = -0.29 [95% CI -0.55, -0.03], p = 0.038), adjusting for maternal education or childhood IQ did not alter the primary results. These findings do not support the notion of advanced brain age in older adolescents with PEs. However, they weakly suggest there might be a younger-looking brain in those individuals, indicative of subtle delays in structural brain maturation. Future studies with larger samples covering a wider age range and multimodal measures could further investigate brain age as a marker of psychotic experiences in youth.
Collapse
Affiliation(s)
| | - Doretta Caramaschi
- Department of Psychology, Faculty of Health and Life Sciences, University of Exeter, UK
| | - Stanley Zammit
- MRC Centre for Neuropsychiatric Genetics and Genomics, Division of Psychological Medicine and Clinical Neurosciences, Cardiff University School of Medicine, Cardiff, UK
- Centre for Academic Mental Health, Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
| | - Tom P Freeman
- Addiction and Mental Health Group (AIM), Department of Psychology, University of Bath, UK
| | | |
Collapse
|
5
|
Fang Z, Pan N, Liu S, Li H, Pan M, Zhang J, Li Z, Liu M, Ge X. Comparative analysis of brain age prediction using structural and diffusion MRIs in neonates. Neuroimage 2024; 299:120815. [PMID: 39191358 DOI: 10.1016/j.neuroimage.2024.120815] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Revised: 08/02/2024] [Accepted: 08/24/2024] [Indexed: 08/29/2024] Open
Abstract
Using machine learning techniques to predict brain age from multimodal data has become a crucial biomarker for assessing brain development. Among various types of brain imaging data, structural magnetic resonance imaging (sMRI) and diffusion magnetic resonance imaging (dMRI) are the most commonly used modalities. sMRI focuses on depicting macrostructural features of the brain, while dMRI reveals the orientation of major white matter fibers and changes in tissue microstructure. However, their differential capabilities in reflecting newborn age and clinical implications have not been systematically studied. This study aims to explore the impact of sMRI and dMRI on brain age prediction. Comparing predictions based on T2-weighted(T2w) and fractional anisotropy (FA) images, we found their mean absolute errors (MAE) in predicting infant age to be similar. Exploratory analysis revealed for T2w images, areas such as the cerebral cortex and ventricles contribute most significantly to age prediction, whereas FA images highlight the cerebral cortex and regions of the main white matter tracts. Despite both modalities focusing on the cerebral cortex, they exhibit significant region-wise differences, reflecting developmental disparities in macro- and microstructural aspects of the cortex. Additionally, we examined the effects of prematurity, gender, and hemispherical asymmetry of the brain on age prediction for both modalities. Results showed significant differences (p<0.05) in age prediction biases based on FA images across gender and hemispherical asymmetry, whereas no significant differences were observed with T2w images. This study underscores the differences between T2w and FA images in predicting infant brain age, offering new perspectives for studying infant brain development and aiding more effective assessment and tracking of infant development.
Collapse
Affiliation(s)
- Zhicong Fang
- School of Information Science and Engineering, Shandong Normal University, Shandong, China
| | - Ningning Pan
- School of Information Science and Engineering, Shandong Normal University, Shandong, China
| | - Shujuan Liu
- School of Information Science and Engineering, Shandong Normal University, Shandong, China
| | - Hongzhuang Li
- School of Information Science and Engineering, Shandong Normal University, Shandong, China
| | - Minmin Pan
- School of Information Science and Engineering, Shandong Normal University, Shandong, China
| | - Jiong Zhang
- Laboratory of Advanced Theranostic Materials and Technology, Ningbo Institute of Materials Technology and Engineering, Chinese Academy of Sciences, Ningbo, China
| | - Zhuoshuo Li
- School of Biomedical Engineering, Sun Yat-sen University, Shenzhen, China
| | - Mengting Liu
- School of Biomedical Engineering, Sun Yat-sen University, Shenzhen, China.
| | - Xinting Ge
- School of Information Science and Engineering, Shandong Normal University, Shandong, China.
| |
Collapse
|
6
|
Dove A, Wang J, Huang H, Dunk MM, Sakakibara S, Guitart-Masip M, Papenberg G, Xu W. Diabetes, Prediabetes, and Brain Aging: The Role of Healthy Lifestyle. Diabetes Care 2024; 47:1794-1802. [PMID: 39193914 PMCID: PMC11417282 DOI: 10.2337/dc24-0860] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/25/2024] [Accepted: 07/08/2024] [Indexed: 08/29/2024]
Abstract
OBJECTIVE Diabetes is a well-known risk factor for dementia. We investigated the association between (pre)diabetes and older brain age and whether this can be attenuated by modifiable lifestyle behaviors. RESEARCH DESIGN AND METHODS The study included 31,229 dementia-free adults from the UK Biobank between the ages of 40 and 70 years. Glycemic status (normoglycemia, prediabetes, or diabetes) was ascertained based on medical history, medication use, and HbA1c measured at baseline. Information on cardiometabolic risk factors (obesity, hypertension, low HDL, and high triglycerides) and lifestyle behaviors (smoking, drinking, and physical activity) was also collected at baseline. Participants underwent up to two brain MRI scans over 11 years of follow-up. Brain age was estimated using a machine learning model based on 1,079 brain MRI phenotypes and used to calculate brain age gap (BAG; i.e., brain age minus chronological age). RESULTS At baseline, 13,518 participants (43.3%) had prediabetes and 1,149 (3.7%) had diabetes. Prediabetes (β = 0.22 [95% CI 0.10, 0.34]) and diabetes (2.01 [1.70, 2.32]) were both associated with significantly higher BAG, and diabetes was further associated with significant increase in BAG over time (0.27 [0.01, 0.53]). The association between (pre)diabetes and higher BAG was more pronounced in men and in people with two or more cardiometabolic risk factors. In joint exposure analysis, having a healthy lifestyle (i.e., no smoking, no heavy drinking, and high physical activity) significantly attenuated the diabetes-BAG association. CONCLUSIONS Diabetes and even prediabetes are associated with accelerated brain aging, especially among men and people with poor cardiometabolic health. However, a healthy lifestyle may counteract this.
Collapse
Affiliation(s)
- Abigail Dove
- Aging Research Center, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden
| | - Jiao Wang
- Department of Epidemiology and Biostatistics, School of Public Health, Tianjin Medical University, Tianjin, China
- Department of Epidemiology, College of Preventive Medicine, Third Military Medical University, Chongqing, China
| | - Huijie Huang
- Department of Epidemiology and Biostatistics, School of Public Health, Tianjin Medical University, Tianjin, China
| | - Michelle M. Dunk
- Aging Research Center, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden
| | - Sakura Sakakibara
- Aging Research Center, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden
| | - Marc Guitart-Masip
- Aging Research Center, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden
| | - Goran Papenberg
- Aging Research Center, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden
| | - Weili Xu
- Aging Research Center, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden
- Department of Epidemiology and Biostatistics, School of Public Health, Tianjin Medical University, Tianjin, China
| |
Collapse
|
7
|
Gaser C, Kalc P, Cole JH. A perspective on brain-age estimation and its clinical promise. NATURE COMPUTATIONAL SCIENCE 2024; 4:744-751. [PMID: 39048692 DOI: 10.1038/s43588-024-00659-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/11/2023] [Accepted: 06/12/2024] [Indexed: 07/27/2024]
Abstract
Brain-age estimation has gained increased attention in the neuroscientific community owing to its potential use as a biomarker of brain health. The difference between estimated and chronological age based on neuroimaging data enables a unique perspective on brain development and aging, with multiple open questions still remaining in the brain-age research field. This Perspective presents an overview of current advancements in the field and envisions the future evolution of the brain-age framework before its potential deployment in hospital settings.
Collapse
Affiliation(s)
- Christian Gaser
- Structural Brain Mapping Group, Department of Neurology, Jena University Hospital, Jena, Germany.
- Department of Psychiatry and Psychotherapy, Jena University Hospital, Jena, Germany.
- German Centre for Mental Health (DZPG), Jena-Halle-Magdeburg, Jena, Germany.
| | - Polona Kalc
- Structural Brain Mapping Group, Department of Neurology, Jena University Hospital, Jena, Germany
| | - James H Cole
- Centre for Medical Image Computing, Department of Computer Science, University College London, London, UK
- Dementia Research Centre, Queen Square Institute of Neurology, University College London, London, UK
| |
Collapse
|
8
|
Zhu X, Sun S, Lin L, Wu Y, Ma X. Transformer-based approaches for neuroimaging: an in-depth review of their role in classification and regression tasks. Rev Neurosci 2024:revneuro-2024-0088. [PMID: 39333087 DOI: 10.1515/revneuro-2024-0088] [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: 07/02/2024] [Accepted: 09/13/2024] [Indexed: 09/29/2024]
Abstract
In the ever-evolving landscape of deep learning (DL), the transformer model emerges as a formidable neural network architecture, gaining significant traction in neuroimaging-based classification and regression tasks. This paper presents an extensive examination of transformer's application in neuroimaging, surveying recent literature to elucidate its current status and research advancement. Commencing with an exposition on the fundamental principles and structures of the transformer model and its variants, this review navigates through the methodologies and experimental findings pertaining to their utilization in neuroimage classification and regression tasks. We highlight the transformer model's prowess in neuroimaging, showcasing its exceptional performance in classification endeavors while also showcasing its burgeoning potential in regression tasks. Concluding with an assessment of prevailing challenges and future trajectories, this paper proffers insights into prospective research directions. By elucidating the current landscape and envisaging future trends, this review enhances comprehension of transformer's role in neuroimaging tasks, furnishing valuable guidance for further inquiry.
Collapse
Affiliation(s)
- Xinyu Zhu
- Department of Biomedical Engineering, 12496 College of Chemistry and Life Sciences, Beijing University of Technology , Beijing, 100124, China
| | - Shen Sun
- Department of Biomedical Engineering, 12496 College of Chemistry and Life Sciences, Beijing University of Technology , Beijing, 100124, China
| | - Lan Lin
- Department of Biomedical Engineering, 12496 College of Chemistry and Life Sciences, Beijing University of Technology , Beijing, 100124, China
| | - Yutong Wu
- Department of Biomedical Engineering, 12496 College of Chemistry and Life Sciences, Beijing University of Technology , Beijing, 100124, China
| | - Xiangge Ma
- Department of Biomedical Engineering, 12496 College of Chemistry and Life Sciences, Beijing University of Technology , Beijing, 100124, China
| |
Collapse
|
9
|
Feng L, Milleson HS, Ye Z, Canida T, Ke H, Liang M, Gao S, Chen S, Hong LE, Kochunov P, Lei DKY, Ma T. Nongenetic and Genetic Factors Associated with White Matter Brain Aging: Exposome-Wide and Genome-Wide Association Study. Genes (Basel) 2024; 15:1285. [PMID: 39457408 PMCID: PMC11507416 DOI: 10.3390/genes15101285] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2024] [Revised: 09/18/2024] [Accepted: 09/19/2024] [Indexed: 10/28/2024] Open
Abstract
BACKGROUND/OBJECTIVES Human brain aging is a complex process that affects various aspects of brain function and structure, increasing susceptibility to neurological and psychiatric disorders. A number of nongenetic (e.g., environmental and lifestyle) and genetic risk factors are found to contribute to the varying rates at which the brain ages among individuals. METHODS In this paper, we conducted both an exposome-wide association study (XWAS) and a genome-wide association study (GWAS) on white matter brain aging in the UK Biobank, revealing the multifactorial nature of brain aging. We applied a machine learning algorithm and leveraged fractional anisotropy tract measurements from diffusion tensor imaging data to predict the white matter brain age gap (BAG) and treated it as the marker of brain aging. For XWAS, we included 107 variables encompassing five major categories of modifiable exposures that potentially impact brain aging and performed both univariate and multivariate analysis to select the final set of nongenetic risk factors. RESULTS We found current tobacco smoking, dietary habits including oily fish, beef, lamb, cereal, and coffee intake, length of mobile phone use, use of UV protection, and frequency of solarium/sunlamp use were associated with the BAG. In genetic analysis, we identified several SNPs on chromosome 3 mapped to genes IP6K1, GMNC, OSTN, and SLC25A20 significantly associated with the BAG, showing the high heritability and polygenic architecture of human brain aging. CONCLUSIONS The critical nongenetic and genetic risk factors identified in our study provide insights into the causal relationship between white matter brain aging and neurodegenerative diseases.
Collapse
Affiliation(s)
- Li Feng
- Department of Nutrition and Food Science, College of Agriculture & Natural Resources, University of Maryland, College Park, MD 20740, USA; (L.F.); (D.K.Y.L.)
- Department of Epidemiology and Biostatistics, School of Public Health, University of Maryland, College Park, MD 20740, USA; (H.S.M.); (T.C.); (H.K.); (M.L.)
| | - Halley S. Milleson
- Department of Epidemiology and Biostatistics, School of Public Health, University of Maryland, College Park, MD 20740, USA; (H.S.M.); (T.C.); (H.K.); (M.L.)
- Department of Mathematics, The College of Computer, Mathematical, and Natural Sciences, University of Maryland, College Park, MD 20740, USA
| | - Zhenyao Ye
- Maryland Psychiatric Research Center, Department of Psychiatry, School of Medicine, University of Maryland, Baltimore, MD 21228, USA; (Z.Y.); (S.G.); (S.C.)
- Division of Biostatistics and Bioinformatics, Department of Epidemiology and Public Health, School of Medicine, University of Maryland, Baltimore, MD 21201, USA
| | - Travis Canida
- Department of Epidemiology and Biostatistics, School of Public Health, University of Maryland, College Park, MD 20740, USA; (H.S.M.); (T.C.); (H.K.); (M.L.)
- Department of Mathematics, The College of Computer, Mathematical, and Natural Sciences, University of Maryland, College Park, MD 20740, USA
| | - Hongjie Ke
- Department of Epidemiology and Biostatistics, School of Public Health, University of Maryland, College Park, MD 20740, USA; (H.S.M.); (T.C.); (H.K.); (M.L.)
| | - Menglu Liang
- Department of Epidemiology and Biostatistics, School of Public Health, University of Maryland, College Park, MD 20740, USA; (H.S.M.); (T.C.); (H.K.); (M.L.)
| | - Si Gao
- Maryland Psychiatric Research Center, Department of Psychiatry, School of Medicine, University of Maryland, Baltimore, MD 21228, USA; (Z.Y.); (S.G.); (S.C.)
- Louis A. Faillace Department of Psychiatry & Behavioral Sciences, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX 77030, USA; (L.E.H.); (P.K.)
| | - Shuo Chen
- Maryland Psychiatric Research Center, Department of Psychiatry, School of Medicine, University of Maryland, Baltimore, MD 21228, USA; (Z.Y.); (S.G.); (S.C.)
- Division of Biostatistics and Bioinformatics, Department of Epidemiology and Public Health, School of Medicine, University of Maryland, Baltimore, MD 21201, USA
| | - L. Elliot Hong
- Louis A. Faillace Department of Psychiatry & Behavioral Sciences, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX 77030, USA; (L.E.H.); (P.K.)
| | - Peter Kochunov
- Louis A. Faillace Department of Psychiatry & Behavioral Sciences, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX 77030, USA; (L.E.H.); (P.K.)
| | - David K. Y. Lei
- Department of Nutrition and Food Science, College of Agriculture & Natural Resources, University of Maryland, College Park, MD 20740, USA; (L.F.); (D.K.Y.L.)
| | - Tianzhou Ma
- Department of Epidemiology and Biostatistics, School of Public Health, University of Maryland, College Park, MD 20740, USA; (H.S.M.); (T.C.); (H.K.); (M.L.)
| |
Collapse
|
10
|
Thornton V, Chang Y, Chaloemtoem A, Anokhin AP, Bijsterbosch J, Foraker R, Hancock DB, Johnson EO, White JD, Hartz SM, Bierut LJ. Alcohol, smoking, and brain structure: common or substance specific associations. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.09.25.24313371. [PMID: 39399056 PMCID: PMC11469368 DOI: 10.1101/2024.09.25.24313371] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 10/15/2024]
Abstract
Alcohol use and smoking are common substance-use behaviors with well-established negative health effects, including decreased brain health. We examined whether alcohol use and smoking were associated with the same neuroimaging-derived brain measures. We further explored whether the effects of alcohol use and smoking on the brain were additive or interactive. We leveraged a cohort of 36,309 participants with neuroimaging data from the UK Biobank. We used linear regression to determine the association between 354 neuroimaging-derived brain measures and alcohol use defined as drinks per week, pack years of smoking, and drinks per week × pack years smoking interaction. To assess whether the brain associations with alcohol are broadly similar or different from the associations with smoking, we calculated the correlation between z-scores of association for drinks per week and pack years smoking. Results indicated overall moderate positive correlation in the associations across measures representing brain structure, magnetic susceptibility, and white matter tract microstructure, indicating greater similarity than difference in the brain measures associated with alcohol use and smoking. The only evidence of an interaction between drinks per week and pack years smoking was seen in measures representing magnetic susceptibility in subcortical structures. The effects of alcohol use and smoking on brain health appeared to be additive rather than multiplicative for all other brain measures studied. 97% (224/230) of associations with alcohol and 100% (167/167) of the associations with smoking that surpassed a p value threshold are in a direction that can be interpreted to reflect reduced brain health. Our results underscore the similarity of the adverse associations between use of these substances and neuroimaging derived brain measures.
Collapse
Affiliation(s)
- Vera Thornton
- Department of Psychiatry, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Yoonhoo Chang
- Department of Psychiatry, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Ariya Chaloemtoem
- Department of Psychiatry, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Andrey P. Anokhin
- Department of Psychiatry, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Janine Bijsterbosch
- Department of Radiology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Randi Foraker
- Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Dana B. Hancock
- GenOmics and Translational Research Center, RTI International, Research Triangle Park, North Carolina, USA
| | - Eric O. Johnson
- GenOmics and Translational Research Center, RTI International, Research Triangle Park, North Carolina, USA
- Fellow Program, RTI International, Research Triangle Park, North Carolina, USA
| | - Julie D. White
- GenOmics and Translational Research Center, RTI International, Research Triangle Park, North Carolina, USA
| | - Sarah M. Hartz
- Department of Psychiatry, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Laura J. Bierut
- Department of Psychiatry, Washington University School of Medicine, St. Louis, Missouri, USA
| |
Collapse
|
11
|
Subramaniapillai S, Schindler LS, Redmond P, Bastin ME, Wardlaw JM, Valdés Hernández M, Maniega SM, Aribisala B, Westlye LT, Coath W, Groves J, Cash DM, Barnes J, James SN, Sudre CH, Barkhof F, Richards M, Corley J, Russ TC, Cox SR, Schott JM, Cole JH, de Lange AMG. Sex-Dependent Effects of Cardiometabolic Health and APOE4 on Brain Age: A Longitudinal Cohort Study. Neurology 2024; 103:e209744. [PMID: 39173100 PMCID: PMC11379441 DOI: 10.1212/wnl.0000000000209744] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/24/2024] Open
Abstract
BACKGROUND AND OBJECTIVES The aging population is growing faster than all other demographic strata. With older age comes a greater risk of health conditions such as obesity and high blood pressure (BP). These cardiometabolic risk factors (CMRs) exhibit prominent sex differences in midlife and aging, yet their influence on brain health in females vs males is largely unexplored. In this study, we investigated sex differences in relationships between BP, body mass index (BMI), and brain age over time and tested for interactions with APOE ε4 genotype (APOE4), a known genetic risk factor of Alzheimer disease. METHODS The sample included participants from 2 United Kingdom-based longitudinal birth cohorts, the Lothian Birth Cohort (1936) and Insight 46 (1946). Participants with MRI data from at least 1 time point were included to evaluate sex differences in associations between CMRs and brain age. The open-access software package brainageR 2.1 was used to estimate brain age for each participant. Linear mixed-effects models were used to assess the relationships between brain age, BMI, BP, and APOE4 status (i.e., carrier vs noncarrier) in males and females over time. RESULTS The combined sample comprised 1,120 participants (48% female) with a mean age (SD) of 73 (0.72) years in the Lothian Birth Cohort and 71 (0.68) years in Insight 46 at the time point 1 assessment. Approximately 30% of participants were APOE4 carriers. Higher systolic and diastolic BP was significantly associated with older brain age in females only (β = 0.43-0.56, p < 0.05). Among males, higher BMI was associated with older brain age across time points and APOE4 groups (β = 0.72-0.77, p < 0.05). In females, higher BMI was linked to older brain age among APOE4 noncarriers (β = 0.68-0.99, p < 0.05), whereas higher BMI was linked to younger brain age among carriers, particularly at the last time point (β = -1.75, p < 0.05). DISCUSSION This study indicates sex-dependent and time-dependent relationships between CMRs, APOE4 status, and brain age. Our findings highlight the necessity of sex-stratified analyses to elucidate the role of CMRs in individual aging trajectories, providing a basis for developing personalized preventive interventions.
