1
|
Giacomel A, Martins D, Nordio G, Easmin R, Howes O, Selvaggi P, Williams SCR, Turkheimer F, De Groot M, Dipasquale O, Veronese M. Investigating dopaminergic abnormalities in schizophrenia and first-episode psychosis with normative modelling and multisite molecular neuroimaging. Mol Psychiatry 2025:10.1038/s41380-025-02938-w. [PMID: 40021831 DOI: 10.1038/s41380-025-02938-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/21/2023] [Revised: 01/09/2025] [Accepted: 02/19/2025] [Indexed: 03/03/2025]
Abstract
Molecular neuroimaging techniques, like PET and SPECT, offer invaluable insights into the brain's in-vivo biology and its dysfunction in neuropsychiatric patients. However, the transition of molecular neuroimaging into diagnostics and precision medicine has been limited to a few clinical applications, hindered by issues like practical feasibility, high costs, and high between-subject heterogeneity of neuroimaging measures. In this study, we explore the use of normative modelling (NM) to identify individual patient alterations by describing the physiological variability of molecular functions. NM potentially addresses challenges such as small sample sizes and diverse acquisition protocols typical of molecular neuroimaging studies. We applied NM to two PET radiotracers targeting the dopaminergic system ([11C]-(+)-PHNO and [18F]FDOPA) to create a reference-cohort model of healthy controls. The models were subsequently utilized on different independent cohorts of patients with psychosis in different disease stages and treatment outcomes. Our results showed that patients with psychosis exhibited a higher degree of extreme deviations (~3-fold increase) than controls, although this pattern was heterogeneous, with minimal overlap of extreme deviations topology (max 20%). We also confirmed that striatal [18F]FDOPA signal, when referenced to a normative distribution, can predict treatment response (striatal AUC ROC: 0.77-0.83). In conclusion, our results indicate that normative modelling can be effectively applied to molecular neuroimaging after proper harmonization, enabling insights into disease mechanisms and advancing precision medicine. In addition, the method is valuable in understanding the heterogeneity of patient populations and can contribute to maximising cost efficiency in studies aimed at comparing cases and controls.
Collapse
Affiliation(s)
- Alessio Giacomel
- Centre for Neuroimaging Sciences, Institute of Psychology, Psychiatry and Neuroscience (IoPPN), King's College London, London, UK.
| | - Daniel Martins
- Centre for Neuroimaging Sciences, Institute of Psychology, Psychiatry and Neuroscience (IoPPN), King's College London, London, UK
- Division of Adult Psychiatry, Department of Psychiatry, Geneva University Hospitals, Rue Gabrielle Perret-Gentil 4, 1205, Geneva, Switzerland
| | - Giovanna Nordio
- Centre for Neuroimaging Sciences, Institute of Psychology, Psychiatry and Neuroscience (IoPPN), King's College London, London, UK
| | - Rubaida Easmin
- Centre for Neuroimaging Sciences, Institute of Psychology, Psychiatry and Neuroscience (IoPPN), King's College London, London, UK
| | - Oliver Howes
- Department of Psychosis Studies, Institute of Psychology, Psychiatry and Neuroscience (IoPPN), King's College London, London, UK
- MRC Laboratory of Medical Science, Imperial College London, London, UK
- South London and Maudsley NHS Foundation Trust, London, UK
| | - Pierluigi Selvaggi
- Centre for Neuroimaging Sciences, Institute of Psychology, Psychiatry and Neuroscience (IoPPN), King's College London, London, UK
- Department of Translational Biomedicine and Neuroscience, University of Bari "Aldo Moro", Bari, Italy
| | - Steven C R Williams
- Centre for Neuroimaging Sciences, Institute of Psychology, Psychiatry and Neuroscience (IoPPN), King's College London, London, UK
| | - Federico Turkheimer
- Centre for Neuroimaging Sciences, Institute of Psychology, Psychiatry and Neuroscience (IoPPN), King's College London, London, UK
| | - Marius De Groot
- GSK R&D, Clinical Pharmacology and Experimental Medicine, Clinical Imaging, Stevenage, UK
| | - Ottavia Dipasquale
- Centre for Neuroimaging Sciences, Institute of Psychology, Psychiatry and Neuroscience (IoPPN), King's College London, London, UK
| | - Mattia Veronese
- Centre for Neuroimaging Sciences, Institute of Psychology, Psychiatry and Neuroscience (IoPPN), King's College London, London, UK.