Collapse
Affiliation(s)
- Sivaniya Subramaniapillai
- From the Department of Clinical Neuroscience (S.S., L.S.S., A.-M.G.d.L.), Lausanne University Hospital and University of Lausanne, Switzerland; Department of Psychology (P.R., M.E.B., J.M.W., M.V.H., S.M.M., B.A., J.C., T.C.R., S.R.C.), University of Edinburgh, United Kingdom; Department of Psychology (L.T.W.), University of Oslo, Norway; Dementia Research Centre (W.C., J.G., D.M.C., J.B., S.-N.J., C.H.S., J.M.S.), Centre for Medical Image Computing (C.H.S., F.B., J.H.C.), and MRC Unit for Lifelong Health and Ageing (M.R., S.-N.J., C.H.S.), University College London, United Kingdom
| | - Louise S Schindler
- From the Department of Clinical Neuroscience (S.S., L.S.S., A.-M.G.d.L.), Lausanne University Hospital and University of Lausanne, Switzerland; Department of Psychology (P.R., M.E.B., J.M.W., M.V.H., S.M.M., B.A., J.C., T.C.R., S.R.C.), University of Edinburgh, United Kingdom; Department of Psychology (L.T.W.), University of Oslo, Norway; Dementia Research Centre (W.C., J.G., D.M.C., J.B., S.-N.J., C.H.S., J.M.S.), Centre for Medical Image Computing (C.H.S., F.B., J.H.C.), and MRC Unit for Lifelong Health and Ageing (M.R., S.-N.J., C.H.S.), University College London, United Kingdom
| | - Paul Redmond
- From the Department of Clinical Neuroscience (S.S., L.S.S., A.-M.G.d.L.), Lausanne University Hospital and University of Lausanne, Switzerland; Department of Psychology (P.R., M.E.B., J.M.W., M.V.H., S.M.M., B.A., J.C., T.C.R., S.R.C.), University of Edinburgh, United Kingdom; Department of Psychology (L.T.W.), University of Oslo, Norway; Dementia Research Centre (W.C., J.G., D.M.C., J.B., S.-N.J., C.H.S., J.M.S.), Centre for Medical Image Computing (C.H.S., F.B., J.H.C.), and MRC Unit for Lifelong Health and Ageing (M.R., S.-N.J., C.H.S.), University College London, United Kingdom
| | - Mark E Bastin
- From the Department of Clinical Neuroscience (S.S., L.S.S., A.-M.G.d.L.), Lausanne University Hospital and University of Lausanne, Switzerland; Department of Psychology (P.R., M.E.B., J.M.W., M.V.H., S.M.M., B.A., J.C., T.C.R., S.R.C.), University of Edinburgh, United Kingdom; Department of Psychology (L.T.W.), University of Oslo, Norway; Dementia Research Centre (W.C., J.G., D.M.C., J.B., S.-N.J., C.H.S., J.M.S.), Centre for Medical Image Computing (C.H.S., F.B., J.H.C.), and MRC Unit for Lifelong Health and Ageing (M.R., S.-N.J., C.H.S.), University College London, United Kingdom
| | - Joanna M Wardlaw
- From the Department of Clinical Neuroscience (S.S., L.S.S., A.-M.G.d.L.), Lausanne University Hospital and University of Lausanne, Switzerland; Department of Psychology (P.R., M.E.B., J.M.W., M.V.H., S.M.M., B.A., J.C., T.C.R., S.R.C.), University of Edinburgh, United Kingdom; Department of Psychology (L.T.W.), University of Oslo, Norway; Dementia Research Centre (W.C., J.G., D.M.C., J.B., S.-N.J., C.H.S., J.M.S.), Centre for Medical Image Computing (C.H.S., F.B., J.H.C.), and MRC Unit for Lifelong Health and Ageing (M.R., S.-N.J., C.H.S.), University College London, United Kingdom
| | - Maria Valdés Hernández
- From the Department of Clinical Neuroscience (S.S., L.S.S., A.-M.G.d.L.), Lausanne University Hospital and University of Lausanne, Switzerland; Department of Psychology (P.R., M.E.B., J.M.W., M.V.H., S.M.M., B.A., J.C., T.C.R., S.R.C.), University of Edinburgh, United Kingdom; Department of Psychology (L.T.W.), University of Oslo, Norway; Dementia Research Centre (W.C., J.G., D.M.C., J.B., S.-N.J., C.H.S., J.M.S.), Centre for Medical Image Computing (C.H.S., F.B., J.H.C.), and MRC Unit for Lifelong Health and Ageing (M.R., S.-N.J., C.H.S.), University College London, United Kingdom
| | - Susana Muñoz Maniega
- From the Department of Clinical Neuroscience (S.S., L.S.S., A.-M.G.d.L.), Lausanne University Hospital and University of Lausanne, Switzerland; Department of Psychology (P.R., M.E.B., J.M.W., M.V.H., S.M.M., B.A., J.C., T.C.R., S.R.C.), University of Edinburgh, United Kingdom; Department of Psychology (L.T.W.), University of Oslo, Norway; Dementia Research Centre (W.C., J.G., D.M.C., J.B., S.-N.J., C.H.S., J.M.S.), Centre for Medical Image Computing (C.H.S., F.B., J.H.C.), and MRC Unit for Lifelong Health and Ageing (M.R., S.-N.J., C.H.S.), University College London, United Kingdom
| | - Benjamin Aribisala
- From the Department of Clinical Neuroscience (S.S., L.S.S., A.-M.G.d.L.), Lausanne University Hospital and University of Lausanne, Switzerland; Department of Psychology (P.R., M.E.B., J.M.W., M.V.H., S.M.M., B.A., J.C., T.C.R., S.R.C.), University of Edinburgh, United Kingdom; Department of Psychology (L.T.W.), University of Oslo, Norway; Dementia Research Centre (W.C., J.G., D.M.C., J.B., S.-N.J., C.H.S., J.M.S.), Centre for Medical Image Computing (C.H.S., F.B., J.H.C.), and MRC Unit for Lifelong Health and Ageing (M.R., S.-N.J., C.H.S.), University College London, United Kingdom
| | - Lars T Westlye
- From the Department of Clinical Neuroscience (S.S., L.S.S., A.-M.G.d.L.), Lausanne University Hospital and University of Lausanne, Switzerland; Department of Psychology (P.R., M.E.B., J.M.W., M.V.H., S.M.M., B.A., J.C., T.C.R., S.R.C.), University of Edinburgh, United Kingdom; Department of Psychology (L.T.W.), University of Oslo, Norway; Dementia Research Centre (W.C., J.G., D.M.C., J.B., S.-N.J., C.H.S., J.M.S.), Centre for Medical Image Computing (C.H.S., F.B., J.H.C.), and MRC Unit for Lifelong Health and Ageing (M.R., S.-N.J., C.H.S.), University College London, United Kingdom
| | - William Coath
- From the Department of Clinical Neuroscience (S.S., L.S.S., A.-M.G.d.L.), Lausanne University Hospital and University of Lausanne, Switzerland; Department of Psychology (P.R., M.E.B., J.M.W., M.V.H., S.M.M., B.A., J.C., T.C.R., S.R.C.), University of Edinburgh, United Kingdom; Department of Psychology (L.T.W.), University of Oslo, Norway; Dementia Research Centre (W.C., J.G., D.M.C., J.B., S.-N.J., C.H.S., J.M.S.), Centre for Medical Image Computing (C.H.S., F.B., J.H.C.), and MRC Unit for Lifelong Health and Ageing (M.R., S.-N.J., C.H.S.), University College London, United Kingdom
| | - James Groves
- From the Department of Clinical Neuroscience (S.S., L.S.S., A.-M.G.d.L.), Lausanne University Hospital and University of Lausanne, Switzerland; Department of Psychology (P.R., M.E.B., J.M.W., M.V.H., S.M.M., B.A., J.C., T.C.R., S.R.C.), University of Edinburgh, United Kingdom; Department of Psychology (L.T.W.), University of Oslo, Norway; Dementia Research Centre (W.C., J.G., D.M.C., J.B., S.-N.J., C.H.S., J.M.S.), Centre for Medical Image Computing (C.H.S., F.B., J.H.C.), and MRC Unit for Lifelong Health and Ageing (M.R., S.-N.J., C.H.S.), University College London, United Kingdom
| | - David M Cash
- From the Department of Clinical Neuroscience (S.S., L.S.S., A.-M.G.d.L.), Lausanne University Hospital and University of Lausanne, Switzerland; Department of Psychology (P.R., M.E.B., J.M.W., M.V.H., S.M.M., B.A., J.C., T.C.R., S.R.C.), University of Edinburgh, United Kingdom; Department of Psychology (L.T.W.), University of Oslo, Norway; Dementia Research Centre (W.C., J.G., D.M.C., J.B., S.-N.J., C.H.S., J.M.S.), Centre for Medical Image Computing (C.H.S., F.B., J.H.C.), and MRC Unit for Lifelong Health and Ageing (M.R., S.-N.J., C.H.S.), University College London, United Kingdom
| | - Josephine Barnes
- From the Department of Clinical Neuroscience (S.S., L.S.S., A.-M.G.d.L.), Lausanne University Hospital and University of Lausanne, Switzerland; Department of Psychology (P.R., M.E.B., J.M.W., M.V.H., S.M.M., B.A., J.C., T.C.R., S.R.C.), University of Edinburgh, United Kingdom; Department of Psychology (L.T.W.), University of Oslo, Norway; Dementia Research Centre (W.C., J.G., D.M.C., J.B., S.-N.J., C.H.S., J.M.S.), Centre for Medical Image Computing (C.H.S., F.B., J.H.C.), and MRC Unit for Lifelong Health and Ageing (M.R., S.-N.J., C.H.S.), University College London, United Kingdom
| | - Sarah-Naomi James
- From the Department of Clinical Neuroscience (S.S., L.S.S., A.-M.G.d.L.), Lausanne University Hospital and University of Lausanne, Switzerland; Department of Psychology (P.R., M.E.B., J.M.W., M.V.H., S.M.M., B.A., J.C., T.C.R., S.R.C.), University of Edinburgh, United Kingdom; Department of Psychology (L.T.W.), University of Oslo, Norway; Dementia Research Centre (W.C., J.G., D.M.C., J.B., S.-N.J., C.H.S., J.M.S.), Centre for Medical Image Computing (C.H.S., F.B., J.H.C.), and MRC Unit for Lifelong Health and Ageing (M.R., S.-N.J., C.H.S.), University College London, United Kingdom
| | - Carole H Sudre
- From the Department of Clinical Neuroscience (S.S., L.S.S., A.-M.G.d.L.), Lausanne University Hospital and University of Lausanne, Switzerland; Department of Psychology (P.R., M.E.B., J.M.W., M.V.H., S.M.M., B.A., J.C., T.C.R., S.R.C.), University of Edinburgh, United Kingdom; Department of Psychology (L.T.W.), University of Oslo, Norway; Dementia Research Centre (W.C., J.G., D.M.C., J.B., S.-N.J., C.H.S., J.M.S.), Centre for Medical Image Computing (C.H.S., F.B., J.H.C.), and MRC Unit for Lifelong Health and Ageing (M.R., S.-N.J., C.H.S.), University College London, United Kingdom
| | - Frederik Barkhof
- From the Department of Clinical Neuroscience (S.S., L.S.S., A.-M.G.d.L.), Lausanne University Hospital and University of Lausanne, Switzerland; Department of Psychology (P.R., M.E.B., J.M.W., M.V.H., S.M.M., B.A., J.C., T.C.R., S.R.C.), University of Edinburgh, United Kingdom; Department of Psychology (L.T.W.), University of Oslo, Norway; Dementia Research Centre (W.C., J.G., D.M.C., J.B., S.-N.J., C.H.S., J.M.S.), Centre for Medical Image Computing (C.H.S., F.B., J.H.C.), and MRC Unit for Lifelong Health and Ageing (M.R., S.-N.J., C.H.S.), University College London, United Kingdom
| | - Marcus Richards
- From the Department of Clinical Neuroscience (S.S., L.S.S., A.-M.G.d.L.), Lausanne University Hospital and University of Lausanne, Switzerland; Department of Psychology (P.R., M.E.B., J.M.W., M.V.H., S.M.M., B.A., J.C., T.C.R., S.R.C.), University of Edinburgh, United Kingdom; Department of Psychology (L.T.W.), University of Oslo, Norway; Dementia Research Centre (W.C., J.G., D.M.C., J.B., S.-N.J., C.H.S., J.M.S.), Centre for Medical Image Computing (C.H.S., F.B., J.H.C.), and MRC Unit for Lifelong Health and Ageing (M.R., S.-N.J., C.H.S.), University College London, United Kingdom
| | - Janie Corley
- From the Department of Clinical Neuroscience (S.S., L.S.S., A.-M.G.d.L.), Lausanne University Hospital and University of Lausanne, Switzerland; Department of Psychology (P.R., M.E.B., J.M.W., M.V.H., S.M.M., B.A., J.C., T.C.R., S.R.C.), University of Edinburgh, United Kingdom; Department of Psychology (L.T.W.), University of Oslo, Norway; Dementia Research Centre (W.C., J.G., D.M.C., J.B., S.-N.J., C.H.S., J.M.S.), Centre for Medical Image Computing (C.H.S., F.B., J.H.C.), and MRC Unit for Lifelong Health and Ageing (M.R., S.-N.J., C.H.S.), University College London, United Kingdom
| | - Tom C Russ
- From the Department of Clinical Neuroscience (S.S., L.S.S., A.-M.G.d.L.), Lausanne University Hospital and University of Lausanne, Switzerland; Department of Psychology (P.R., M.E.B., J.M.W., M.V.H., S.M.M., B.A., J.C., T.C.R., S.R.C.), University of Edinburgh, United Kingdom; Department of Psychology (L.T.W.), University of Oslo, Norway; Dementia Research Centre (W.C., J.G., D.M.C., J.B., S.-N.J., C.H.S., J.M.S.), Centre for Medical Image Computing (C.H.S., F.B., J.H.C.), and MRC Unit for Lifelong Health and Ageing (M.R., S.-N.J., C.H.S.), University College London, United Kingdom
| | - Simon R Cox
- From the Department of Clinical Neuroscience (S.S., L.S.S., A.-M.G.d.L.), Lausanne University Hospital and University of Lausanne, Switzerland; Department of Psychology (P.R., M.E.B., J.M.W., M.V.H., S.M.M., B.A., J.C., T.C.R., S.R.C.), University of Edinburgh, United Kingdom; Department of Psychology (L.T.W.), University of Oslo, Norway; Dementia Research Centre (W.C., J.G., D.M.C., J.B., S.-N.J., C.H.S., J.M.S.), Centre for Medical Image Computing (C.H.S., F.B., J.H.C.), and MRC Unit for Lifelong Health and Ageing (M.R., S.-N.J., C.H.S.), University College London, United Kingdom
| | - Jonathan M Schott
- From the Department of Clinical Neuroscience (S.S., L.S.S., A.-M.G.d.L.), Lausanne University Hospital and University of Lausanne, Switzerland; Department of Psychology (P.R., M.E.B., J.M.W., M.V.H., S.M.M., B.A., J.C., T.C.R., S.R.C.), University of Edinburgh, United Kingdom; Department of Psychology (L.T.W.), University of Oslo, Norway; Dementia Research Centre (W.C., J.G., D.M.C., J.B., S.-N.J., C.H.S., J.M.S.), Centre for Medical Image Computing (C.H.S., F.B., J.H.C.), and MRC Unit for Lifelong Health and Ageing (M.R., S.-N.J., C.H.S.), University College London, United Kingdom
| | - James H Cole
- From the Department of Clinical Neuroscience (S.S., L.S.S., A.-M.G.d.L.), Lausanne University Hospital and University of Lausanne, Switzerland; Department of Psychology (P.R., M.E.B., J.M.W., M.V.H., S.M.M., B.A., J.C., T.C.R., S.R.C.), University of Edinburgh, United Kingdom; Department of Psychology (L.T.W.), University of Oslo, Norway; Dementia Research Centre (W.C., J.G., D.M.C., J.B., S.-N.J., C.H.S., J.M.S.), Centre for Medical Image Computing (C.H.S., F.B., J.H.C.), and MRC Unit for Lifelong Health and Ageing (M.R., S.-N.J., C.H.S.), University College London, United Kingdom
| | - Ann-Marie G de Lange
- From the Department of Clinical Neuroscience (S.S., L.S.S., A.-M.G.d.L.), Lausanne University Hospital and University of Lausanne, Switzerland; Department of Psychology (P.R., M.E.B., J.M.W., M.V.H., S.M.M., B.A., J.C., T.C.R., S.R.C.), University of Edinburgh, United Kingdom; Department of Psychology (L.T.W.), University of Oslo, Norway; Dementia Research Centre (W.C., J.G., D.M.C., J.B., S.-N.J., C.H.S., J.M.S.), Centre for Medical Image Computing (C.H.S., F.B., J.H.C.), and MRC Unit for Lifelong Health and Ageing (M.R., S.-N.J., C.H.S.), University College London, United Kingdom
| |
Collapse
|
12
|
Dular L, Špiclin Ž. Analysis of Brain Age Gap across Subject Cohorts and Prediction Model Architectures. Biomedicines 2024; 12:2139. [PMID: 39335651 PMCID: PMC11428686 DOI: 10.3390/biomedicines12092139] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2024] [Revised: 09/12/2024] [Accepted: 09/16/2024] [Indexed: 09/30/2024] Open
Abstract
Background: Brain age prediction from brain MRI scans and the resulting brain age gap (BAG)-the difference between predicted brain age and chronological age-is a general biomarker for a variety of neurological, psychiatric, and other diseases or disorders. Methods: This study examined the differences in BAG values derived from T1-weighted scans using five state-of-the-art deep learning model architectures previously used in the brain age literature: 2D/3D VGG, RelationNet, ResNet, and SFCN. The models were evaluated on healthy controls and cohorts with sleep apnea, diabetes, multiple sclerosis, Parkinson's disease, mild cognitive impairment, and Alzheimer's disease, employing rigorous statistical analysis, including repeated model training and linear mixed-effects models. Results: All five models consistently identified a statistically significant positive BAG for diabetes (ranging from 0.79 years with RelationNet to 2.13 years with SFCN), multiple sclerosis (2.67 years with 3D VGG to 4.24 years with 2D VGG), mild cognitive impairment (2.13 years with 2D VGG to 2.59 years with 3D VGG), and Alzheimer's dementia (5.54 years with ResNet to 6.48 years with SFCN). For Parkinson's disease, a statistically significant BAG increase was observed in all models except ResNet (1.30 years with 2D VGG to 2.59 years with 3D VGG). For sleep apnea, a statistically significant BAG increase was only detected with the SFCN model (1.59 years). Additionally, we observed a trend of decreasing BAG with increasing chronological age, which was more pronounced in diseased cohorts, particularly those with the largest BAG, such as multiple sclerosis (-0.34 to -0.2), mild cognitive impairment (-0.37 to -0.26), and Alzheimer's dementia (-0.66 to -0.47), compared to healthy controls (-0.18 to -0.1). Conclusions: Consistent with previous research, Alzheimer's dementia and multiple sclerosis exhibited the largest BAG across all models, with SFCN predicting the highest BAG overall. The negative BAG trend suggests a complex interplay of survival bias, disease progression, adaptation, and therapy that influences brain age prediction across the age spectrum.
Collapse
Affiliation(s)
| | - Žiga Špiclin
- University of Ljubljana, Faculty of Electrical Engineering, Tržaška cesta 25, 1000 Ljubljana, Slovenia
| | | | | |
Collapse
|
13
|
Jiang Y, Mu Y, Xu Z, Liu Q, Wang S, Wang H, Feng J. Identifying individual brain development using multimodality brain network. Commun Biol 2024; 7:1163. [PMID: 39289448 PMCID: PMC11408623 DOI: 10.1038/s42003-024-06876-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2024] [Accepted: 09/10/2024] [Indexed: 09/19/2024] Open
Abstract
The cortical development of our brains is in a hierarchical manner and promotes the emergence of large-scale functional hierarchy. However, under interindividual heterogenicity, how the spatiotemporal features of brain networks reflect brain development and mental health remains unclear. Here we collect both resting-state electroencephalography and functional magnetic resonance imaging data from the Child Mind Institute Biobank to demonstrate that during brain growth, the global dynamic patterns of brain states become more active and the dominant networks shift from sensory to higher-level networks; the individual functional network patterns become more similar to that of adults and their spatial coupling tends to be invariable. Furthermore, the properties of multimodality brain networks are sufficiently robust to identify healthy brain age and mental disorders at specific ages. Therefore, multimodality brain networks provide new insights into the functional development of the brain and a more reliable and reasonable approach for age prediction and individual diagnosis.
Collapse
Affiliation(s)
- Yuwei Jiang
- Institute of Science and Technology for Brain-Inspired Intelligence, Fudan University, Shanghai, China.
- Key Laboratory of Computational Neuroscience and Brain-Inspired Intelligence, Fudan University, Ministry of Education, Shanghai, China.
| | - Yangjiayi Mu
- Institute of Science and Technology for Brain-Inspired Intelligence, Fudan University, Shanghai, China
- Key Laboratory of Computational Neuroscience and Brain-Inspired Intelligence, Fudan University, Ministry of Education, Shanghai, China
| | - Zhao Xu
- Institute of Science and Technology for Brain-Inspired Intelligence, Fudan University, Shanghai, China
- Key Laboratory of Computational Neuroscience and Brain-Inspired Intelligence, Fudan University, Ministry of Education, Shanghai, China
| | - Qingyang Liu
- Institute of Science and Technology for Brain-Inspired Intelligence, Fudan University, Shanghai, China
- Key Laboratory of Computational Neuroscience and Brain-Inspired Intelligence, Fudan University, Ministry of Education, Shanghai, China
| | - Shouyan Wang
- Institute of Science and Technology for Brain-Inspired Intelligence, Fudan University, Shanghai, China
- Key Laboratory of Computational Neuroscience and Brain-Inspired Intelligence, Fudan University, Ministry of Education, Shanghai, China
| | - He Wang
- Institute of Science and Technology for Brain-Inspired Intelligence, Fudan University, Shanghai, China.
- Key Laboratory of Computational Neuroscience and Brain-Inspired Intelligence, Fudan University, Ministry of Education, Shanghai, China.
| | - Jianfeng Feng
- Institute of Science and Technology for Brain-Inspired Intelligence, Fudan University, Shanghai, China.
- Key Laboratory of Computational Neuroscience and Brain-Inspired Intelligence, Fudan University, Ministry of Education, Shanghai, China.
| |
Collapse
|
14
|
Gage AT, Stone JR, Wilde EA, McCauley SR, Welsh RC, Mugler JP, Tustison N, Avants B, Whitlow CT, Lancashire L, Bhatt SD, Haas M. Normative Neuroimaging Library: Designing a Comprehensive and Demographically Diverse Dataset of Healthy Controls to Support Traumatic Brain Injury Diagnostic and Therapeutic Development. J Neurotrauma 2024. [PMID: 39235436 DOI: 10.1089/neu.2024.0128] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/06/2024] Open
Abstract
The past decade has seen impressive advances in neuroimaging, moving from qualitative to quantitative outputs. Available techniques now allow for the inference of microscopic changes occurring in white and gray matter, along with alterations in physiology and function. These existing and emerging techniques hold the potential of providing unprecedented capabilities in achieving a diagnosis and predicting outcomes for traumatic brain injury (TBI) and a variety of other neurological diseases. To see this promise move from the research lab into clinical care, an understanding is needed of what normal data look like for all age ranges, sex, and other demographic and socioeconomic categories. Clinicians can only use the results of imaging scans to support their decision-making if they know how the results for their patient compare with a normative standard. This potential for utilizing magnetic resonance imaging (MRI) in TBI diagnosis motivated the American College of Radiology and Cohen Veterans Bioscience to create a reference database of healthy individuals with neuroimaging, demographic data, and characterization of psychological functioning and neurocognitive data that will serve as a normative resource for clinicians and researchers for development of diagnostics and therapeutics for TBI and other brain disorders. The goal of this article is to introduce the large, well-curated Normative Neuroimaging Library (NNL) to the research community. NNL consists of data collected from ∼1900 healthy participants. The highlights of NNL are (1) data are collected across a diverse population, including civilians, veterans, and active-duty service members with an age range (18-64 years) not well represented in existing datasets; (2) comprehensive structural and functional neuroimaging acquisition with state-of-the-art sequences (including structural, diffusion, and functional MRI; raw scanner data are preserved, allowing higher quality data to be derived in the future; standardized imaging acquisition protocols across sites reflect sequences and parameters often recommended for use with various neurological and psychiatric conditions, including TBI, post-traumatic stress disorder, stroke, neurodegenerative disorders, and neoplastic disease); and (3) the collection of comprehensive demographic details, medical history, and a broad structured clinical assessment, including cognition and psychological scales, relevant to multiple neurological conditions with functional sequelae. Thus, NNL provides a demographically diverse population of healthy individuals who can serve as a comparison group for brain injury study and clinical samples, providing a strong foundation for precision medicine. Use cases include the creation of imaging-derived phenotypes (IDPs), derivation of reference ranges of imaging measures, and use of IDPs as training samples for artificial intelligence-based biomarker development and for normative modeling to help identify injury-induced changes as outliers for precision diagnosis and targeted therapeutic development. On its release, NNL is poised to support the use of advanced imaging in clinician decision support tools, the validation of imaging biomarkers, and the investigation of brain-behavior anomalies, moving the field toward precision medicine.
Collapse
Affiliation(s)
| | - James R Stone
- Department of Radiology and Medical Imaging, University of Virginia, Charlottesville, Virginia, USA
| | - Elisabeth A Wilde
- George E. Wahlen VA, Salt Lake City Healthcare System, Salt Lake City, Utah, USA
| | - Stephen R McCauley
- Department of Neurology, Baylor College of Medicine, Houston, Texas, USA
| | - Robert C Welsh
- Department of Psychiatry and Biobehavioral Sciences, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California, USA
| | - John P Mugler
- Department of Radiology and Medical Imaging, University of Virginia, Charlottesville, Virginia, USA
| | - Nick Tustison
- Department of Radiology and Medical Imaging, University of Virginia, Charlottesville, Virginia, USA
| | - Brian Avants
- Department of Radiology and Medical Imaging, University of Virginia, Charlottesville, Virginia, USA
| | - Christopher T Whitlow
- Department of Radiology, Wake Forest University School of Medicine, Winston-Salem, North Carolina, USA
| | | | | | - Magali Haas
- Cohen Veterans Bioscience, New York, New York, USA
| |
Collapse
|
15
|
Melendez J, Sung YJ, Orr M, Yoo A, Schindler S, Cruchaga C, Bateman R. An interpretable machine learning-based cerebrospinal fluid proteomics clock for predicting age reveals novel insights into brain aging. Aging Cell 2024; 23:e14230. [PMID: 38923730 PMCID: PMC11488306 DOI: 10.1111/acel.14230] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2024] [Revised: 05/10/2024] [Accepted: 05/15/2024] [Indexed: 06/28/2024] Open
Abstract
Machine learning can be used to create "biologic clocks" that predict age. However, organs, tissues, and biofluids may age at different rates from the organism as a whole. We sought to understand how cerebrospinal fluid (CSF) changes with age to inform the development of brain aging-related disease mechanisms and identify potential anti-aging therapeutic targets. Several epigenetic clocks exist based on plasma and neuronal tissues; however, plasma may not reflect brain aging specifically and tissue-based clocks require samples that are difficult to obtain from living participants. To address these problems, we developed a machine learning clock that uses CSF proteomics to predict the chronological age of individuals with a 0.79 Pearson correlation and mean estimated error (MAE) of 4.30 years in our validation cohort. Additionally, we analyzed proteins highly weighted by the algorithm to gain insights into changes in CSF and uncover novel insights into brain aging. We also demonstrate a novel method to create a minimal protein clock that uses just 109 protein features from the original clock to achieve a similar accuracy (0.75 correlation, MAE 5.41). Finally, we demonstrate that our clock identifies novel proteins that are highly predictive of age in interactions with other proteins, but do not directly correlate with chronological age themselves. In conclusion, we propose that our CSF protein aging clock can identify novel proteins that influence the rate of aging of the central nervous system (CNS), in a manner that would not be identifiable by examining their individual relationships with age.
Collapse
Affiliation(s)
- Justin Melendez
- Tracy Family SILQ CenterWashington University in St. LouisSt. LouisMissouriUSA
- Department of NeurologyWashington University in St. LouisSt. LouisMissouriUSA
| | - Yun Ju Sung
- Department of PsychiatryWashington University in St. LouisSt. LouisMissouriUSA
- Department of BiostatisticsWashington University in St. LouisSt. LouisMissouriUSA
| | - Miranda Orr
- Department of Internal MedicineWake Forest School of Medicine Section of Gerontology and Geriatric Medicine Medical Center BoulevardWinston‐SalemNorth CarolinaUSA
| | - Andrew Yoo
- Department of Developmental BiologyWashington University in St. LouisSt. LouisMissouriUSA
| | - Suzanne Schindler
- Department of NeurologyWashington University in St. LouisSt. LouisMissouriUSA
| | - Carlos Cruchaga
- Department of NeurologyWashington University in St. LouisSt. LouisMissouriUSA
- Department of PsychiatryWashington University in St. LouisSt. LouisMissouriUSA
| | - Randall Bateman
- Tracy Family SILQ CenterWashington University in St. LouisSt. LouisMissouriUSA
- Department of NeurologyWashington University in St. LouisSt. LouisMissouriUSA
| |
Collapse
|
16
|
Gustavson DE, Elman JA, Reynolds CA, Eyler LT, Fennema-Notestine C, Puckett OK, Panizzon MS, Gillespie NA, Neale MC, Lyons MJ, Franz CE, Kremen WS. Brain reserve in midlife is associated with executive function changes across 12 years. Neurobiol Aging 2024; 141:113-120. [PMID: 38852544 PMCID: PMC11246793 DOI: 10.1016/j.neurobiolaging.2024.05.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Revised: 04/17/2024] [Accepted: 05/08/2024] [Indexed: 06/11/2024]
Abstract
We examined how brain reserve in midlife, measured by brain-predicted age difference scores (Brain-PADs), predicted executive function concurrently and longitudinally into early old age, and whether these associations were moderated by young adult cognitive reserve or APOE genotype. 508 men in the Vietnam Era Twin Study of Aging (VETSA) completed neuroimaging assessments at mean age 56 and six executive function tasks at mean ages 56, 62, and 68 years. Results indicated that greater brain reserve at age 56 was associated with better concurrent executive function (r=.10, p=.040) and less decline in executive function over 12 years (r=.34, p=.001). These associations were not moderated by cognitive reserve or APOE genotype. Twin analysis suggested associations with executive function slopes were driven by genetic influences. Our findings suggest that greater brain reserve allowed for better cognitive maintenance from middle- to old age, driven by a genetic association. The results are consistent with differential preservation of executive function based on brain reserve that is independent of young adult cognitive reserve or APOE genotype.
Collapse
Affiliation(s)
- Daniel E Gustavson
- Institute for Behavioral Genetics, University of Colorado Boulder, Boulder, CO, USA.
| | - Jeremy A Elman
- Department of Psychiatry, University of California San Diego, La Jolla, CA, USA; Center for Behavior Genetics of Aging, University of California San Diego, La Jolla, CA, USA
| | - Chandra A Reynolds
- Institute for Behavioral Genetics, University of Colorado Boulder, Boulder, CO, USA
| | - Lisa T Eyler
- Department of Psychiatry, University of California San Diego, La Jolla, CA, USA; Center for Behavior Genetics of Aging, University of California San Diego, La Jolla, CA, USA
| | - Christine Fennema-Notestine
- Department of Psychiatry, University of California San Diego, La Jolla, CA, USA; Center for Behavior Genetics of Aging, University of California San Diego, La Jolla, CA, USA; Department of Radiology, University of California San Diego, La Jolla, CA, USA
| | - Olivia K Puckett
- Center for Behavior Genetics of Aging, University of California San Diego, La Jolla, CA, USA; Department of Radiology, University of California San Diego, La Jolla, CA, USA
| | - Matthew S Panizzon
- Department of Psychiatry, University of California San Diego, La Jolla, CA, USA; Center for Behavior Genetics of Aging, University of California San Diego, La Jolla, CA, USA
| | - Nathan A Gillespie
- Virginia Institute for Psychiatric and Behavior Genetics, Virginia Commonwealth University, Richmond, VA, USA
| | - Michael C Neale
- Virginia Institute for Psychiatric and Behavior Genetics, Virginia Commonwealth University, Richmond, VA, USA
| | - Michael J Lyons
- Department of Psychological and Brain Sciences, Boston University, Boston, MA, USA
| | - Carol E Franz
- Department of Psychiatry, University of California San Diego, La Jolla, CA, USA; Center for Behavior Genetics of Aging, University of California San Diego, La Jolla, CA, USA
| | - William S Kremen
- Department of Psychiatry, University of California San Diego, La Jolla, CA, USA; Center for Behavior Genetics of Aging, University of California San Diego, La Jolla, CA, USA
| |
Collapse
|
17
|
Karoly HC, Kirk‐Provencher KT, Schacht JP, Gowin JL. Alcohol and brain structure across the lifespan: A systematic review of large-scale neuroimaging studies. Addict Biol 2024; 29:e13439. [PMID: 39317645 PMCID: PMC11421948 DOI: 10.1111/adb.13439] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2024] [Revised: 08/29/2024] [Accepted: 09/01/2024] [Indexed: 09/26/2024]
Abstract
Alcohol exposure affects brain structure, but the extent to which its effects differ across development remains unclear. Several countries are considering changes to recommended guidelines for alcohol consumption, so high-quality evidence is needed. Many studies have been conducted among small samples, but recent efforts have been made to acquire large samples to characterize alcohol's effects on the brain on a population level. Several large-scale consortia have acquired such samples, but this evidence has not been synthesized across the lifespan. We conducted a systematic review of large-scale neuroimaging studies examining effects of alcohol exposure on brain structure at multiple developmental stages. We included studies with an alcohol-exposed sample of at least N = 100 from the following consortia: ABCD, ENIGMA, NCANDA, IMAGEN, Framingham Offspring Study, HCP and UK BioBank. Twenty-seven studies were included, examining prenatal (N = 1), adolescent (N = 9), low-to-moderate-level adult (N = 11) and heavy adult (N = 7) exposure. Prenatal exposure was associated with greater brain volume at ages 9-10, but contemporaneous alcohol consumption during adolescence and adulthood was associated with smaller volume/thickness. Both low-to-moderate consumption and heavy consumption were characterized by smaller volume and thickness in frontal, temporal and parietal regions, and reductions in insula, cingulate and subcortical structures. Adolescent consumption had similar effects, with less consistent evidence for smaller cingulate, insula and subcortical volume. In sum, prenatal exposure was associated with larger volume, while adolescent and adult alcohol exposure was associated with smaller volume and thickness, suggesting that regional patterns of effects of alcohol are similar in adolescence and adulthood.
Collapse
Affiliation(s)
- Hollis C. Karoly
- Department of PsychologyColorado State UniversityFort CollinsColoradoUSA
| | - Katelyn T. Kirk‐Provencher
- Department of Radiology, School of MedicineUniversity of Colorado Anschutz Medical CampusAuroraColoradoUSA
| | - Joseph P. Schacht
- Department of Psychiatry, School of MedicineUniversity of Colorado Anschutz Medical CampusAuroraColoradoUSA
| | - Joshua L. Gowin
- Department of Radiology, School of MedicineUniversity of Colorado Anschutz Medical CampusAuroraColoradoUSA
| |
Collapse
|
18
|
Hua JPY, Abram SV, Loewy RL, Stuart B, Fryer SL, Vinogradov S, Mathalon DH. Brain Age Gap in Early Illness Schizophrenia and the Clinical High-Risk Syndrome: Associations With Experiential Negative Symptoms and Conversion to Psychosis. Schizophr Bull 2024; 50:1159-1170. [PMID: 38815987 PMCID: PMC11349027 DOI: 10.1093/schbul/sbae074] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 06/01/2024]
Abstract
BACKGROUND AND HYPOTHESIS Brain development/aging is not uniform across individuals, spawning efforts to characterize brain age from a biological perspective to model the effects of disease and maladaptive life processes on the brain. The brain age gap represents the discrepancy between estimated brain biological age and chronological age (in this case, based on structural magnetic resonance imaging, MRI). Structural MRI studies report an increased brain age gap (biological age > chronological age) in schizophrenia, with a greater brain age gap related to greater negative symptom severity. Less is known regarding the nature of this gap early in schizophrenia (ESZ), if this gap represents a psychosis conversion biomarker in clinical high-risk (CHR-P) individuals, and how altered brain development and/or aging map onto specific symptom facets. STUDY DESIGN Using structural MRI, we compared the brain age gap among CHR-P (n = 51), ESZ (n = 78), and unaffected comparison participants (UCP; n = 90), and examined associations with CHR-P psychosis conversion (CHR-P converters n = 10; CHR-P non-converters; n = 23) and positive and negative symptoms. STUDY RESULTS ESZ showed a greater brain age gap relative to UCP and CHR-P (Ps < .010). CHR-P individuals who converted to psychosis showed a greater brain age gap (P = .043) relative to CHR-P non-converters. A larger brain age gap in ESZ was associated with increased experiential (P = .008), but not expressive negative symptom severity. CONCLUSIONS Consistent with schizophrenia pathophysiological models positing abnormal brain maturation, results suggest abnormal brain development is present early in psychosis. An increased brain age gap may be especially relevant to motivational and functional deficits in schizophrenia.
Collapse
Affiliation(s)
- Jessica P Y Hua
- Sierra Pacific Mental Illness Research Education and Clinical Centers, San Francisco VA Medical Center, University of California, San Francisco, CA, USA
- Mental Health Service, San Francisco VA Health Care System, San Francisco, CA, USA
- Department of Psychiatry and Behavioral Sciences, University of California San Francisco, San Francisco, CA, USA
| | - Samantha V Abram
- Mental Health Service, San Francisco VA Health Care System, San Francisco, CA, USA
- Department of Psychiatry and Behavioral Sciences, University of California San Francisco, San Francisco, CA, USA
| | - Rachel L Loewy
- Department of Psychiatry and Behavioral Sciences, University of California San Francisco, San Francisco, CA, USA
| | - Barbara Stuart
- Department of Psychiatry and Behavioral Sciences, University of California San Francisco, San Francisco, CA, USA
| | - Susanna L Fryer
- Mental Health Service, San Francisco VA Health Care System, San Francisco, CA, USA
- Department of Psychiatry and Behavioral Sciences, University of California San Francisco, San Francisco, CA, USA
| | - Sophia Vinogradov
- Department of Psychiatry and Behavioral Sciences, University of Minnesota, Minneapolis, MN, USA
| | - Daniel H Mathalon
- Mental Health Service, San Francisco VA Health Care System, San Francisco, CA, USA
- Department of Psychiatry and Behavioral Sciences, University of California San Francisco, San Francisco, CA, USA
| |
Collapse
|
19
|
Klempíř O, Krupička R. Analyzing Wav2Vec 1.0 Embeddings for Cross-Database Parkinson's Disease Detection and Speech Features Extraction. SENSORS (BASEL, SWITZERLAND) 2024; 24:5520. [PMID: 39275431 PMCID: PMC11398018 DOI: 10.3390/s24175520] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Revised: 08/22/2024] [Accepted: 08/24/2024] [Indexed: 09/16/2024]
Abstract
Advancements in deep learning speech representations have facilitated the effective use of extensive unlabeled speech datasets for Parkinson's disease (PD) modeling with minimal annotated data. This study employs the non-fine-tuned wav2vec 1.0 architecture to develop machine learning models for PD speech diagnosis tasks, such as cross-database classification and regression to predict demographic and articulation characteristics. The primary aim is to analyze overlapping components within the embeddings on both classification and regression tasks, investigating whether latent speech representations in PD are shared across models, particularly for related tasks. Firstly, evaluation using three multi-language PD datasets showed that wav2vec accurately detected PD based on speech, outperforming feature extraction using mel-frequency cepstral coefficients in the proposed cross-database classification scenarios. In cross-database scenarios using Italian and English-read texts, wav2vec demonstrated performance comparable to intra-dataset evaluations. We also compared our cross-database findings against those of other related studies. Secondly, wav2vec proved effective in regression, modeling various quantitative speech characteristics related to articulation and aging. Ultimately, subsequent analysis of important features examined the presence of significant overlaps between classification and regression models. The feature importance experiments discovered shared features across trained models, with increased sharing for related tasks, further suggesting that wav2vec contributes to improved generalizability. The study proposes wav2vec embeddings as a next promising step toward a speech-based universal model to assist in the evaluation of PD.
Collapse
Affiliation(s)
| | - Radim Krupička
- Department of Biomedical Informatics, Faculty of Biomedical Engineering, Czech Technical University in Prague, 16000 Prague, Czech Republic;
| |
Collapse
|
20
|
Zhang X, Pan Y, Wu T, Zhao W, Zhang H, Ding J, Ji Q, Jia X, Li X, Lee Z, Zhang J, Bai L. Brain age prediction using interpretable multi-feature-based convolutional neural network in mild traumatic brain injury. Neuroimage 2024; 297:120751. [PMID: 39048043 DOI: 10.1016/j.neuroimage.2024.120751] [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: 04/20/2024] [Revised: 07/15/2024] [Accepted: 07/22/2024] [Indexed: 07/27/2024] Open
Abstract
BACKGROUND Convolutional neural network (CNN) can capture the structural features changes of brain aging based on MRI, thus predict brain age in healthy individuals accurately. However, most studies use single feature to predict brain age in healthy individuals, ignoring adding information from multiple sources and the changes in brain aging patterns after mild traumatic brain injury (mTBI) were still unclear. METHODS Here, we leveraged the structural data from a large, heterogeneous dataset (N = 1464) to implement an interpretable 3D combined CNN model for brain-age prediction. In addition, we also built an atlas-based occlusion analysis scheme with a fine-grained human Brainnetome Atlas to reveal the age-sstratified contributed brain regions for brain-age prediction in healthy controls (HCs) and mTBI patients. The correlations between brain predicted age gaps (brain-PAG) following mTBI and individual's cognitive impairment, as well as the level of plasma neurofilament light were also examined. RESULTS Our model utilized multiple 3D features derived from T1w data as inputs, and reduced the mean absolute error (MAE) of age prediction to 3.08 years and improved Pearson's r to 0.97 on 154 HCs. The strong generalizability of our model was also validated across different centers. Regions contributing the most significantly to brain age prediction were the caudate and thalamus for HCs and patients with mTBI, and the contributive regions were mostly located in the subcortical areas throughout the adult lifespan. The left hemisphere was confirmed to contribute more in brain age prediction throughout the adult lifespan. Our research showed that brain-PAG in mTBI patients was significantly higher than that in HCs in both acute and chronic phases. The increased brain-PAG in mTBI patients was also highly correlated with cognitive impairment and a higher level of plasma neurofilament light, a marker of neurodegeneration. The higher brain-PAG and its correlation with severe cognitive impairment showed a longitudinal and persistent nature in patients with follow-up examinations. CONCLUSION We proposed an interpretable deep learning framework on a relatively large dataset to accurately predict brain age in both healthy individuals and mTBI patients. The interpretable analysis revealed that the caudate and thalamus became the most contributive role across the adult lifespan in both HCs and patients with mTBI. The left hemisphere contributed significantly to brain age prediction may enlighten us to be concerned about the lateralization of brain abnormality in neurological diseases in the future. The proposed interpretable deep learning framework might also provide hope for testing the performance of related drugs and treatments in the future.
Collapse
Affiliation(s)
- Xiang Zhang
- The Key Laboratory of Biomedical Information Engineering, Ministry of Education, Department of Biomedical Engineering, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an 710049, China
| | - Yizhen Pan
- The Key Laboratory of Biomedical Information Engineering, Ministry of Education, Department of Biomedical Engineering, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an 710049, China
| | - Tingting Wu
- The Key Laboratory of Biomedical Information Engineering, Ministry of Education, Department of Biomedical Engineering, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an 710049, China
| | - Wenpu Zhao
- The Key Laboratory of Biomedical Information Engineering, Ministry of Education, Department of Biomedical Engineering, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an 710049, China
| | - Haonan Zhang
- The Key Laboratory of Biomedical Information Engineering, Ministry of Education, Department of Biomedical Engineering, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an 710049, China
| | - Jierui Ding
- The Key Laboratory of Biomedical Information Engineering, Ministry of Education, Department of Biomedical Engineering, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an 710049, China
| | - Qiuyu Ji
- The Key Laboratory of Biomedical Information Engineering, Ministry of Education, Department of Biomedical Engineering, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an 710049, China
| | - Xiaoyan Jia
- The Key Laboratory of Biomedical Information Engineering, Ministry of Education, Department of Biomedical Engineering, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an 710049, China
| | - Xuan Li
- The Key Laboratory of Biomedical Information Engineering, Ministry of Education, Department of Biomedical Engineering, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an 710049, China
| | - Zhiqi Lee
- The Key Laboratory of Biomedical Information Engineering, Ministry of Education, Department of Biomedical Engineering, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an 710049, China
| | - Jie Zhang
- Department of Radiation Medicine, School of Preventive Medicine, Air Force Medical University, Xi'an 710032, China.
| | - Lijun Bai
- The Key Laboratory of Biomedical Information Engineering, Ministry of Education, Department of Biomedical Engineering, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an 710049, China.
| |
Collapse
|
21
|
Wu Y, Gao H, Zhang C, Ma X, Zhu X, Wu S, Lin L. Machine Learning and Deep Learning Approaches in Lifespan Brain Age Prediction: A Comprehensive Review. Tomography 2024; 10:1238-1262. [PMID: 39195728 PMCID: PMC11359833 DOI: 10.3390/tomography10080093] [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: 07/18/2024] [Revised: 08/09/2024] [Accepted: 08/09/2024] [Indexed: 08/29/2024] Open
Abstract
The concept of 'brain age', derived from neuroimaging data, serves as a crucial biomarker reflecting cognitive vitality and neurodegenerative trajectories. In the past decade, machine learning (ML) and deep learning (DL) integration has transformed the field, providing advanced models for brain age estimation. However, achieving precise brain age prediction across all ages remains a significant analytical challenge. This comprehensive review scrutinizes advancements in ML- and DL-based brain age prediction, analyzing 52 peer-reviewed studies from 2020 to 2024. It assesses various model architectures, highlighting their effectiveness and nuances in lifespan brain age studies. By comparing ML and DL, strengths in forecasting and methodological limitations are revealed. Finally, key findings from the reviewed articles are summarized and a number of major issues related to ML/DL-based lifespan brain age prediction are discussed. Through this study, we aim at the synthesis of the current state of brain age prediction, emphasizing both advancements and persistent challenges, guiding future research, technological advancements, and improving early intervention strategies for neurodegenerative diseases.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Lan Lin
- Department of Biomedical Engineering, College of Chemistry and Life Science, Beijing University of Technology, Beijing 100124, China; (Y.W.); (H.G.); (C.Z.); (X.M.); (X.Z.); (S.W.)
| |
Collapse
|
22
|
Kapogiannis D, Manolopoulos A, Mullins R, Avgerinos K, Delgado-Peraza F, Mustapic M, Nogueras-Ortiz C, Yao PJ, Pucha KA, Brooks J, Chen Q, Haas SS, Ge R, Hartnell LM, Cookson MR, Egan JM, Frangou S, Mattson MP. Brain responses to intermittent fasting and the healthy living diet in older adults. Cell Metab 2024; 36:1668-1678.e5. [PMID: 38901423 PMCID: PMC11305918 DOI: 10.1016/j.cmet.2024.05.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Revised: 03/29/2024] [Accepted: 05/29/2024] [Indexed: 06/22/2024]
Abstract
Diet may promote brain health in metabolically impaired older individuals. In an 8-week randomized clinical trial involving 40 cognitively intact older adults with insulin resistance, we examined the effects of 5:2 intermittent fasting and the healthy living diet on brain health. Although intermittent fasting induced greater weight loss, the two diets had comparable effects in improving insulin signaling biomarkers in neuron-derived extracellular vesicles, decreasing the brain-age-gap estimate (reflecting the pace of biological aging of the brain) on magnetic resonance imaging, reducing brain glucose on magnetic resonance spectroscopy, and improving blood biomarkers of carbohydrate and lipid metabolism, with minimal changes in cerebrospinal fluid biomarkers for Alzheimer's disease. Intermittent fasting and healthy living improved executive function and memory, with intermittent fasting benefiting more certain cognitive measures. In exploratory analyses, sex, body mass index, and apolipoprotein E and SLC16A7 genotypes modulated diet effects. The study provides a blueprint for assessing brain effects of dietary interventions and motivates further research on intermittent fasting and continuous diets for brain health optimization. For further information, please see ClinicalTrials.gov registration: NCT02460783.
Collapse
Affiliation(s)
- Dimitrios Kapogiannis
- Intramural Research Program, Laboratory of Clinical Investigation, National Institute on Aging, Baltimore, MD, USA; Department of Neurology, Johns Hopkins University, Baltimore, MD, USA.
| | - Apostolos Manolopoulos
- Intramural Research Program, Laboratory of Clinical Investigation, National Institute on Aging, Baltimore, MD, USA
| | - Roger Mullins
- Morgan State University, Core Lab, Baltimore, MD, USA
| | | | - Francheska Delgado-Peraza
- Intramural Research Program, Laboratory of Clinical Investigation, National Institute on Aging, Baltimore, MD, USA
| | - Maja Mustapic
- Intramural Research Program, Laboratory of Clinical Investigation, National Institute on Aging, Baltimore, MD, USA
| | - Carlos Nogueras-Ortiz
- Intramural Research Program, Laboratory of Clinical Investigation, National Institute on Aging, Baltimore, MD, USA
| | - Pamela J Yao
- Intramural Research Program, Laboratory of Clinical Investigation, National Institute on Aging, Baltimore, MD, USA
| | - Krishna A Pucha
- Intramural Research Program, Laboratory of Clinical Investigation, National Institute on Aging, Baltimore, MD, USA
| | - Janet Brooks
- Intramural Research Program, Laboratory of Neurogenetics, National Institute on Aging, Bethesda, MD, USA
| | - Qinghua Chen
- Intramural Research Program, Laboratory of Clinical Investigation, National Institute on Aging, Baltimore, MD, USA
| | - Shalaila S Haas
- Mt. Sinai School of Medicine, Department of Psychiatry, New York, NY, USA
| | - Ruiyang Ge
- Center for Brain Health, University of British Columbia, Vancouver, BC, Canada
| | - Lisa M Hartnell
- Intramural Research Program, Translational Gerontology Branch, National Institute on Aging, Baltimore, MD, USA
| | - Mark R Cookson
- Intramural Research Program, Laboratory of Neurogenetics, National Institute on Aging, Bethesda, MD, USA
| | - Josephine M Egan
- Intramural Research Program, Laboratory of Clinical Investigation, National Institute on Aging, Baltimore, MD, USA
| | - Sophia Frangou
- Mt. Sinai School of Medicine, Department of Psychiatry, New York, NY, USA; Center for Brain Health, University of British Columbia, Vancouver, BC, Canada
| | - Mark P Mattson
- Department of Neuroscience, Johns Hopkins University, Baltimore, MD, USA
| |
Collapse
|
23
|
Casanova R, Walker KA, Justice JN, Anderson A, Duggan MR, Cordon J, Barnard RT, Lu L, Hsu FC, Sedaghat S, Prizment A, Kritchevsky SB, Wagenknecht LE, Hughes TM. Associations of plasma proteomics and age-related outcomes with brain age in a diverse cohort. GeroScience 2024; 46:3861-3873. [PMID: 38438772 PMCID: PMC11226584 DOI: 10.1007/s11357-024-01112-4] [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: 12/07/2023] [Accepted: 02/26/2024] [Indexed: 03/06/2024] Open
Abstract
Machine learning models are increasingly being used to estimate "brain age" from neuroimaging data. The gap between chronological age and the estimated brain age gap (BAG) is potentially a measure of accelerated and resilient brain aging. Brain age calculated in this fashion has been shown to be associated with mortality, measures of physical function, health, and disease. Here, we estimate the BAG using a voxel-based elastic net regression approach, and then, we investigate its associations with mortality, cognitive status, and measures of health and disease in participants from Atherosclerosis Risk in Communities (ARIC) study who had a brain MRI at visit 5 of the study. Finally, we used the SOMAscan assay containing 4877 proteins to examine the proteomic associations with the MRI-defined BAG. Among N = 1849 participants (age, 76.4 (SD 5.6)), we found that increased values of BAG were strongly associated with increased mortality and increased severity of the cognitive status. Strong associations with mortality persisted when the analyses were performed in cognitively normal participants. In addition, it was strongly associated with BMI, diabetes, measures of physical function, hypertension, prevalent heart disease, and stroke. Finally, we found 33 proteins associated with BAG after a correction for multiple comparisons. The top proteins with positive associations to brain age were growth/differentiation factor 15 (GDF-15), Sushi, von Willebrand factor type A, EGF, and pentraxin domain-containing protein 1 (SEVP 1), matrilysin (MMP7), ADAMTS-like protein 2 (ADAMTS), and heat shock 70 kDa protein 1B (HSPA1B) while EGF-receptor (EGFR), mast/stem-cell-growth-factor-receptor (KIT), coagulation-factor-VII, and cGMP-dependent-protein-kinase-1 (PRKG1) were negatively associated to brain age. Several of these proteins were previously associated with dementia in ARIC. These results suggest that circulating proteins implicated in biological aging, cellular senescence, angiogenesis, and coagulation are associated with a neuroimaging measure of brain aging.
Collapse
Affiliation(s)
- Ramon Casanova
- Department of Biostatistics and Data Science, Wake Forest University School of Medicine, Medical Center Blvd., Winston-Salem, NC, USA.
| | | | - Jamie N Justice
- Department of Internal Medicine, Wake Forest University School of Medicine, Winston-Salem, NC, USA
| | - Andrea Anderson
- Department of Biostatistics and Data Science, Wake Forest University School of Medicine, Medical Center Blvd., Winston-Salem, NC, USA
| | | | | | - Ryan T Barnard
- Department of Biostatistics and Data Science, Wake Forest University School of Medicine, Medical Center Blvd., Winston-Salem, NC, USA
| | - Lingyi Lu
- Department of Biostatistics and Data Science, Wake Forest University School of Medicine, Medical Center Blvd., Winston-Salem, NC, USA
| | - Fang-Chi Hsu
- Department of Biostatistics and Data Science, Wake Forest University School of Medicine, Medical Center Blvd., Winston-Salem, NC, USA
| | - Sanaz Sedaghat
- School of Public Health, Oncology and Transplantation, University of Minnesota, Minneapolis, MN, USA
| | - Anna Prizment
- Division of Epidemiology and Community Health, School of Public Health, University of Minnesota, Minneapolis, MN, USA
| | - Stephen B Kritchevsky
- Department of Internal Medicine, Wake Forest University School of Medicine, Winston-Salem, NC, USA
| | - Lynne E Wagenknecht
- Division of Public Health Sciences, Wake Forest University School of Medicine, Winston-Salem, NC, USA
| | - Timothy M Hughes
- Department of Internal Medicine, Wake Forest University School of Medicine, Winston-Salem, NC, USA
| |
Collapse
|
24
|
Ng C, Huang P, Cho Y, Lee P, Liu Y, Chang T. Frontoparietal and salience network synchronizations during nonsymbolic magnitude processing predict brain age and mathematical performance in youth. Hum Brain Mapp 2024; 45:e26777. [PMID: 39046114 PMCID: PMC11267564 DOI: 10.1002/hbm.26777] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2023] [Revised: 06/03/2024] [Accepted: 06/19/2024] [Indexed: 07/25/2024] Open
Abstract
The development and refinement of functional brain circuits crucial to human cognition is a continuous process that spans from childhood to adulthood. Research increasingly focuses on mapping these evolving configurations, with the aim to identify markers for functional impairments and atypical development. Among human cognitive systems, nonsymbolic magnitude representations serve as a foundational building block for future success in mathematical learning and achievement for individuals. Using task-based frontoparietal (FPN) and salience network (SN) features during nonsymbolic magnitude processing alongside machine learning algorithms, we developed a framework to construct brain age prediction models for participants aged 7-30. Our study revealed differential developmental profiles in the synchronization within and between FPN and SN networks. Specifically, we observed a linear increase in FPN connectivity, concomitant with a decline in SN connectivity across the age span. A nonlinear U-shaped trajectory in the connectivity between the FPN and SN was discerned, revealing reduced FPN-SN synchronization among adolescents compared to both pediatric and adult cohorts. Leveraging the Gradient Boosting machine learning algorithm and nested fivefold stratified cross-validation with independent training datasets, we demonstrated that functional connectivity measures of the FPN and SN nodes predict chronological age, with a correlation coefficient of .727 and a mean absolute error of 2.944 between actual and predicted ages. Notably, connectivity within the FPN emerged as the most contributing feature for age prediction. Critically, a more matured brain age estimate is associated with better arithmetic performance. Our findings shed light on the intricate developmental changes occurring in the neural networks supporting magnitude representations. We emphasize brain age estimation as a potent tool for understanding cognitive development and its relationship to mathematical abilities across the critical developmental period of youth. PRACTITIONER POINTS: This study investigated the prolonged changes in the brain's architecture across childhood, adolescence, and adulthood, with a focus on task-state frontoparietal and salience networks. Distinct developmental pathways were identified: frontoparietal synchronization strengthens consistently throughout development, while salience network connectivity diminishes with age. Furthermore, adolescents show a unique dip in connectivity between these networks. Leveraging advanced machine learning methods, we accurately predicted individuals' ages based on these brain circuits, with a more mature estimated brain age correlating with better math skills.
Collapse
Affiliation(s)
- Chan‐Tat Ng
- Department of PsychologyNational Chengchi UniversityTaipeiTaiwan
| | - Po‐Hsien Huang
- Department of PsychologyNational Chengchi UniversityTaipeiTaiwan
- Research Center for Mind, Brain & LearningNational Chengchi UniversityTaipeiTaiwan
| | - Yi‐Cheng Cho
- Department of PsychologyNational Chengchi UniversityTaipeiTaiwan
| | - Pei‐Hong Lee
- Research Center for Mind, Brain & LearningNational Chengchi UniversityTaipeiTaiwan
| | - Yi‐Chang Liu
- Research Center for Mind, Brain & LearningNational Chengchi UniversityTaipeiTaiwan
| | - Ting‐Ting Chang
- Department of PsychologyNational Chengchi UniversityTaipeiTaiwan
- Research Center for Mind, Brain & LearningNational Chengchi UniversityTaipeiTaiwan
| |
Collapse
|
25
|
Kim WS, Heo DW, Maeng J, Shen J, Tsogt U, Odkhuu S, Zhang X, Cheraghi S, Kim SW, Ham BJ, Rami FZ, Sui J, Kang CY, Suk HI, Chung YC. Deep Learning-based Brain Age Prediction in Patients With Schizophrenia Spectrum Disorders. Schizophr Bull 2024; 50:804-814. [PMID: 38085061 PMCID: PMC11283195 DOI: 10.1093/schbul/sbad167] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 07/29/2024]
Abstract
BACKGROUND AND HYPOTHESIS The brain-predicted age difference (brain-PAD) may serve as a biomarker for neurodegeneration. We investigated the brain-PAD in patients with schizophrenia (SCZ), first-episode schizophrenia spectrum disorders (FE-SSDs), and treatment-resistant schizophrenia (TRS) using structural magnetic resonance imaging (sMRI). STUDY DESIGN We employed a convolutional network-based regression (SFCNR), and compared its performance with models based on three machine learning (ML) algorithms. We pretrained the SFCNR with sMRI data of 7590 healthy controls (HCs) selected from the UK Biobank. The parameters of the pretrained model were transferred to the next training phase with a new set of HCs (n = 541). The brain-PAD was analyzed in independent HCs (n = 209) and patients (n = 233). Correlations between the brain-PAD and clinical measures were investigated. STUDY RESULTS The SFCNR model outperformed three commonly used ML models. Advanced brain aging was observed in patients with SCZ, FE-SSDs, and TRS compared to HCs. A significant difference in brain-PAD was observed between FE-SSDs and TRS with ridge regression but not with the SFCNR model. Chlorpromazine equivalent dose and cognitive function were correlated with the brain-PAD in SCZ and FE-SSDs. CONCLUSIONS Our findings indicate that there is advanced brain aging in patients with SCZ and higher brain-PAD in SCZ can be used as a surrogate marker for cognitive dysfunction. These findings warrant further investigations on the causes of advanced brain age in SCZ. In addition, possible psychosocial and pharmacological interventions targeting brain health should be considered in early-stage SCZ patients with advanced brain age.
Collapse
Affiliation(s)
- Woo-Sung Kim
- Department of Psychiatry, Jeonbuk National University, Medical School, Jeonju, Korea
- Research Institute of Clinical Medicine of Jeonbuk National University-Biomedical Research Institute of Jeonbuk National University Hospital, Jeonju, Korea
| | - Da-Woon Heo
- Department of Artificial Intelligence, Korea University, Seoul, Korea
| | - Junyeong Maeng
- Department of Artificial Intelligence, Korea University, Seoul, Korea
| | - Jie Shen
- Department of Psychiatry, Jeonbuk National University, Medical School, Jeonju, Korea
- Research Institute of Clinical Medicine of Jeonbuk National University-Biomedical Research Institute of Jeonbuk National University Hospital, Jeonju, Korea
- Department of Psychiatry, Yanbian University, Medical School, Yanji, China
| | - Uyanga Tsogt
- Department of Psychiatry, Jeonbuk National University, Medical School, Jeonju, Korea
- Research Institute of Clinical Medicine of Jeonbuk National University-Biomedical Research Institute of Jeonbuk National University Hospital, Jeonju, Korea
| | - Soyolsaikhan Odkhuu
- Department of Psychiatry, Jeonbuk National University, Medical School, Jeonju, Korea
- Research Institute of Clinical Medicine of Jeonbuk National University-Biomedical Research Institute of Jeonbuk National University Hospital, Jeonju, Korea
| | - Xuefeng Zhang
- Research Institute of Clinical Medicine of Jeonbuk National University-Biomedical Research Institute of Jeonbuk National University Hospital, Jeonju, Korea
| | - Sahar Cheraghi
- Department of Psychiatry, Jeonbuk National University, Medical School, Jeonju, Korea
- Research Institute of Clinical Medicine of Jeonbuk National University-Biomedical Research Institute of Jeonbuk National University Hospital, Jeonju, Korea
| | - Sung-Wan Kim
- Department of Psychiatry, Chonnam National University Medical School, Gwangju, Korea
| | - Byung-Joo Ham
- Department of Psychiatry, Korea University Anam Hospital, Korea University College of Medicine, Seoul, Korea
| | - Fatima Zahra Rami
- Department of Psychiatry, Jeonbuk National University, Medical School, Jeonju, Korea
- Research Institute of Clinical Medicine of Jeonbuk National University-Biomedical Research Institute of Jeonbuk National University Hospital, Jeonju, Korea
| | - Jing Sui
- Tri-Institutional Center for Translational Research in Neuroimaging and Data Science (TReNDS), Georgia State University, Georgia Institute of Technology, Emory University, Atlanta, GA, USA
- State Key Laboratory of Cognitive Neuroscience and Learning, Beijing Normal University, Beijing, China
| | - Chae Yeong Kang
- Research Institute of Clinical Medicine of Jeonbuk National University-Biomedical Research Institute of Jeonbuk National University Hospital, Jeonju, Korea
| | - Heung-Il Suk
- Department of Artificial Intelligence, Korea University, Seoul, Korea
- Department of Brain and Cognitive Engineering, Korea University, Seoul, Korea
| | - Young-Chul Chung
- Department of Psychiatry, Jeonbuk National University, Medical School, Jeonju, Korea
- Department of Psychiatry, Jeonbuk National University Hospital, Jeonju, Korea
- Research Institute of Clinical Medicine of Jeonbuk National University-Biomedical Research Institute of Jeonbuk National University Hospital, Jeonju, Korea
| |
Collapse
|
26
|
Huang H, Wang J, Dunk MM, Guo J, Dove A, Ma J, Bennett DA, Xu W. Association of Cardiovascular Health With Brain Age Estimated Using Machine Learning Methods in Middle-Aged and Older Adults. Neurology 2024; 103:e209530. [PMID: 38889383 PMCID: PMC11226327 DOI: 10.1212/wnl.0000000000209530] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Accepted: 04/05/2024] [Indexed: 06/20/2024] Open
Abstract
BACKGROUND AND OBJECTIVES Cardiovascular health (CVH) has been associated with cognitive decline and dementia, but the extent to which CVH affects brain health remains unclear. We investigated the association of CVH, assessed using Life's Essential 8 (LE8), with neuroimaging-based brain age and brain-predicted age difference (brain-PAD). METHODS This longitudinal community-based study was based on UK Biobank participants aged 40-69 years who were free from dementia and other neurologic diseases at baseline. LE8 score at baseline was assessed with 8 measures and tertiled as low, moderate, and high CVH. Structural and functional brain MRI scans were performed approximately 9 years after baseline, and 1,079 measures from 6 neuroimaging modalities were used to model brain age. A Least Absolute Shrinkage and Selection Operator regression model was trained in 4,355 healthy participants and then used to calculate brain age and brain-PAD in the whole population. Data were analyzed using linear regression models. RESULTS The study included 32,646 participants (mean age at baseline 54.74 years; 53.44% female; mean LE8 score: 71.90). In multivariable-adjusted linear regression, higher LE8 score was associated with younger brain age (β [95% CI] -0.037 [-0.043 to -0.031]) and more negative brain-PAD (β [95% CI] -0.043 [-0.048 to -0.038]) (brain looks younger for chronological age). Compared with high CVH, low/moderate CVH was associated with older brain age (β [95% CI] 1.030 [0.852-1.208]/0.475 [0.303-0.647]) and increased brain-PAD (β [95% CI] 1.193 [1.029-1.357]/0.528 [0.370-0.686]). The associations between low CVH and older brain age/brain-PAD remained similar and significant in both middle-aged (β [95% CI] 1.199 [0.992-1.405]/1.351 [1.159-1.542]) and older adults (β [95% CI] 0.764 [0.417-1.110]/0.948 [0.632-1.263]). DISCUSSION Low CVH is associated with older brain age and greater brain-PAD, even among middle-aged adults. Our findings suggest that optimizing CVH could support brain health. The main limitation of our study is that the study sample was healthier than the general population, thus caution is required when generalizing our findings to other populations.
Collapse
Affiliation(s)
- Huijie Huang
- From the Department of Epidemiology and Biostatistics (H.H., J.M., W.X.), School of Public Health, Tianjin Medical University; Department of Epidemiology (J.W.), College of Preventive Medicine, Third Military Medical University, China; Aging Research Center (M.M.D., J.G., A.D., W.X.), Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden; Department of Nutrition and Health (J.G.), China Agricultural University, Beijing, China; and Rush Alzheimer's Disease Center (D.A.B.), Rush University Medical Center, Chicago, IL
| | - Jiao Wang
- From the Department of Epidemiology and Biostatistics (H.H., J.M., W.X.), School of Public Health, Tianjin Medical University; Department of Epidemiology (J.W.), College of Preventive Medicine, Third Military Medical University, China; Aging Research Center (M.M.D., J.G., A.D., W.X.), Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden; Department of Nutrition and Health (J.G.), China Agricultural University, Beijing, China; and Rush Alzheimer's Disease Center (D.A.B.), Rush University Medical Center, Chicago, IL
| | - Michelle M Dunk
- From the Department of Epidemiology and Biostatistics (H.H., J.M., W.X.), School of Public Health, Tianjin Medical University; Department of Epidemiology (J.W.), College of Preventive Medicine, Third Military Medical University, China; Aging Research Center (M.M.D., J.G., A.D., W.X.), Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden; Department of Nutrition and Health (J.G.), China Agricultural University, Beijing, China; and Rush Alzheimer's Disease Center (D.A.B.), Rush University Medical Center, Chicago, IL
| | - Jie Guo
- From the Department of Epidemiology and Biostatistics (H.H., J.M., W.X.), School of Public Health, Tianjin Medical University; Department of Epidemiology (J.W.), College of Preventive Medicine, Third Military Medical University, China; Aging Research Center (M.M.D., J.G., A.D., W.X.), Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden; Department of Nutrition and Health (J.G.), China Agricultural University, Beijing, China; and Rush Alzheimer's Disease Center (D.A.B.), Rush University Medical Center, Chicago, IL
| | - Abigail Dove
- From the Department of Epidemiology and Biostatistics (H.H., J.M., W.X.), School of Public Health, Tianjin Medical University; Department of Epidemiology (J.W.), College of Preventive Medicine, Third Military Medical University, China; Aging Research Center (M.M.D., J.G., A.D., W.X.), Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden; Department of Nutrition and Health (J.G.), China Agricultural University, Beijing, China; and Rush Alzheimer's Disease Center (D.A.B.), Rush University Medical Center, Chicago, IL
| | - Jun Ma
- From the Department of Epidemiology and Biostatistics (H.H., J.M., W.X.), School of Public Health, Tianjin Medical University; Department of Epidemiology (J.W.), College of Preventive Medicine, Third Military Medical University, China; Aging Research Center (M.M.D., J.G., A.D., W.X.), Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden; Department of Nutrition and Health (J.G.), China Agricultural University, Beijing, China; and Rush Alzheimer's Disease Center (D.A.B.), Rush University Medical Center, Chicago, IL
| | - David A Bennett
- From the Department of Epidemiology and Biostatistics (H.H., J.M., W.X.), School of Public Health, Tianjin Medical University; Department of Epidemiology (J.W.), College of Preventive Medicine, Third Military Medical University, China; Aging Research Center (M.M.D., J.G., A.D., W.X.), Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden; Department of Nutrition and Health (J.G.), China Agricultural University, Beijing, China; and Rush Alzheimer's Disease Center (D.A.B.), Rush University Medical Center, Chicago, IL
| | - Weili Xu
- From the Department of Epidemiology and Biostatistics (H.H., J.M., W.X.), School of Public Health, Tianjin Medical University; Department of Epidemiology (J.W.), College of Preventive Medicine, Third Military Medical University, China; Aging Research Center (M.M.D., J.G., A.D., W.X.), Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden; Department of Nutrition and Health (J.G.), China Agricultural University, Beijing, China; and Rush Alzheimer's Disease Center (D.A.B.), Rush University Medical Center, Chicago, IL
| |
Collapse
|
27
|
Lu H, Li J. MRI-informed machine learning-driven brain age models for classifying mild cognitive impairment converters. J Cent Nerv Syst Dis 2024; 16:11795735241266556. [PMID: 39049837 PMCID: PMC11268046 DOI: 10.1177/11795735241266556] [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: 01/10/2024] [Accepted: 06/02/2024] [Indexed: 07/27/2024] Open
Abstract
BACKGROUND Brain age model, including estimated brain age and brain-predicted age difference (brain-PAD), has shown great potentials for serving as imaging markers for monitoring normal ageing, as well as for identifying the individuals in the pre-diagnostic phase of neurodegenerative diseases. PURPOSE This study aimed to investigate the brain age models in normal ageing and mild cognitive impairments (MCI) converters and their values in classifying MCI conversion. METHODS Pre-trained brain age model was constructed using the structural magnetic resonance imaging (MRI) data from the Cambridge Centre for Ageing and Neuroscience (Cam-CAN) project (N = 609). The tested brain age model was built using the baseline, 1-year and 3-year follow-up MRI data from normal ageing (NA) adults (n = 32) and MCI converters (n = 22) drew from the Open Access Series of Imaging Studies (OASIS-2). The quantitative measures of morphometry included total intracranial volume (TIV), gray matter volume (GMV) and cortical thickness. Brain age models were calculated based on the individual's morphometric features using the support vector machine (SVM) algorithm. RESULTS With comparable chronological age, MCI converters showed significant increased TIV-based (Baseline: P = 0.021; 1-year follow-up: P = 0.037; 3-year follow-up: P = 0.001) and left GMV-based brain age than NA adults at all time points. Higher brain-PAD scores were associated with worse global cognition. Acceptable classification performance of TIV-based (AUC = 0.698) and left GMV-based brain age (AUC = 0.703) was found, which could differentiate the MCI converters from NA adults at the baseline. CONCLUSIONS This is the first demonstration that MRI-informed brain age models exhibit feature-specific patterns. The greater GMV-based brain age observed in MCI converters may provide new evidence for identifying the individuals at the early stage of neurodegeneration. Our findings added value to existing quantitative imaging markers and might help to improve disease monitoring and accelerate personalized treatments in clinical practice.
Collapse
Affiliation(s)
- Hanna Lu
- Department of Psychiatry, The Chinese University of Hong Kong, Hong Kong, China
- Department of Neurology, The Affiliated Brain Hospital of Guangzhou Medical University, Guangzhou, China
| | - Jing Li
- Department of Psychiatry, The Chinese University of Hong Kong, Hong Kong, China
| |
Collapse
|
28
|
Yu Y, Cui H, Haas SS, New F, Sanford N, Yu K, Zhan D, Yang G, Gao J, Wei D, Qiu J, Banaj N, Boomsma DI, Breier A, Brodaty H, Buckner RL, Buitelaar JK, Cannon DM, Caseras X, Clark VP, Conrod PJ, Crivello F, Crone EA, Dannlowski U, Davey CG, de Haan L, de Zubicaray GI, Di Giorgio A, Fisch L, Fisher SE, Franke B, Glahn DC, Grotegerd D, Gruber O, Gur RE, Gur RC, Hahn T, Harrison BJ, Hatton S, Hickie IB, Hulshoff Pol HE, Jamieson AJ, Jernigan TL, Jiang J, Kalnin AJ, Kang S, Kochan NA, Kraus A, Lagopoulos J, Lazaro L, McDonald BC, McDonald C, McMahon KL, Mwangi B, Piras F, Rodriguez‐Cruces R, Royer J, Sachdev PS, Satterthwaite TD, Saykin AJ, Schumann G, Sevaggi P, Smoller JW, Soares JC, Spalletta G, Tamnes CK, Trollor JN, Van't Ent D, Vecchio D, Walter H, Wang Y, Weber B, Wen W, Wierenga LM, Williams SCR, Wu M, Zunta‐Soares GB, Bernhardt B, Thompson P, Frangou S, Ge R. Brain-age prediction: Systematic evaluation of site effects, and sample age range and size. Hum Brain Mapp 2024; 45:e26768. [PMID: 38949537 PMCID: PMC11215839 DOI: 10.1002/hbm.26768] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Revised: 05/15/2024] [Accepted: 06/10/2024] [Indexed: 07/02/2024] Open
Abstract
Structural neuroimaging data have been used to compute an estimate of the biological age of the brain (brain-age) which has been associated with other biologically and behaviorally meaningful measures of brain development and aging. The ongoing research interest in brain-age has highlighted the need for robust and publicly available brain-age models pre-trained on data from large samples of healthy individuals. To address this need we have previously released a developmental brain-age model. Here we expand this work to develop, empirically validate, and disseminate a pre-trained brain-age model to cover most of the human lifespan. To achieve this, we selected the best-performing model after systematically examining the impact of seven site harmonization strategies, age range, and sample size on brain-age prediction in a discovery sample of brain morphometric measures from 35,683 healthy individuals (age range: 5-90 years; 53.59% female). The pre-trained models were tested for cross-dataset generalizability in an independent sample comprising 2101 healthy individuals (age range: 8-80 years; 55.35% female) and for longitudinal consistency in a further sample comprising 377 healthy individuals (age range: 9-25 years; 49.87% female). This empirical examination yielded the following findings: (1) the accuracy of age prediction from morphometry data was higher when no site harmonization was applied; (2) dividing the discovery sample into two age-bins (5-40 and 40-90 years) provided a better balance between model accuracy and explained age variance than other alternatives; (3) model accuracy for brain-age prediction plateaued at a sample size exceeding 1600 participants. These findings have been incorporated into CentileBrain (https://centilebrain.org/#/brainAGE2), an open-science, web-based platform for individualized neuroimaging metrics.
Collapse
Affiliation(s)
- Yuetong Yu
- Djavad Mowafaghian Centre for Brain Health, Department of PsychiatryUniversity of British ColumbiaVancouverBritish ColumbiaCanada
| | - Hao‐Qi Cui
- Djavad Mowafaghian Centre for Brain Health, Department of PsychiatryUniversity of British ColumbiaVancouverBritish ColumbiaCanada
| | - Shalaila S. Haas
- Department of PsychiatryIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
| | - Faye New
- Department of PsychiatryIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
| | - Nicole Sanford
- Djavad Mowafaghian Centre for Brain Health, Department of PsychiatryUniversity of British ColumbiaVancouverBritish ColumbiaCanada
| | - Kevin Yu
- Djavad Mowafaghian Centre for Brain Health, Department of PsychiatryUniversity of British ColumbiaVancouverBritish ColumbiaCanada
| | - Denghuang Zhan
- School of Population and Public HealthUniversity of British ColumbiaVancouverBritish ColumbiaCanada
| | - Guoyuan Yang
- Advanced Research Institute of Multidisciplinary Sciences, School of Medical Technology, School of Life ScienceBeijing Institute of TechnologyBeijingChina
| | - Jia‐Hong Gao
- Center for MRI ResearchPeking UniversityBeijingChina
| | - Dongtao Wei
- School of PsychologySouthwest UniversityChongqingChina
| | - Jiang Qiu
- School of PsychologySouthwest UniversityChongqingChina
| | - Nerisa Banaj
- Laboratory of Neuropsychiatry, Department of Clinical and Behavioral NeurologyIRCCS Santa Lucia FoundationRomeItaly
| | - Dorret I. Boomsma
- Department of Biological PsychologyVrije Universiteit AmsterdamAmsterdamThe Netherlands
| | - Alan Breier
- Department of PsychiatryIndiana University School of MedicineIndianapolisIndianaUSA
| | - Henry Brodaty
- Centre for Healthy Brain Ageing (CHeBA), Discipline of Psychiatry and Mental Health, School of Clinical MedicineUniversity of New South WalesSydneyNew South WalesAustralia
| | - Randy L. Buckner
- Department of Psychology, Center for Brain ScienceHarvard UniversityBostonMassachusettsUSA
- Department of Psychiatry, Massachusetts General HospitalHarvard Medical SchoolBostonMassachusettsUSA
| | - Jan K. Buitelaar
- Department of Cognitive NeuroscienceDonders Institute for Brain, Cognition and Behaviour, Radboud University Medical CenterNijmegenThe Netherlands
| | - Dara M. Cannon
- Centre for Neuroimaging & Cognitive Genomics (NICOG), Clinical Neuroimaging Laboratory, Galway Neuroscience CentreCollege of Medicine Nursing and Health Sciences, University of GalwayGalwayIreland
| | - Xavier Caseras
- Centre for Neuropsychiatric Genetics and Genomics, Division of Psychological Medicine and Clinical NeurosciencesCardiff UniversityCardiffUK
| | - Vincent P. Clark
- Psychology Clinical Neuroscience Center, Department of PsychologyUniversity of New MexicoAlbuquerqueNew MexicoUSA
| | - Patricia J. Conrod
- Department of Psychiatry and AddictionUniversité de Montréal, CHU Ste JustineMontrealQuebecCanada
| | - Fabrice Crivello
- Institut des Maladies NeurodégénérativesUniversité de BordeauxBordeauxFrance
| | - Eveline A. Crone
- Department of Psychology, Faculty of Social SciencesLeiden UniversityLeidenThe Netherlands
- Erasmus School of Social and Behavioral SciencesErasmus University RotterdamRotterdamThe Netherlands
| | - Udo Dannlowski
- Institute for Translational PsychiatryUniversity of MünsterMünsterGermany
| | | | - Lieuwe de Haan
- Department of PsychiatryAmsterdam UMCAmsterdamThe Netherlands
| | - Greig I. de Zubicaray
- Faculty of Health, School of Psychology & CounsellingQueensland University of TechnologyBrisbaneQueenslandAustralia
| | | | - Lukas Fisch
- Institute for Translational PsychiatryUniversity of MünsterMünsterGermany
| | - Simon E. Fisher
- Language and Genetics DepartmentMax Planck Institute for PsycholinguisticsNijmegenThe Netherlands
- Donders Institute for Brain, Cognition and BehaviourRadboud UniversityNijmegenThe Netherlands
| | - Barbara Franke
- Donders Institute for Brain, Cognition and BehaviourRadboud UniversityNijmegenThe Netherlands
- Department of Cognitive NeuroscienceRadboud University Medical CenterNijmegenThe Netherlands
- Department of Human GeneticsRadboud University Medical CenterNijmegenThe Netherlands
| | - David C. Glahn
- Department of Psychiatry, Tommy Fuss Center for Neuropsychiatric Disease Research, Boston Children's HospitalHarvard Medical SchoolBostonMassachusettsUSA
| | - Dominik Grotegerd
- Institute for Translational PsychiatryUniversity of MünsterMünsterGermany
| | - Oliver Gruber
- Section for Experimental Psychopathology and Neuroimaging, Department of General PsychiatryHeidelberg UniversityHeidelbergGermany
| | - Raquel E. Gur
- Department of PsychiatryUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
| | - Ruben C. Gur
- Department of PsychiatryUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
| | - Tim Hahn
- Institute for Translational PsychiatryUniversity of MünsterMünsterGermany
| | - Ben J. Harrison
- Department of PsychiatryThe University of MelbourneMelbourneVictoriaAustralia
| | - Sean Hatton
- Brain and Mind CentreThe University of SydneySydneyNew South WalesAustralia
| | - Ian B. Hickie
- Brain and Mind CentreThe University of SydneySydneyNew South WalesAustralia
| | - Hilleke E. Hulshoff Pol
- Department of PsychiatryIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
- Department of PsychologyUtrecht UniversityUtrechtThe Netherlands
- Department of PsychiatryUniversity Medical Center UtrechtUtrechtThe Netherlands
| | - Alec J. Jamieson
- Department of PsychiatryThe University of MelbourneMelbourneVictoriaAustralia
| | - Terry L. Jernigan
- Center for Human Development, Departments of Cognitive Science, Psychiatry, and RadiologyUniversity of CaliforniaSan DiegoCaliforniaUSA
| | - Jiyang Jiang
- Centre for Healthy Brain Ageing (CHeBA), Discipline of Psychiatry and Mental Health, School of Clinical MedicineUniversity of New South WalesSydneyNew South WalesAustralia
| | - Andrew J. Kalnin
- Department of RadiologyThe Ohio State University College of MedicineColumbusOhioUSA
| | - Sim Kang
- West Region, Institute of Mental HealthSingaporeSingapore
| | - Nicole A. Kochan
- Centre for Healthy Brain Ageing (CHeBA), Discipline of Psychiatry and Mental Health, School of Clinical MedicineUniversity of New South WalesSydneyNew South WalesAustralia
| | - Anna Kraus
- Institute for Translational PsychiatryUniversity of MünsterMünsterGermany
| | - Jim Lagopoulos
- Brain and Mind CentreThe University of SydneySydneyNew South WalesAustralia
| | - Luisa Lazaro
- Department of Child and Adolescent Psychiatry and PsychologyHospital Clínic, IDIBAPS, CIBERSAM, University of BarcelonaBarcelonaSpain
| | - Brenna C. McDonald
- Department of Radiology and Imaging SciencesIndiana University School of MedicineIndianapolisIndianaUSA
| | - Colm McDonald
- Centre for Neuroimaging & Cognitive Genomics (NICOG), Clinical Neuroimaging Laboratory, Galway Neuroscience CentreCollege of Medicine Nursing and Health Sciences, University of GalwayGalwayIreland
| | - Katie L. McMahon
- School of Clinical Sciences, Centre for Biomedical TechnologiesQueensland University of TechnologyBrisbaneQueenslandAustralia
| | - Benson Mwangi
- Louis A. Faillace, MD, Department of Psychiatry and Behavioral SciencesThe University of Texas Health Science Center at HoustonHoustonTexasUSA
| | - Fabrizio Piras
- Laboratory of Neuropsychiatry, Department of Clinical and Behavioral NeurologyIRCCS Santa Lucia FoundationRomeItaly
| | | | - Jessica Royer
- McConnell Brain Imaging CentreMcGill UniversityMontrealQuebecCanada
| | - Perminder S. Sachdev
- Centre for Healthy Brain Ageing (CHeBA), Discipline of Psychiatry and Mental Health, School of Clinical MedicineUniversity of New South WalesSydneyNew South WalesAustralia
| | | | - Andrew J. Saykin
- Department of Radiology and Imaging SciencesIndiana University School of MedicineIndianapolisIndianaUSA
| | - Gunter Schumann
- Department of PsychiatryCCM, Charite Universitaetsmedizin BerlinBerlinGermany
- Centre for Population Neuroscience and Stratified Medicine (PONS), ISTBIFudan UniversityShanghaiChina
| | - Pierluigi Sevaggi
- Department of Translational Biomedicine and NeuroscienceUniversity of Bari Aldo MoroBariItaly
| | - Jordan W. Smoller
- Department of Psychiatry, Massachusetts General HospitalHarvard Medical SchoolBostonMassachusettsUSA
- Center for Genomic MedicineMassachusetts General HospitalBostonMassachusettsUSA
- Center for Precision PsychiatryMassachusetts General HospitalBostonMassachusettsUSA
| | - Jair C. Soares
- Louis A. Faillace, MD, Department of Psychiatry and Behavioral SciencesThe University of Texas Health Science Center at HoustonHoustonTexasUSA
| | - Gianfranco Spalletta
- Laboratory of Neuropsychiatry, Department of Clinical and Behavioral NeurologyIRCCS Santa Lucia FoundationRomeItaly
| | - Christian K. Tamnes
- PROMENTA Research Center, Department of PsychologyUniversity of OsloOsloNorway
| | - Julian N. Trollor
- Centre for Healthy Brain Ageing (CHeBA), Discipline of Psychiatry and Mental Health, School of Clinical MedicineUniversity of New South WalesSydneyNew South WalesAustralia
- Department of Developmental Disability Neuropsychiatry, School of Clinical MedicineUniversity of New South WalesSydneyNew South WalesAustralia
| | - Dennis Van't Ent
- Department of Biological PsychologyVrije Universiteit AmsterdamAmsterdamThe Netherlands
| | - Daniela Vecchio
- Laboratory of Neuropsychiatry, Department of Clinical and Behavioral NeurologyIRCCS Santa Lucia FoundationRomeItaly
| | - Henrik Walter
- Department of Psychiatry and Psychotherapy, Charité Universitätsmedizin BerlinCorporate Member of FU Berlin and Humboldt Universität zu BerlinBerlinGermany
| | - Yang Wang
- Department of RadiologyMedical College of WisconsinMilwaukeeWisconsinUSA
| | - Bernd Weber
- Institute for Experimental Epileptology and Cognition ResearchUniversity of Bonn and University Hospital BonnBonnGermany
| | - Wei Wen
- Centre for Healthy Brain Ageing (CHeBA), Discipline of Psychiatry and Mental Health, School of Clinical MedicineUniversity of New South WalesSydneyNew South WalesAustralia
| | - Lara M. Wierenga
- Department of Psychology, Faculty of Social SciencesLeiden UniversityLeidenThe Netherlands
| | - Steven C. R. Williams
- Department of NeuroimagingInstitute of Psychiatry, Psychology and Neuroscience, King's College LondonLondonUK
| | - Mon‐Ju Wu
- Louis A. Faillace, MD, Department of Psychiatry and Behavioral SciencesThe University of Texas Health Science Center at HoustonHoustonTexasUSA
| | - Giovana B. Zunta‐Soares
- Louis A. Faillace, MD, Department of Psychiatry and Behavioral SciencesThe University of Texas Health Science Center at HoustonHoustonTexasUSA
| | - Boris Bernhardt
- McConnell Brain Imaging CentreMcGill UniversityMontrealQuebecCanada
| | - Paul Thompson
- Imaging Genetics Center, Mark and Mary Stevens Neuroimaging and Informatics Institute, Keck School of MedicineUniversity of Southern CaliforniaMarina del ReyCaliforniaUSA
| | - Sophia Frangou
- Djavad Mowafaghian Centre for Brain Health, Department of PsychiatryUniversity of British ColumbiaVancouverBritish ColumbiaCanada
- Department of PsychiatryIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
| | - Ruiyang Ge
- Djavad Mowafaghian Centre for Brain Health, Department of PsychiatryUniversity of British ColumbiaVancouverBritish ColumbiaCanada
| | | |
Collapse
|
29
|
Lu H, Li J, Chan SSM, Ma SL, Mok VCT, Shi L, Mak ADP, Lam LCW. Predictive values of pre-treatment brain age models to rTMS effects in neurocognitive disorder with depression: Secondary analysis of a randomised sham-controlled clinical trial. DIALOGUES IN CLINICAL NEUROSCIENCE 2024; 26:38-52. [PMID: 38963341 PMCID: PMC11225634 DOI: 10.1080/19585969.2024.2373075] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/18/2024] [Accepted: 06/21/2024] [Indexed: 07/05/2024]
Abstract
INTRODUCTION One major challenge in developing personalised repetitive transcranial magnetic stimulation (rTMS) is that the treatment responses exhibited high inter-individual variations. Brain morphometry might contribute to these variations. This study sought to determine whether individual's brain morphometry could predict the rTMS responders and remitters. METHODS This was a secondary analysis of data from a randomised clinical trial that included fifty-five patients over the age of 60 with both comorbid depression and neurocognitive disorder. Based on magnetic resonance imaging scans, estimated brain age was calculated with morphometric features using a support vector machine. Brain-predicted age difference (brain-PAD) was computed as the difference between brain age and chronological age. RESULTS The rTMS responders and remitters had younger brain age. Every additional year of brain-PAD decreased the odds of relieving depressive symptoms by ∼25.7% in responders (Odd ratio [OR] = 0.743, p = .045) and by ∼39.5% in remitters (OR = 0.605, p = .022) in active rTMS group. Using brain-PAD score as a feature, responder-nonresponder classification accuracies of 85% (3rd week) and 84% (12th week), respectively were achieved. CONCLUSION In elderly patients, younger brain age appears to be associated with better treatment responses to active rTMS. Pre-treatment brain age models informed by morphometry might be used as an indicator to stratify suitable patients for rTMS treatment. TRIAL REGISTRATION ClinicalTrials.gov Identifier: ChiCTR-IOR-16008191.
Collapse
Affiliation(s)
- Hanna Lu
- Department of Psychiatry, The Chinese University of Hong Kong, Hong Kong SAR, China
- The Affiliated Brain Hospital of Guangzhou Medical University, Guangzhou, China
| | - Jing Li
- Department of Psychiatry, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Sandra Sau Man Chan
- Department of Psychiatry, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Suk Ling Ma
- Department of Psychiatry, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Vincent Chung Tong Mok
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Lin Shi
- Department of Imaging and Interventional Radiology, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Arthur Dun-Ping Mak
- Department of Psychiatry, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Linda Chiu Wa Lam
- Department of Psychiatry, The Chinese University of Hong Kong, Hong Kong SAR, China
| |
Collapse
|
30
|
Lin L, Wu Y, Liu L, Sun S, Wu S. Understanding the Temporal Dynamics of Accelerated Brain Aging and Resilient Brain Aging: Insights from Discriminative Event-Based Analysis of UK Biobank Data. Bioengineering (Basel) 2024; 11:647. [PMID: 39061729 PMCID: PMC11273398 DOI: 10.3390/bioengineering11070647] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2024] [Revised: 06/14/2024] [Accepted: 06/21/2024] [Indexed: 07/28/2024] Open
Abstract
The intricate dynamics of brain aging, especially the neurodegenerative mechanisms driving accelerated (ABA) and resilient brain aging (RBA), are pivotal in neuroscience. Understanding the temporal dynamics of these phenotypes is crucial for identifying vulnerabilities to cognitive decline and neurodegenerative diseases. Currently, there is a lack of comprehensive understanding of the temporal dynamics and neuroimaging biomarkers linked to ABA and RBA. This study addressed this gap by utilizing a large-scale UK Biobank (UKB) cohort, with the aim to elucidate brain aging heterogeneity and establish the foundation for targeted interventions. Employing Lasso regression on multimodal neuroimaging data, structural MRI (sMRI), diffusion MRI (dMRI), and resting-state functional MRI (rsfMRI), we predicted the brain age and classified individuals into ABA and RBA cohorts. Our findings identified 1949 subjects (6.2%) as representative of the ABA subpopulation and 3203 subjects (10.1%) as representative of the RBA subpopulation. Additionally, the Discriminative Event-Based Model (DEBM) was applied to estimate the sequence of biomarker changes across aging trajectories. Our analysis unveiled distinct central ordering patterns between the ABA and RBA cohorts, with profound implications for understanding cognitive decline and vulnerability to neurodegenerative disorders. Specifically, the ABA cohort exhibited early degeneration in four functional networks and two cognitive domains, with cortical thinning initially observed in the right hemisphere, followed by the temporal lobe. In contrast, the RBA cohort demonstrated initial degeneration in the three functional networks, with cortical thinning predominantly in the left hemisphere and white matter microstructural degeneration occurring at more advanced stages. The detailed aging progression timeline constructed through our DEBM analysis positioned subjects according to their estimated stage of aging, offering a nuanced view of the aging brain's alterations. This study holds promise for the development of targeted interventions aimed at mitigating age-related cognitive decline.
Collapse
Affiliation(s)
- Lan Lin
- Department of Biomedical Engineering, College of Chemistry and Life Science, Beijing University of Technology, Beijing 100124, China; (Y.W.); (L.L.); (S.W.)
| | - Yutong Wu
- Department of Biomedical Engineering, College of Chemistry and Life Science, Beijing University of Technology, Beijing 100124, China; (Y.W.); (L.L.); (S.W.)
| | - Lingyu Liu
- Department of Biomedical Engineering, College of Chemistry and Life Science, Beijing University of Technology, Beijing 100124, China; (Y.W.); (L.L.); (S.W.)
| | - Shen Sun
- Intelligent Physiological Measurement and Clinical Translation, Beijing International Base for Scientific and Technological Cooperation, Beijing University of Technology, Beijing 100124, China
| | - Shuicai Wu
- Department of Biomedical Engineering, College of Chemistry and Life Science, Beijing University of Technology, Beijing 100124, China; (Y.W.); (L.L.); (S.W.)
| |
Collapse
|
31
|
Pastika L, Sau A, Patlatzoglou K, Sieliwonczyk E, Ribeiro AH, McGurk KA, Khan S, Mandic D, Scott WR, Ware JS, Peters NS, Ribeiro ALP, Kramer DB, Waks JW, Ng FS. Artificial intelligence-enhanced electrocardiography derived body mass index as a predictor of future cardiometabolic disease. NPJ Digit Med 2024; 7:167. [PMID: 38918595 PMCID: PMC11199586 DOI: 10.1038/s41746-024-01170-0] [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: 01/24/2024] [Accepted: 06/14/2024] [Indexed: 06/27/2024] Open
Abstract
The electrocardiogram (ECG) can capture obesity-related cardiac changes. Artificial intelligence-enhanced ECG (AI-ECG) can identify subclinical disease. We trained an AI-ECG model to predict body mass index (BMI) from the ECG alone. Developed from 512,950 12-lead ECGs from the Beth Israel Deaconess Medical Center (BIDMC), a secondary care cohort, and validated on UK Biobank (UKB) (n = 42,386), the model achieved a Pearson correlation coefficient (r) of 0.65 and 0.62, and an R2 of 0.43 and 0.39 in the BIDMC cohort and UK Biobank, respectively for AI-ECG BMI vs. measured BMI. We found delta-BMI, the difference between measured BMI and AI-ECG-predicted BMI (AI-ECG-BMI), to be a biomarker of cardiometabolic health. The top tertile of delta-BMI showed increased risk of future cardiometabolic disease (BIDMC: HR 1.15, p < 0.001; UKB: HR 1.58, p < 0.001) and diabetes mellitus (BIDMC: HR 1.25, p < 0.001; UKB: HR 2.28, p < 0.001) after adjusting for covariates including measured BMI. Significant enhancements in model fit, reclassification and improvements in discriminatory power were observed with the inclusion of delta-BMI in both cohorts. Phenotypic profiling highlighted associations between delta-BMI and cardiometabolic diseases, anthropometric measures of truncal obesity, and pericardial fat mass. Metabolic and proteomic profiling associates delta-BMI positively with valine, lipids in small HDL, syntaxin-3, and carnosine dipeptidase 1, and inversely with glutamine, glycine, colipase, and adiponectin. A genome-wide association study revealed associations with regulators of cardiovascular/metabolic traits, including SCN10A, SCN5A, EXOG and RXRG. In summary, our AI-ECG-BMI model accurately predicts BMI and introduces delta-BMI as a non-invasive biomarker for cardiometabolic risk stratification.
Collapse
Affiliation(s)
- Libor Pastika
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Arunashis Sau
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
- Department of Cardiology, Imperial College Healthcare NHS Trust, London, United Kingdom
| | | | - Ewa Sieliwonczyk
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
- MRC Laboratory of Medical Sciences, Imperial College London, London, United Kingdom
| | - Antônio H Ribeiro
- Department of Information Technology, Uppsala University, Uppsala, Sweden
| | - Kathryn A McGurk
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
- MRC Laboratory of Medical Sciences, Imperial College London, London, United Kingdom
| | - Sadia Khan
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
- Chelsea and Westminster NHS Foundation Trust, London, United Kingdom
| | - Danilo Mandic
- Department of Electrical and Electronic Engineering, Imperial College London, London, United Kingdom
| | - William R Scott
- MRC Laboratory of Medical Sciences, Imperial College London, London, United Kingdom
- Institute of Clinical Sciences, Faculty of Medicine, Imperial College London, London, United Kingdom
| | - James S Ware
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
- MRC Laboratory of Medical Sciences, Imperial College London, London, United Kingdom
| | - Nicholas S Peters
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
- Department of Cardiology, Imperial College Healthcare NHS Trust, London, United Kingdom
| | - Antonio Luiz P Ribeiro
- Department of Internal Medicine, Faculdade de Medicina, and Telehealth Center and Cardiology Service, Hospital das Clínicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Daniel B Kramer
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
- Richard A. and Susan F. Smith Center for Outcomes Research in Cardiology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Jonathan W Waks
- Harvard-Thorndike Electrophysiology Institute, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Fu Siong Ng
- National Heart and Lung Institute, Imperial College London, London, United Kingdom.
- Department of Cardiology, Imperial College Healthcare NHS Trust, London, United Kingdom.
- Chelsea and Westminster NHS Foundation Trust, London, United Kingdom.
| |
Collapse
|
32
|
Tetereva A, Pat N. Brain age has limited utility as a biomarker for capturing fluid cognition in older individuals. eLife 2024; 12:RP87297. [PMID: 38869938 PMCID: PMC11175613 DOI: 10.7554/elife.87297] [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: 06/14/2024] Open
Abstract
One well-known biomarker candidate that supposedly helps capture fluid cognition is Brain Age, or a predicted value based on machine-learning models built to predict chronological age from brain MRI. To formally evaluate the utility of Brain Age for capturing fluid cognition, we built 26 age-prediction models for Brain Age based on different combinations of MRI modalities, using the Human Connectome Project in Aging (n=504, 36-100 years old). First, based on commonality analyses, we found a large overlap between Brain Age and chronological age: Brain Age could uniquely add only around 1.6% in explaining variation in fluid cognition over and above chronological age. Second, the age-prediction models that performed better at predicting chronological age did NOT necessarily create better Brain Age for capturing fluid cognition over and above chronological age. Instead, better-performing age-prediction models created Brain Age that overlapped larger with chronological age, up to around 29% out of 32%, in explaining fluid cognition. Third, Brain Age missed around 11% of the total variation in fluid cognition that could have been explained by the brain variation. That is, directly predicting fluid cognition from brain MRI data (instead of relying on Brain Age and chronological age) could lead to around a 1/3-time improvement of the total variation explained. Accordingly, we demonstrated the limited utility of Brain Age as a biomarker for fluid cognition and made some suggestions to ensure the utility of Brain Age in explaining fluid cognition and other phenotypes of interest.
Collapse
Affiliation(s)
- Alina Tetereva
- Department of Psychology, University of OtagoDunedinNew Zealand
| | - Narun Pat
- Department of Psychology, University of OtagoDunedinNew Zealand
| |
Collapse
|
33
|
Pu F, Chen W, Li C, Fu J, Gao W, Ma C, Cao X, Zhang L, Hao M, Zhou J, Huang R, Ma Y, Hu K, Liu Z. Heterogeneous associations of multiplexed environmental factors and multidimensional aging metrics. Nat Commun 2024; 15:4921. [PMID: 38858361 PMCID: PMC11164970 DOI: 10.1038/s41467-024-49283-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2023] [Accepted: 05/31/2024] [Indexed: 06/12/2024] Open
Abstract
Complicated associations between multiplexed environmental factors and aging are poorly understood. We manipulated aging using multidimensional metrics such as phenotypic age, brain age, and brain volumes in the UK Biobank. Weighted quantile sum regression was used to examine the relative individual contributions of multiplexed environmental factors to aging, and self-organizing maps (SOMs) were used to examine joint effects. Air pollution presented a relatively large contribution in most cases. We also found fair heterogeneities in which the same environmental factor contributed inconsistently to different aging metrics. Particulate matter contributed the most to variance in aging, while noise and green space showed considerable contribution to brain volumes. SOM identified five subpopulations with distinct environmental exposure patterns and the air pollution subpopulation had the worst aging status. This study reveals the heterogeneous associations of multiplexed environmental factors with multidimensional aging metrics and serves as a proof of concept when analyzing multifactors and multiple outcomes.
Collapse
Affiliation(s)
- Fan Pu
- 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 Province, Zhejiang University School of Medicine, Hangzhou, 310058, Zhejiang, China
| | - Weiran Chen
- 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 Province, Zhejiang University School of Medicine, Hangzhou, 310058, Zhejiang, China
| | - Chenxi Li
- 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 Province, Zhejiang University School of Medicine, Hangzhou, 310058, Zhejiang, China
| | - Jingqiao Fu
- Ocean College, Zhejiang University, Zhoushan, 316021, Zhejiang, China
| | - Weijing Gao
- 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 Province, Zhejiang University School of Medicine, Hangzhou, 310058, Zhejiang, China
| | - Chao Ma
- School of Economics and Management, Southeast University, Nanjing, 211189, Jiangsu, China
| | - Xingqi Cao
- Center for Clinical Big Data and Analytics of the Second Affiliated Hospital, and Department of Big Data in Health Science School of Public Health, The Key Laboratory of Intelligent Preventive Medicine of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, 310058, Zhejiang, China
| | - Lingzhi 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 Province, Zhejiang University School of Medicine, Hangzhou, 310058, Zhejiang, China
| | - Meng Hao
- State Key Laboratory of Genetic Engineering, Collaborative Innovation Center for Genetics and Development, School of Life Sciences and Human Phenome Institute, Fudan University, Shanghai, 200433, China
| | - Jin Zhou
- Key Laboratory of Environmental Stress and Chronic Disease Control & Prevention, Ministry of Education, China Medical University; Department of Biostatistics and Epidemiology, School of Public Health, China Medical University, Shenyang, 110122, Liaoning, China
| | - Rong Huang
- Key Laboratory of Environmental Stress and Chronic Disease Control & Prevention, Ministry of Education, China Medical University; Department of Biostatistics and Epidemiology, School of Public Health, China Medical University, Shenyang, 110122, Liaoning, China
| | - Yanan Ma
- Key Laboratory of Environmental Stress and Chronic Disease Control & Prevention, Ministry of Education, China Medical University; Department of Biostatistics and Epidemiology, School of Public Health, China Medical University, Shenyang, 110122, Liaoning, China.
| | - Kejia Hu
- Department of Big Data in Health Science School of Public Health, Zhejiang University School of Medicine, Hangzhou, 310058, Zhejiang, China.
| | - Zuyun Liu
- Center for Clinical Big Data and Analytics of the Second Affiliated Hospital, and Department of Big Data in Health Science School of Public Health, The Key Laboratory of Intelligent Preventive Medicine of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, 310058, Zhejiang, China.
| |
Collapse
|
34
|
Aghaei A, Ebrahimi Moghaddam M. Brain age gap estimation using attention-based ResNet method for Alzheimer's disease detection. Brain Inform 2024; 11:16. [PMID: 38833039 DOI: 10.1186/s40708-024-00230-1] [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: 11/03/2023] [Accepted: 05/27/2024] [Indexed: 06/06/2024] Open
Abstract
This study investigates the correlation between brain age and chronological age in healthy individuals using brain MRI images, aiming to identify potential biomarkers for neurodegenerative diseases like Alzheimer's. To achieve this, a novel attention-based ResNet method, 3D-Attention-Resent-SVR, is proposed to accurately estimate brain age and distinguish between Cognitively Normal (CN) and Alzheimer's disease (AD) individuals by computing the brain age gap (BAG). Unlike conventional methods, which often rely on single datasets, our approach addresses potential biases by employing four datasets for training and testing. The results, based on a combined dataset from four public sources comprising 3844 data points, demonstrate the model's efficacy with a mean absolute error (MAE) of 2.05 for brain age gap estimation. Moreover, the model's generalizability is showcased by training on three datasets and testing on a separate one, yielding a remarkable MAE of 2.4. Furthermore, leveraging BAG as the sole biomarker, our method achieves an accuracy of 92% and an AUC of 0.87 in Alzheimer's disease detection on the ADNI dataset. These findings underscore the potential of our approach in assisting with early detection and disease monitoring, emphasizing the strong correlation between BAG and AD.
Collapse
Affiliation(s)
- Atefe Aghaei
- Faculty of Computer Science and Engineering, Shahid Beheshti University, Tehran, Iran
| | | |
Collapse
|
35
|
Yang X, Sullivan PF, Li B, Fan Z, Ding D, Shu J, Guo Y, Paschou P, Bao J, Shen L, Ritchie MD, Nave G, Platt ML, Li T, Zhu H, Zhao B. Multi-organ imaging-derived polygenic indexes for brain and body health. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2023.04.18.23288769. [PMID: 38883759 PMCID: PMC11177904 DOI: 10.1101/2023.04.18.23288769] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/18/2024]
Abstract
The UK Biobank (UKB) imaging project is a crucial resource for biomedical research, but is limited to 100,000 participants due to cost and accessibility barriers. Here we used genetic data to predict heritable imaging-derived phenotypes (IDPs) for a larger cohort. We developed and evaluated 4,375 IDP genetic scores (IGS) derived from UKB brain and body images. When applied to UKB participants who were not imaged, IGS revealed links to numerous phenotypes and stratified participants at increased risk for both brain and somatic diseases. For example, IGS identified individuals at higher risk for Alzheimer's disease and multiple sclerosis, offering additional insights beyond traditional polygenic risk scores of these diseases. When applied to independent external cohorts, IGS also stratified those at high disease risk in the All of Us Research Program and the Alzheimer's Disease Neuroimaging Initiative study. Our results demonstrate that, while the UKB imaging cohort is largely healthy and may not be the most enriched for disease risk management, it holds immense potential for stratifying the risk of various brain and body diseases in broader external genetic cohorts.
Collapse
Affiliation(s)
- Xiaochen Yang
- Department of Statistics, Purdue University, West Lafayette, IN 47907, USA
| | - Patrick F. Sullivan
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Bingxuan Li
- UCLA Samueli School of Engineering, Los Angeles, CA 90095, USA
| | - Zirui Fan
- Department of Statistics and Data Science, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Dezheng Ding
- Department of Electrical and Systems Engineering, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Juan Shu
- Department of Statistics, Purdue University, West Lafayette, IN 47907, USA
| | - Yuxin Guo
- Department of Biological Sciences, Purdue University, West Lafayette, IN 47907, USA
| | - Peristera Paschou
- Department of Biological Sciences, Purdue University, West Lafayette, IN 47907, USA
| | - Jingxuan Bao
- Department of Biostatistics, Epidemiology and Informatics, University of Pennsylvania, Philadelphia, PA 19104, USA
- Graduate Group in Genomics and Computational Biology, University of Pennsylvania, Philadelphia, PA, USA
| | - Li Shen
- Department of Biostatistics, Epidemiology and Informatics, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Marylyn D. Ritchie
- Department of Genetics, University of Pennsylvania, Philadelphia, PA 19104, USA
- Institute for Biomedical Informatics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Gideon Nave
- Marketing Department, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Michael L. Platt
- Marketing Department, University of Pennsylvania, Philadelphia, PA 19104, USA
- Department of Neuroscience, University of Pennsylvania, Philadelphia, PA 19104, USA
- Department of Psychology, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Tengfei Li
- Department of Radiology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Biomedical Research Imaging Center, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Hongtu Zhu
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Biomedical Research Imaging Center, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Department of Biostatistics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Department of Computer Science, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Department of Statistics and Operations Research, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Bingxin Zhao
- Department of Statistics and Data Science, University of Pennsylvania, Philadelphia, PA 19104, USA
- Institute for Biomedical Informatics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
- Applied Mathematics and Computational Science Graduate Group, University of Pennsylvania, Philadelphia, PA 19104, USA
- Center for AI and Data Science for Integrated Diagnostics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Population Aging Research Center, University of Pennsylvania, Philadelphia, PA 19104, USA
- Institute for Translational Medicine and Therapeutics, University of Pennsylvania, Philadelphia, PA 19104, USA
| |
Collapse
|
36
|
Sandalova E, Maier AB. Targeting the epigenetically older individuals for geroprotective trials: the use of DNA methylation clocks. Biogerontology 2024; 25:423-431. [PMID: 37968337 DOI: 10.1007/s10522-023-10077-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2023] [Accepted: 10/15/2023] [Indexed: 11/17/2023]
Abstract
Chronological age is the most important risk factor for the incidence of age-related diseases. The pace of ageing determines the magnitude of that risk and can be expressed as biological age. Targeting fundamental pathways of human aging with geroprotectors has the potential to lower the biological age and therewith prolong the healthspan, the period of life one spends in good health. Target populations for geroprotective interventions should be chosen based on the ageing mechanisms being addressed and the expected effect of the geroprotector on the primary outcome. Biomarkers of ageing, such as DNA methylation age, can be used to select populations for geroprotective interventions and as a surrogate outcome. Here, the use of DNA methylation clocks for selecting target populations for geroprotective intervention is explored.
Collapse
Affiliation(s)
- Elena Sandalova
- Healthy Longevity Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.
- Centre for Healthy Longevity, @AgeSingapore, National University Health System, Singapore, Singapore.
| | - Andrea B Maier
- Healthy Longevity Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.
- Centre for Healthy Longevity, @AgeSingapore, National University Health System, Singapore, Singapore.
- Department of Human Movement Sciences, @AgeAmsterdam, Amsterdam Movement Sciences, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands.
| |
Collapse
|
37
|
Paliwal V, Das K, Doesburg SM, Medvedev G, Xi P, Ribary U, Pachori RB, Vakorin VA. Classifying Routine Clinical Electroencephalograms With Multivariate Iterative Filtering and Convolutional Neural Networks. IEEE Trans Neural Syst Rehabil Eng 2024; 32:2038-2048. [PMID: 38768007 DOI: 10.1109/tnsre.2024.3403198] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/22/2024]
Abstract
Electroencephalogram (EEG) is widely used in basic and clinical neuroscience to explore neural states in various populations, and classifying these EEG recordings is a fundamental challenge. While machine learning shows promising results in classifying long multivariate time series, optimal prediction models and feature extraction methods for EEG classification remain elusive. Our study addressed the problem of EEG classification under the framework of brain age prediction, applying a deep learning model on EEG time series. We hypothesized that decomposing EEG signals into oscillatory modes would yield more accurate age predictions than using raw or canonically frequency-filtered EEG. Specifically, we employed multivariate intrinsic mode functions (MIMFs), an empirical mode decomposition (EMD) variant based on multivariate iterative filtering (MIF), with a convolutional neural network (CNN) model. Testing a large dataset of routine clinical EEG scans (n = 6540) from patients aged 1 to 103 years, we found that an ad-hoc CNN model without fine-tuning could reasonably predict brain age from EEGs. Crucially, MIMF decomposition significantly improved performance compared to canonical brain rhythms (from delta to lower gamma oscillations). Our approach achieved a mean absolute error (MAE) of 13.76 ± 0.33 and a correlation coefficient of 0.64 ± 0.01 in brain age prediction over the entire lifespan. Our findings indicate that CNN models applied to EEGs, preserving their original temporal structure, remains a promising framework for EEG classification, wherein the adaptive signal decompositions such as the MIF can enhance CNN models' performance in this task.
Collapse
|
38
|
Sorooshyari SK. Brain age monotonicity and functional connectivity differences of healthy subjects. PLoS One 2024; 19:e0300720. [PMID: 38814972 PMCID: PMC11139261 DOI: 10.1371/journal.pone.0300720] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Accepted: 03/04/2024] [Indexed: 06/01/2024] Open
Abstract
Alterations in the brain's connectivity or the interactions among brain regions have been studied with the aid of resting state (rs)fMRI data attained from large numbers of healthy subjects of various demographics. This has been instrumental in providing insight into how a phenotype as fundamental as age affects the brain. Although machine learning (ML) techniques have already been deployed in such studies, novel questions are investigated in this work. We study whether young brains develop properties that progressively resemble those of aged brains, and if the aging dynamics of older brains provide information about the aging trajectory in young subjects. The degree of a prospective monotonic relationship will be quantified, and hypotheses of brain aging trajectories will be tested via ML. Furthermore, the degree of functional connectivity across the age spectrum of three datasets will be compared at a population level and across sexes. The findings scrutinize similarities and differences among the male and female subjects at greater detail than previously performed.
Collapse
Affiliation(s)
- Siamak K. Sorooshyari
- Department of Statistics, Stanford University, Stanford, CA, United States of America
| |
Collapse
|
39
|
Ruffle JK, Gray RJ, Mohinta S, Pombo G, Kaul C, Hyare H, Rees G, Nachev P. Computational limits to the legibility of the imaged human brain. Neuroimage 2024; 291:120600. [PMID: 38569979 DOI: 10.1016/j.neuroimage.2024.120600] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Revised: 03/08/2024] [Accepted: 03/31/2024] [Indexed: 04/05/2024] Open
Abstract
Our knowledge of the organisation of the human brain at the population-level is yet to translate into power to predict functional differences at the individual-level, limiting clinical applications and casting doubt on the generalisability of inferred mechanisms. It remains unknown whether the difficulty arises from the absence of individuating biological patterns within the brain, or from limited power to access them with the models and compute at our disposal. Here we comprehensively investigate the resolvability of such patterns with data and compute at unprecedented scale. Across 23 810 unique participants from UK Biobank, we systematically evaluate the predictability of 25 individual biological characteristics, from all available combinations of structural and functional neuroimaging data. Over 4526 GPU*hours of computation, we train, optimize, and evaluate out-of-sample 700 individual predictive models, including fully-connected feed-forward neural networks of demographic, psychological, serological, chronic disease, and functional connectivity characteristics, and both uni- and multi-modal 3D convolutional neural network models of macro- and micro-structural brain imaging. We find a marked discrepancy between the high predictability of sex (balanced accuracy 99.7%), age (mean absolute error 2.048 years, R2 0.859), and weight (mean absolute error 2.609Kg, R2 0.625), for which we set new state-of-the-art performance, and the surprisingly low predictability of other characteristics. Neither structural nor functional imaging predicted an individual's psychology better than the coincidence of common chronic disease (p < 0.05). Serology predicted chronic disease (p < 0.05) and was best predicted by it (p < 0.001), followed by structural neuroimaging (p < 0.05). Our findings suggest either more informative imaging or more powerful models will be needed to decipher individual level characteristics from the human brain. We make our models and code openly available.
Collapse
Affiliation(s)
- James K Ruffle
- Queen Square Institute of Neurology, University College London, London, United Kingdom.
| | - Robert J Gray
- Queen Square Institute of Neurology, University College London, London, United Kingdom
| | - Samia Mohinta
- Queen Square Institute of Neurology, University College London, London, United Kingdom
| | - Guilherme Pombo
- Queen Square Institute of Neurology, University College London, London, United Kingdom
| | - Chaitanya Kaul
- School of Computing Science, University of Glasgow, Glasgow, United Kingdom
| | - Harpreet Hyare
- Queen Square Institute of Neurology, University College London, London, United Kingdom
| | - Geraint Rees
- Queen Square Institute of Neurology, University College London, London, United Kingdom
| | - Parashkev Nachev
- Queen Square Institute of Neurology, University College London, London, United Kingdom.
| |
Collapse
|
40
|
Dular L, Pernuš F, Špiclin Ž. Extensive T1-weighted MRI preprocessing improves generalizability of deep brain age prediction models. Comput Biol Med 2024; 173:108320. [PMID: 38531250 DOI: 10.1016/j.compbiomed.2024.108320] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Revised: 01/09/2024] [Accepted: 03/12/2024] [Indexed: 03/28/2024]
Abstract
Brain age is an estimate of chronological age obtained from T1-weighted magnetic resonance images (T1w MRI), representing a straightforward diagnostic biomarker of brain aging and associated diseases. While the current best accuracy of brain age predictions on T1w MRIs of healthy subjects ranges from two to three years, comparing results across studies is challenging due to differences in the datasets, T1w preprocessing pipelines, and evaluation protocols used. This paper investigates the impact of T1w image preprocessing on the performance of four deep learning brain age models from recent literature. Four preprocessing pipelines, which differed in terms of registration transform, grayscale correction, and software implementation, were evaluated. The results showed that the choice of software or preprocessing steps could significantly affect the prediction error, with a maximum increase of 0.75 years in mean absolute error (MAE) for the same model and dataset. While grayscale correction had no significant impact on MAE, using affine rather than rigid registration to brain atlas statistically significantly improved MAE. Models trained on 3D images with isotropic 1mm3 resolution exhibited less sensitivity to the T1w preprocessing variations compared to 2D models or those trained on downsampled 3D images. Our findings indicate that extensive T1w preprocessing improves MAE, especially when predicting on a new dataset. This runs counter to prevailing research literature, which suggests that models trained on minimally preprocessed T1w scans are better suited for age predictions on MRIs from unseen scanners. We demonstrate that, irrespective of the model or T1w preprocessing used during training, applying some form of offset correction is essential to enable the model's performance to generalize effectively on datasets from unseen sites, regardless of whether they have undergone the same or different T1w preprocessing as the training set.
Collapse
Affiliation(s)
- Lara Dular
- University of Ljubljana, Faculty of Electrical Engineering, Tržaška cesta 25, Ljubljana 1000, Slovenia
| | - Franjo Pernuš
- University of Ljubljana, Faculty of Electrical Engineering, Tržaška cesta 25, Ljubljana 1000, Slovenia
| | - Žiga Špiclin
- University of Ljubljana, Faculty of Electrical Engineering, Tržaška cesta 25, Ljubljana 1000, Slovenia.
| |
Collapse
|
41
|
Kirschen RM, Leaver AM. Hearing function moderates age-related changes in brain morphometry in the HCP Aging cohort. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.22.590589. [PMID: 38712119 PMCID: PMC11071358 DOI: 10.1101/2024.04.22.590589] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/08/2024]
Abstract
Introduction There are well-established relationships between aging and neurodegenerative changes, and between aging and hearing loss. The goal of this study was to determine how structural brain aging is influenced by hearing loss. Methods Human Connectome Project Aging (HCP-A) data were analyzed, including T1-weighted MRI and Words in Noise (WIN) thresholds (n=623). Freesurfer extracted gray and white matter volume, and cortical thickness, area, and curvature. Linear regression models targeted (1) interactions between age and WIN threshold and (2) correlations with WIN threshold adjusted for age, both corrected for false discovery rate (pFDR<0.05). Results WIN threshold moderated age-related increase in volume in bilateral inferior lateral ventricles, with higher threshold associated with increased age-related ventricle expansion. Age-related deterioration in occipital cortex was also increased with higher WIN thresholds. When controlling for age, high WIN threshold was correlated with reduced cortical thickness in Heschl's gyrus, calcarine sulcus, and other sensory regions, and reduced temporal lobe white matter. Older volunteers with poorer hearing and cognitive scores had the lowest volume in left parahippocampal white matter. Conclusions Preserved hearing abilities in aging associated with a reduction of age-related changes to medial temporal lobe, and preserved hearing at any age associated with preserved cortical tissue in auditory and other sensory regions. Future longitudinal studies are needed to assess the causal nature of these relationships, but these results indicate interventions which preserve hearing function may combat some neurodegenerative changes in aging.
Collapse
Affiliation(s)
| | - Amber M. Leaver
- Department of Radiology, Northwestern University, Chicago, IL, 60611
| |
Collapse
|
42
|
Sorooshyari SK. Beyond network connectivity: A classification approach to brain age prediction with resting-state fMRI. Neuroimage 2024; 290:120570. [PMID: 38467344 DOI: 10.1016/j.neuroimage.2024.120570] [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: 11/20/2023] [Revised: 03/06/2024] [Accepted: 03/08/2024] [Indexed: 03/13/2024] Open
Abstract
The brain is a complex, dynamic organ that shows differences in the same subject at various periods. Understanding how brain activity changes across age as a function of the brain networks has been greatly abetted by fMRI. Canonical analysis consists of determining how alterations in connectivity patterns (CPs) of certain regions are affected. An alternative approach is taken here by not considering connectivity but rather features computed from recordings at the regions of interest (ROIs). Using machine learning (ML) we assess how neural signals are altered by and prospectively predictive of age and sex via a methodology that is novel in drawing upon pairwise classification across six decades of subjects' chronological ages. ML is used to answer the equally important questions of what properties of the computed features are most predictive as well as which brain networks are most affected by aging. It was found that there is decreased differentiation among the neural signals of older subjects that are separated in age by the same number of years as younger subjects. Furthermore, the burstiness of the signals change at different rates between males and females. The findings provide insight into brain aging via an ROI-based analysis, the consideration of several feature groups, and a novel classification-based ML pipeline. There is also a contribution to understanding the effects of data aggregated from different recording centers on the conclusions of fMRI studies.
Collapse
|
43
|
Wang G, Jiang N, Ma Y, Suo D, Liu T, Funahashi S, Yan T. Using a deep generation network reveals neuroanatomical specificity in hemispheres. PATTERNS (NEW YORK, N.Y.) 2024; 5:100930. [PMID: 38645770 PMCID: PMC11026975 DOI: 10.1016/j.patter.2024.100930] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Revised: 01/08/2024] [Accepted: 01/15/2024] [Indexed: 04/23/2024]
Abstract
Asymmetry is an important property of brain organization, but its nature is still poorly understood. Capturing the neuroanatomical components specific to each hemisphere facilitates the understanding of the establishment of brain asymmetry. Since deep generative networks (DGNs) have powerful inference and recovery capabilities, we use one hemisphere to predict the opposite hemisphere by training the DGNs, which automatically fit the built-in dependencies between the left and right hemispheres. After training, the reconstructed images approximate the homologous components in the hemisphere. We use the difference between the actual and reconstructed hemispheres to measure hemisphere-specific components due to asymmetric expression of environmental and genetic factors. The results show that our model is biologically plausible and that our proposed metric of hemispheric specialization is reliable, representing a wide range of individual variation. Together, this work provides promising tools for exploring brain asymmetry and new insights into self-supervised DGNs for representing the brain.
Collapse
Affiliation(s)
- Gongshu Wang
- School of Medical Technology, Beijing Institute of Technology, Beijing 100081, China
| | - Ning Jiang
- School of Medical Technology, Beijing Institute of Technology, Beijing 100081, China
| | - Yunxiao Ma
- School of Medical Technology, Beijing Institute of Technology, Beijing 100081, China
| | - Dingjie Suo
- School of Medical Technology, Beijing Institute of Technology, Beijing 100081, China
| | - Tiantian Liu
- School of Medical Technology, Beijing Institute of Technology, Beijing 100081, China
| | - Shintaro Funahashi
- Advanced Research Institute for Multidisciplinary Science, Beijing Institute of Technology, Beijing 100081, China
- Department of Cognitive and Behavioral Sciences, Graduate School of Human and Environmental Science, Kyoto University, Sakyo-ku, Kyoto 606-8501, Japan
- Kokoro Research Center, Kyoto University, Sakyo-ku, Kyoto 606-8501, Japan
| | - Tianyi Yan
- School of Medical Technology, Beijing Institute of Technology, Beijing 100081, China
| |
Collapse
|
44
|
Cumplido-Mayoral I, Brugulat-Serrat A, Sánchez-Benavides G, González-Escalante A, Anastasi F, Milà-Alomà M, López-Martos D, Akinci M, Falcón C, Shekari M, Cacciaglia R, Arenaza-Urquijo EM, Minguillón C, Fauria K, Molinuevo JL, Suárez-Calvet M, Grau-Rivera O, Vilaplana V, Gispert JD. The mediating role of neuroimaging-derived biological brain age in the association between risk factors for dementia and cognitive decline in middle-aged and older individuals without cognitive impairment: a cohort study. THE LANCET. HEALTHY LONGEVITY 2024; 5:e276-e286. [PMID: 38555920 DOI: 10.1016/s2666-7568(24)00025-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2023] [Revised: 01/29/2024] [Accepted: 01/31/2024] [Indexed: 04/02/2024] Open
Abstract
BACKGROUND Neuroimaging-based brain-age delta has been shown to be a mediator linking cardiovascular risk factors to cognitive function. We aimed to assess the mediating role of brain-age delta in the association between modifiable risk factors of dementia and longitudinal cognitive decline in middle-aged and older individuals who are asymptomatic, stratified by Alzheimer's disease pathology. We also explored whether the mediation effect is specific to cognitive domain. METHODS In this cohort study, we included participants from the ALFA+ cohort aged between 45 years and 65 years who were cognitively unimpaired and who had available structural MRI, cerebrospinal fluid β-amyloid (Aβ)42 and Aβ40 measurements obtained within 1 year of each other, modifiable risk factors assessment, and cognitive evaluation over 3 years. Participants were recruited from the Barcelonaβeta Brain Research Center (Barcelona, Spain). Included individuals underwent a first assessment between Oct 25, 2016, and Jan 28, 2020, and a follow-up cognitive assessment 3·28 (SD 0·27) years later. We computed brain-age delta and composites of different cognitive function domains (preclinical Alzheimer's cognitive composite [PACC], attention, executive function, episodic memory, visual processing, and language). We used partial least squares path modelling to explore mediation effects in the associations between modifiable risk factors (including cardiovascular, mental health, mood, metabolic or endocrine history, and alcohol use) and changes in cognitive composites. To assess the role of Alzheimer's disease pathology, we computed separate models for Aβ-negative and Aβ-positive individuals. FINDINGS Of the 419 participants enrolled in ALFA+, 302 met our inclusion criteria, of which 108 participants were classified as Aβ-positive and 194 as Aβ-negative. In Aβ-positive individuals, brain-age delta partially mediated (percent mediation proportion 15·73% [95% CI 14·22-16·66]) the association between modifiable risk factors and decline in overall cognition (across cognitive domains). Brain-age delta fully mediated (mediation proportion 28·03% [26·25-29·21]) the effect of modifiable risk factors on the PACC, wherein increased values for risk factors correlated with an older brain-age delta, and, consequently, an older brain-age delta was linked to greater PACC decline. This effect appears to be primarily driven by memory decline. Mediation was not significant in Aβ-negative individuals (3·52% [0·072-4·17]) on PACC, although path coefficients were not significantly different from those in the Aβ-positive group. INTERPRETATION Our findings suggest that brain-age delta captures the association between modifiable risk factors and longitudinal cognitive decline in middle-aged and older people. In asymptomatic middle-aged and older individuals who are Aβ-positive, the pathology might be the strongest driver of cognitive decline, whereas the effect of risk factors is smaller. Our results highlight the potential of brain-age delta as an objective outcome measure for preventive lifestyle interventions targeting cognitive decline. FUNDING La Caixa Foundation, the TriBEKa Imaging Platform, and the Universities and Research Secretariat of the Catalan Government. TRANSLATION For the Spanish translation of the abstract see Supplementary Materials section.
Collapse
Affiliation(s)
- Irene Cumplido-Mayoral
- Barcelonaβeta Brain Research Center, Pasqual Maragall Foundation, Barcelona, Spain; Biomedicine, Universitat Pompeu Fabra, Barcelona, Spain; Neuroimagen de Enfermedades Neurodegenerativas y Envejecimiento Saludable, Hospital del Mar Research Institute, Barcelona, Spain
| | - Anna Brugulat-Serrat
- Barcelonaβeta Brain Research Center, Pasqual Maragall Foundation, Barcelona, Spain; Neuroimagen de Enfermedades Neurodegenerativas y Envejecimiento Saludable, Hospital del Mar Research Institute, Barcelona, Spain; CIBER Fragilidad y Envejecimiento Saludable, Instituto de Salud Carlos III, Madrid, Spain; Global Brain Health Institute, San Francisco, CA, USA
| | - Gonzalo Sánchez-Benavides
- Barcelonaβeta Brain Research Center, Pasqual Maragall Foundation, Barcelona, Spain; Neuroimagen de Enfermedades Neurodegenerativas y Envejecimiento Saludable, Hospital del Mar Research Institute, Barcelona, Spain; CIBER Fragilidad y Envejecimiento Saludable, Instituto de Salud Carlos III, Madrid, Spain
| | - Armand González-Escalante
- Barcelonaβeta Brain Research Center, Pasqual Maragall Foundation, Barcelona, Spain; Biomedicine, Universitat Pompeu Fabra, Barcelona, Spain; Neuroimagen de Enfermedades Neurodegenerativas y Envejecimiento Saludable, Hospital del Mar Research Institute, Barcelona, Spain
| | - Federica Anastasi
- Barcelonaβeta Brain Research Center, Pasqual Maragall Foundation, Barcelona, Spain; Neuroimagen de Enfermedades Neurodegenerativas y Envejecimiento Saludable, Hospital del Mar Research Institute, Barcelona, Spain; Centre for Genomic Regulation, Barcelona Institute of Science and Technology, Barcelona, Spain
| | - Marta Milà-Alomà
- Barcelonaβeta Brain Research Center, Pasqual Maragall Foundation, Barcelona, Spain; Department of Veterans Affairs Medical Center, Northern California Institute for Research and Education, San Francisco, CA, USA; Department of Radiology, University of California, San Francisco, CA, USA
| | - David López-Martos
- Barcelonaβeta Brain Research Center, Pasqual Maragall Foundation, Barcelona, Spain; Neuroimagen de Enfermedades Neurodegenerativas y Envejecimiento Saludable, Hospital del Mar Research Institute, Barcelona, Spain
| | - Muge Akinci
- Barcelonaβeta Brain Research Center, Pasqual Maragall Foundation, Barcelona, Spain; Biomedicine, Universitat Pompeu Fabra, Barcelona, Spain; Neuroimagen de Enfermedades Neurodegenerativas y Envejecimiento Saludable, Hospital del Mar Research Institute, Barcelona, Spain; Barcelona Institute of Global Health, Barcelona, Spain
| | - Carles Falcón
- Barcelonaβeta Brain Research Center, Pasqual Maragall Foundation, Barcelona, Spain
| | - Mahnaz Shekari
- Barcelonaβeta Brain Research Center, Pasqual Maragall Foundation, Barcelona, Spain; Biomedicine, Universitat Pompeu Fabra, Barcelona, Spain; Neuroimagen de Enfermedades Neurodegenerativas y Envejecimiento Saludable, Hospital del Mar Research Institute, Barcelona, Spain
| | - Raffaele Cacciaglia
- Barcelonaβeta Brain Research Center, Pasqual Maragall Foundation, Barcelona, Spain; Neuroimagen de Enfermedades Neurodegenerativas y Envejecimiento Saludable, Hospital del Mar Research Institute, Barcelona, Spain; CIBER Fragilidad y Envejecimiento Saludable, Instituto de Salud Carlos III, Madrid, Spain
| | | | - Carolina Minguillón
- Barcelonaβeta Brain Research Center, Pasqual Maragall Foundation, Barcelona, Spain; Neuroimagen de Enfermedades Neurodegenerativas y Envejecimiento Saludable, Hospital del Mar Research Institute, Barcelona, Spain; CIBER Fragilidad y Envejecimiento Saludable, Instituto de Salud Carlos III, Madrid, Spain
| | - Karine Fauria
- Barcelonaβeta Brain Research Center, Pasqual Maragall Foundation, Barcelona, Spain; Neuroimagen de Enfermedades Neurodegenerativas y Envejecimiento Saludable, Hospital del Mar Research Institute, Barcelona, Spain; CIBER Fragilidad y Envejecimiento Saludable, Instituto de Salud Carlos III, Madrid, Spain
| | - José Luis Molinuevo
- Barcelonaβeta Brain Research Center, Pasqual Maragall Foundation, Barcelona, Spain; H Lundbeck, Copenhagen, Denmark
| | - Marc Suárez-Calvet
- Barcelonaβeta Brain Research Center, Pasqual Maragall Foundation, Barcelona, Spain; Neuroimagen de Enfermedades Neurodegenerativas y Envejecimiento Saludable, Hospital del Mar Research Institute, Barcelona, Spain; CIBER Fragilidad y Envejecimiento Saludable, Instituto de Salud Carlos III, Madrid, Spain; Servei de Neurologia, Hospital del Mar, Barcelona, Spain
| | - Oriol Grau-Rivera
- Barcelonaβeta Brain Research Center, Pasqual Maragall Foundation, Barcelona, Spain; Neuroimagen de Enfermedades Neurodegenerativas y Envejecimiento Saludable, Hospital del Mar Research Institute, Barcelona, Spain; CIBER Fragilidad y Envejecimiento Saludable, Instituto de Salud Carlos III, Madrid, Spain; Servei de Neurologia, Hospital del Mar, Barcelona, Spain
| | - Verónica Vilaplana
- Department of Signal Theory and Communications, Universitat Politècnica de Catalunya, Barcelona, Spain
| | - Juan Domingo Gispert
- Barcelonaβeta Brain Research Center, Pasqual Maragall Foundation, Barcelona, Spain; Neuroimagen de Enfermedades Neurodegenerativas y Envejecimiento Saludable, Hospital del Mar Research Institute, Barcelona, Spain; Centro de Investigación Biomédica en Red de Bioingeniería, Biomateriales y Nanomedicina, Instituto de Salud Carlos III, Madrid, Spain; Centro Nacional de Investigaciones Cardiovasculares, Madrid, Spain.
| |
Collapse
|
45
|
Seitz-Holland J, Haas SS, Penzel N, Reichenberg A, Pasternak O. BrainAGE, brain health, and mental disorders: A systematic review. Neurosci Biobehav Rev 2024; 159:105581. [PMID: 38354871 PMCID: PMC11119273 DOI: 10.1016/j.neubiorev.2024.105581] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2023] [Revised: 02/05/2024] [Accepted: 02/09/2024] [Indexed: 02/16/2024]
Abstract
The imaging-based method of brainAGE aims to characterize an individual's vulnerability to age-related brain changes. The present study systematically reviewed brainAGE findings in neuropsychiatric conditions and discussed the potential of brainAGE as a marker for biological age. A systematic PubMed search (from inception to March 6th, 2023) identified 273 articles. The 30 included studies compared brainAGE between neuropsychiatric and healthy groups (n≥50). We presented results qualitatively and adapted a bias risk assessment questionnaire. The imaging modalities, design, and input features varied considerably between studies. While the studies found higher brainAGE in neuropsychiatric conditions (11 mild cognitive impairment/ dementia, 11 schizophrenia spectrum/ other psychotic and bipolar disorder, six depression/ anxiety, two multiple groups), the associations with clinical characteristics were mixed. While brainAGE is sensitive to group differences, limitations include the lack of diverse training samples, multi-modal studies, and external validation. Only a few studies obtained longitudinal data, and all have used algorithms built solely to predict chronological age. These limitations impede the validity of brainAGE as a biological age marker.
Collapse
Affiliation(s)
- Johanna Seitz-Holland
- Department of Psychiatry, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA; Department of Psychiatry, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA.
| | - Shalaila S Haas
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Nora Penzel
- Department of Psychiatry, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Abraham Reichenberg
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Department of Environmental Medicine and Public Health, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Mindich Child Health and Development Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Ofer Pasternak
- Department of Psychiatry, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA; Department of Psychiatry, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
46
|
Sacca V, Chai-Zhang TC, Hodges S, Amores J, Guler S, Todorova N, McDonald CM, Ge T, Kong J. Morphological changes of the limbic system associated with acute and chronic low-back pain: A UK biobank imaging study. Eur J Pain 2024; 28:608-619. [PMID: 38009393 PMCID: PMC10947961 DOI: 10.1002/ejp.2206] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2023] [Revised: 09/29/2023] [Accepted: 11/01/2023] [Indexed: 11/28/2023]
Abstract
BACKGROUND Low back pain (LBP) is a major public health issue that influences physical and emotional factors integral to the limbic system. This study aims to investigate the association between LBP and brain morphometry alterations as the duration of LBP increases (acute vs. chronic). METHODS We used the UK Biobank data to investigate the morphological features of the limbic system in acute LBP (N = 115), chronic LBP (N = 243) and controls (N = 358), and tried to replicate our findings with an independent dataset composed of 45 acute LBP participants evaluated at different timepoints throughout 1 year from the OpenPain database. RESULTS We found that in comparison with chronic LBP and pain-free controls, acute LBP was associated with increased volumes of the nucleus accumbens, amygdala, hippocampus, and thalamus, and increased grey matter volumes in the hippocampus and posterior cingulate gyrus. In the replication cohort, we found non-significantly larger hippocampus and thalamus volumes in the 3-month visit (acute LBP) compared to the 1-year visit (chronic LBP), with similar effect sizes as the UK Biobank dataset. CONCLUSIONS Our results suggest that acute LBP is associated with dramatic morphometric increases in the limbic system and mesolimbic pathway, which may reflect an active brain response and self-regulation in the early stage of LBP. SIGNIFICANCE Our study suggests that LBP in the acute phase is associated with the brain morphometric changes (increase) in some limbic areas, indicating that the acute phase of LBP may represent a crucial stage of self-regulation and active response to the disease's onset.
Collapse
Affiliation(s)
- Valeria Sacca
- Department of Psychiatry, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA 02129, United States
| | - Thalia Celeste Chai-Zhang
- Department of Psychiatry, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA 02129, United States
| | - Sierra Hodges
- Department of Psychiatry, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA 02129, United States
| | - Judith Amores
- Department of Psychiatry, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA 02129, United States
| | - Seyhmus Guler
- Department of Psychiatry, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA 02129, United States
| | - Nevyana Todorova
- Department of Psychiatry, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA 02129, United States
| | - Caroline Merritt McDonald
- Department of Psychiatry, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA 02129, United States
| | - Tian Ge
- Department of Psychiatry, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA 02129, United States
| | - Jian Kong
- Department of Psychiatry, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA 02129, United States
| |
Collapse
|
47
|
Hartmann S, Cearns M, Pantelis C, Dwyer D, Cavve B, Byrne E, Scott I, Yuen HP, Gao C, Allott K, Lin A, Wood SJ, Wigman JTW, Amminger GP, McGorry PD, Yung AR, Nelson B, Clark SR. Combining Clinical With Cognitive or Magnetic Resonance Imaging Data for Predicting Transition to Psychosis in Ultra High-Risk Patients: Data From the PACE 400 Cohort. BIOLOGICAL PSYCHIATRY. COGNITIVE NEUROSCIENCE AND NEUROIMAGING 2024; 9:417-428. [PMID: 38052267 DOI: 10.1016/j.bpsc.2023.11.009] [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: 07/27/2023] [Revised: 10/19/2023] [Accepted: 11/26/2023] [Indexed: 12/07/2023]
Abstract
BACKGROUND Multimodal modeling that combines biological and clinical data shows promise in predicting transition to psychosis in individuals who are at ultra-high risk. Individuals who transition to psychosis are known to have deficits at baseline in cognitive function and reductions in gray matter volume in multiple brain regions identified by magnetic resonance imaging. METHODS In this study, we used Cox proportional hazards regression models to assess the additive predictive value of each modality-cognition, cortical structure information, and the neuroanatomical measure of brain age gap-to a previously developed clinical model using functioning and duration of symptoms prior to service entry as predictors in the Personal Assessment and Crisis Evaluation (PACE) 400 cohort. The PACE 400 study is a well-characterized cohort of Australian youths who were identified as ultra-high risk of transitioning to psychosis using the Comprehensive Assessment of At Risk Mental States (CAARMS) and followed for up to 18 years; it contains clinical data (from N = 416 participants), cognitive data (n = 213), and magnetic resonance imaging cortical parameters extracted using FreeSurfer (n = 231). RESULTS The results showed that neuroimaging, brain age gap, and cognition added marginal predictive information to the previously developed clinical model (fraction of new information: neuroimaging 0%-12%, brain age gap 7%, cognition 0%-16%). CONCLUSIONS In summary, adding a second modality to a clinical risk model predicting the onset of a psychotic disorder in the PACE 400 cohort showed little improvement in the fit of the model for long-term prediction of transition to psychosis.
Collapse
Affiliation(s)
- Simon Hartmann
- Discipline of Psychiatry, Adelaide Medical School, The University of Adelaide, Adelaide, South Australia, Australia; Orygen, Parkville, Victoria, Australia; Centre for Youth Mental Health, The University of Melbourne, Melbourne, Victoria, Australia.
| | - Micah Cearns
- Discipline of Psychiatry, Adelaide Medical School, The University of Adelaide, Adelaide, South Australia, Australia
| | - Christos Pantelis
- Melbourne Neuropsychiatry Centre, Department of Psychiatry, The University of Melbourne, Carlton South, Melbourne, Victoria, Australia; Western Centre for Health Research & Education, Western Hospital Sunshine, The University of Melbourne, St. Albans, Victoria, Australia
| | - Dominic Dwyer
- Orygen, Parkville, Victoria, Australia; Centre for Youth Mental Health, The University of Melbourne, Melbourne, Victoria, Australia
| | - Blake Cavve
- Telethon Kids Institute, The University of Western Australia, Perth, Western Australia, Australia
| | - Enda Byrne
- Child Health Research Center, The University of Queensland, Brisbane, Queensland, Australia
| | - Isabelle Scott
- Orygen, Parkville, Victoria, Australia; Centre for Youth Mental Health, The University of Melbourne, Melbourne, Victoria, Australia
| | - Hok Pan Yuen
- Orygen, Parkville, Victoria, Australia; Centre for Youth Mental Health, The University of Melbourne, Melbourne, Victoria, Australia
| | - Caroline Gao
- Orygen, Parkville, Victoria, Australia; Centre for Youth Mental Health, The University of Melbourne, Melbourne, Victoria, Australia
| | - Kelly Allott
- Orygen, Parkville, Victoria, Australia; Centre for Youth Mental Health, The University of Melbourne, Melbourne, Victoria, Australia
| | - Ashleigh Lin
- Telethon Kids Institute, The University of Western Australia, Perth, Western Australia, Australia
| | - Stephen J Wood
- Orygen, Parkville, Victoria, Australia; Centre for Youth Mental Health, The University of Melbourne, Melbourne, Victoria, Australia; School of Psychology, The University of Birmingham, Birmingham, England, United Kingdom
| | - Johanna T W Wigman
- Department of Psychiatry, Interdisciplinary Center Psychopathology and Emotion Regulation, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - G Paul Amminger
- Orygen, Parkville, Victoria, Australia; Centre for Youth Mental Health, The University of Melbourne, Melbourne, Victoria, Australia
| | - Patrick D McGorry
- Orygen, Parkville, Victoria, Australia; Centre for Youth Mental Health, The University of Melbourne, Melbourne, Victoria, Australia
| | - Alison R Yung
- Institute for Mental and Physical Health and Clinical Translation, Deakin University, Melbourne, Victoria, Australia
| | - Barnaby Nelson
- Orygen, Parkville, Victoria, Australia; Centre for Youth Mental Health, The University of Melbourne, Melbourne, Victoria, Australia
| | - Scott R Clark
- Discipline of Psychiatry, Adelaide Medical School, The University of Adelaide, Adelaide, South Australia, Australia
| |
Collapse
|
48
|
Kamarajan C, Ardekani BA, Pandey AK, Meyers JL, Chorlian DB, Kinreich S, Pandey G, Richard C, de Viteri SS, Kuang W, Porjesz B. Prediction of brain age in individuals with and at risk for alcohol use disorder using brain morphological features. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.01.582844. [PMID: 38496639 PMCID: PMC10942318 DOI: 10.1101/2024.03.01.582844] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/19/2024]
Abstract
Brain age measures predicted from structural and functional brain features are increasingly being used to understand brain integrity, disorders, and health. While there is a vast literature showing aberrations in both structural and functional brain measures in individuals with and at risk for alcohol use disorder (AUD), few studies have investigated brain age in these groups. The current study examines brain age measures predicted using brain morphological features, such as cortical thickness and brain volume, in individuals with a lifetime diagnosis of AUD as well as in those at higher risk to develop AUD from families with multiple members affected with AUD (i.e., higher family history density (FHD) scores). The AUD dataset included a group of 30 adult males (mean age = 41.25 years) with a lifetime diagnosis of AUD and currently abstinent and a group of 30 male controls (mean age = 27.24 years) without any history of AUD. A second dataset of young adults who were categorized based on their FHD scores comprised a group of 40 individuals (20 males) with high FHD of AUD (mean age = 25.33 years) and a group of 31 individuals (18 males) with low FHD (mean age = 25.47 years). Brain age was predicted using 187 brain morphological features of cortical thickness and brain volume in an XGBoost regression model; a bias-correction procedure was applied to the predicted brain age. Results showed that both AUD and high FHD individuals showed an increase of 1.70 and 0.09 years (1.08 months), respectively, in their brain age relative to their chronological age, suggesting accelerated brain aging in AUD and risk for AUD. Increased brain age was associated with poor performance on neurocognitive tests of executive functioning in both AUD and high FHD individuals, indicating that brain age can also serve as a proxy for cognitive functioning and brain health. These findings on brain aging in these groups may have important implications for the prevention and treatment of AUD and ensuing cognitive decline.
Collapse
Affiliation(s)
- Chella Kamarajan
- Henri Begleiter Neurodynamics Lab, SUNY Downstate Health Sciences University, Brooklyn, NY 11203, USA
| | - Babak A. Ardekani
- Center for Advanced Brain Imaging, Nathan Kline Institute for Psychiatric Research, Orangeburg, NY 10962, USA
- Department of Psychiatry, Grossman School of Medicine, New York University, New York, NY 10016, USA
| | - Ashwini K. Pandey
- Henri Begleiter Neurodynamics Lab, SUNY Downstate Health Sciences University, Brooklyn, NY 11203, USA
| | - Jacquelyn L. Meyers
- Henri Begleiter Neurodynamics Lab, SUNY Downstate Health Sciences University, Brooklyn, NY 11203, USA
| | - David B. Chorlian
- Henri Begleiter Neurodynamics Lab, SUNY Downstate Health Sciences University, Brooklyn, NY 11203, USA
| | - Sivan Kinreich
- Henri Begleiter Neurodynamics Lab, SUNY Downstate Health Sciences University, Brooklyn, NY 11203, USA
| | - Gayathri Pandey
- Henri Begleiter Neurodynamics Lab, SUNY Downstate Health Sciences University, Brooklyn, NY 11203, USA
| | - Christian Richard
- Henri Begleiter Neurodynamics Lab, SUNY Downstate Health Sciences University, Brooklyn, NY 11203, USA
| | - Stacey Saenz de Viteri
- Henri Begleiter Neurodynamics Lab, SUNY Downstate Health Sciences University, Brooklyn, NY 11203, USA
| | - Weipeng Kuang
- Henri Begleiter Neurodynamics Lab, SUNY Downstate Health Sciences University, Brooklyn, NY 11203, USA
| | - Bernice Porjesz
- Henri Begleiter Neurodynamics Lab, SUNY Downstate Health Sciences University, Brooklyn, NY 11203, USA
| |
Collapse
|
49
|
Constantinides C, Baltramonaityte V, Caramaschi D, Han LKM, Lancaster TM, Zammit S, Freeman TP, Walton E. Assessing the association between global structural brain age and polygenic risk for schizophrenia in early adulthood: A recall-by-genotype study. Cortex 2024; 172:1-13. [PMID: 38154374 DOI: 10.1016/j.cortex.2023.11.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Revised: 09/22/2023] [Accepted: 11/23/2023] [Indexed: 12/30/2023]
Abstract
Neuroimaging studies consistently show advanced brain age in schizophrenia, suggesting that brain structure is often 'older' than expected at a given chronological age. Whether advanced brain age is linked to genetic liability for schizophrenia remains unclear. In this pre-registered secondary data analysis, we utilised a recall-by-genotype approach applied to a population-based subsample from the Avon Longitudinal Study of Parents and Children to assess brain age differences between young adults aged 21-24 years with relatively high (n = 96) and low (n = 93) polygenic risk for schizophrenia (SCZ-PRS). A global index of brain age (or brain-predicted age) was estimated using a publicly available machine learning model previously trained on a combination of region-wise gray-matter measures, including cortical thickness, surface area and subcortical volumes derived from T1-weighted magnetic resonance imaging (MRI) scans. We found no difference in mean brain-PAD (the difference between brain-predicted age and chronological age) between the high- and low-SCZ-PRS groups, controlling for the effects of sex and age at time of scanning (b = -.21; 95% CI -2.00, 1.58; p = .82; Cohen's d = -.034; partial R2 = .00029). These findings do not support an association between SCZ-PRS and brain-PAD based on global age-related structural brain patterns, suggesting that brain age may not be a vulnerability marker of common genetic risk for SCZ. Future studies with larger samples and multimodal brain age measures could further investigate global or localised effects of SCZ-PRS.
Collapse
Affiliation(s)
| | | | - Doretta Caramaschi
- Department of Psychology, Faculty of Health and Life Sciences, University of Exeter, UK
| | - Laura K M Han
- Centre for Youth Mental Health, The University of Melbourne, Melbourne, Australia; Orygen, Parkville, Australia
| | | | - Stanley Zammit
- Division of Psychological Medicine and Clinical Neurosciences, Cardiff University, Cardiff, UK; Centre for Academic Mental Health, Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
| | - Tom P Freeman
- Addiction and Mental Health Group (AIM), Department of Psychology, University of Bath, UK
| | | |
Collapse
|
50
|
A. Shirsath M, O'Connor JD, Boyle R, Newman L, Knight SP, Hernandez B, Whelan R, Meaney JF, Kenny RA. Slower speed of blood pressure recovery after standing is associated with accelerated brain aging: Evidence from The Irish Longitudinal Study on Ageing (TILDA). CEREBRAL CIRCULATION - COGNITION AND BEHAVIOR 2024; 6:100212. [PMID: 38445293 PMCID: PMC10912350 DOI: 10.1016/j.cccb.2024.100212] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/11/2023] [Revised: 01/17/2024] [Accepted: 02/02/2024] [Indexed: 03/07/2024]
Abstract
Background Impaired recovery of blood pressure (BP) in response to standing up is a prevalent condition in older individuals. We evaluated the relationship between the early recovery of hemodynamic responses to standing and brain health in adults over 50. Methods Participants from The Irish Longitudinal Study on Ageing (TILDA) (n=411; age 67.6 ± 7.3 years; 53.4 % women) performed an active stand challenge while blood pressure and heart rate were continuously monitored. The recovery of these parameters was determined as the slope of the BP and HR response, following the initial drop/rise after standing. We have previously reported a novel and validated measure of brain ageing using MRI data, which measures the difference between biological brain age and chronological age, providing a brain-predicted age difference (brainPAD) score. Results Slower recovery of systolic and diastolic BP was found to be significantly associated with higher brainPAD scores (i.e., biologically older brains), where a one-year increase in brainPAD was associated with a decrease of 0.02 mmHg/s and 0.01 mmHg/s in systolic and diastolic BP recovery, respectively, after standing. Heart rate (HR) recovery was not significantly associated with brainPAD score. Conclusion These results demonstrate that slower systolic and diastolic BP recovery in the early phase after standing is associated with accelerated brain aging in older individuals. This suggests that the BP response to standing, measured using beat-to-beat monitoring, has the potential to be used as a marker of accelerated brain aging, relying on a simple procedure and devices that are easily accessible.
Collapse
Affiliation(s)
- Morgana A. Shirsath
- The Irish Longitudinal Study on Ageing (TILDA), School of Medicine, Trinity College, University of, Ireland
| | - John D. O'Connor
- The Irish Longitudinal Study on Ageing (TILDA), School of Medicine, Trinity College, University of, Ireland
- School of Engineering, Ulster University, Northern Ireland, UK
| | - Rory Boyle
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, USA
| | - Louise Newman
- The Irish Longitudinal Study on Ageing (TILDA), School of Medicine, Trinity College, University of, Ireland
| | - Silvin P. Knight
- The Irish Longitudinal Study on Ageing (TILDA), School of Medicine, Trinity College, University of, Ireland
| | - Belinda Hernandez
- The Irish Longitudinal Study on Ageing (TILDA), School of Medicine, Trinity College, University of, Ireland
| | - Robert Whelan
- Trinity College Institute of Neuroscience, Trinity College, University of Dublin, Ireland
- Global Brain Health Institute, Trinity College, Trinity College Dublin, Ireland
| | - James F. Meaney
- National Centre for Advanced Medical Imaging (CAMI), St. James's Hospital, Dublin, Ireland
| | - Rose Anne Kenny
- The Irish Longitudinal Study on Ageing (TILDA), School of Medicine, Trinity College, University of, Ireland
| |
Collapse
|