- Department of Information Engineering, University of Padova, Padova, Italy.
| |
Collapse
|
2
|
Teipel SJ, Hoffmann H, Storch A, Hermann A, Dyrba M, Schumacher J. Brain age in genetic and idiopathic Parkinson's disease. Brain Commun 2024; 6:fcae382. [PMID: 39713239 PMCID: PMC11660940 DOI: 10.1093/braincomms/fcae382] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Revised: 09/02/2024] [Accepted: 12/18/2024] [Indexed: 12/24/2024] Open
Abstract
The brain-age gap, i.e. the difference between the brain age estimated from structural MRI data and the chronological age of an individual, has been proposed as a summary measure of brain integrity in neurodegenerative diseases. Here, we aimed to determine the brain-age gap in genetic and idiopathic Parkinson's disease and its association with surrogate markers of Alzheimer's disease and Parkinson's disease pathology and with rates of cognitive and motor function decline. We studied 1200 cases from the Parkinson's Progression Markers Initiative cohort, including idiopathic Parkinson's disease, asymptomatic and clinical mutation carriers in the leucine-rich repeat kinase 2 gene (LRRK2) and the glucocerebrosidase gene (GBA), and normal controls using a cohort study design. For comparison, we studied 187 Alzheimer's disease dementia cases and 254 controls from the Alzheimer's Disease Neuroimaging Initiative cohort. We used Bayesian ANOVA to determine associations of the brain-age gap with diagnosis, and baseline measures of motor and cognitive function, dopamine transporter activity and CSF markers of Alzheimer's disease type amyloid-β42 and phosphotau pathology. Associations of brain-age gap with rates of cognitive and motor function decline were determined using Bayesian generalized mixed effect models. The brain-age gap in idiopathic Parkinson's disease patients was 0.7 years compared to controls, but 5.9 years in Alzheimer's disease dementia cases. In contrast, asymptomatic LRRK2 individuals had a 1.1. year younger brain age than controls. Across all cases, the brain-age gap was associated with motor impairment and (in the clinically manifest PD cases) reduced dopamine transporter activity, but less with CSF amyloid-β42 and phosphotau. In idiopathic Parkinson's disease cases, however, the brain-age gap was associated with lower CSF amyloid-β42 levels. In sporadic and genetic Parkinson's disease cases, a higher brain-age gap was associated with faster decline in episodic memory, and executive and motor function, whereas in asymptomatic LRRK2 cases, a smaller brain-age gap was associated with faster cognitive decline. In conclusion, brain age was sensitive to Alzheimer's disease like rather than Parkinson's disease like brain atrophy. Once an individual had idiopathic Parkinson's disease, their brain age was associated with markers of Alzheimer's disease rather than Parkinson's disease. Asymptomatic LRRK2 cases had seemingly younger brains than controls, and in these cases, younger brain age was associated with poorer cognitive outcome. This suggests that the term brain age is misleading when applied to disease stages where reactive brain changes with apparent volume increases rather than atrophy may drive the calculation of the brain age.
Collapse
Affiliation(s)
- Stefan J Teipel
- Deutsches Zentrum für Neurodegenerative Erkrankungen (DZNE) Rostock-Greifswald, Rostock 18147, Germany
- Department of Psychosomatic Medicine, University Medical Center Rostock, Rostock 18147, Germany
| | - Hauke Hoffmann
- Department of Psychosomatic Medicine, University Medical Center Rostock, Rostock 18147, Germany
| | - Alexander Storch
- Deutsches Zentrum für Neurodegenerative Erkrankungen (DZNE) Rostock-Greifswald, Rostock 18147, Germany
- Department of Neurology, University Medical Center Rostock, Rostock 18147, Germany
| | - Andreas Hermann
- Deutsches Zentrum für Neurodegenerative Erkrankungen (DZNE) Rostock-Greifswald, Rostock 18147, Germany
- Department of Neurology, University Medical Center Rostock, Rostock 18147, Germany
- Translational Neurodegeneration Section ‘Albrecht Kossel’, Department of Neurology, University Medical Center Rostock, Rostock 18147, Germany
| | - Martin Dyrba
- Deutsches Zentrum für Neurodegenerative Erkrankungen (DZNE) Rostock-Greifswald, Rostock 18147, Germany
| | - Julia Schumacher
- Deutsches Zentrum für Neurodegenerative Erkrankungen (DZNE) Rostock-Greifswald, Rostock 18147, Germany
- Department of Neurology, University Medical Center Rostock, Rostock 18147, Germany
| |
Collapse
|
3
|
Chai L, Sun J, Zhuo Z, Wei R, Xu X, Duan Y, Tian D, Bai Y, Zhang N, Li H, Li Y, Li Y, Zhou F, Xu J, Cole JH, Barkhof F, Zhang J, Zheng H, Liu Y. Estimated Brain Age in Healthy Aging and Across Multiple Neurological Disorders. J Magn Reson Imaging 2024. [PMID: 39588683 DOI: 10.1002/jmri.29667] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2024] [Revised: 11/13/2024] [Accepted: 11/13/2024] [Indexed: 11/27/2024] Open
Abstract
BACKGROUND The brain aging in the general population and patients with neurological disorders is not well understood. PURPOSE To characterize brain aging in the above conditions and its clinical relevance. STUDY TYPE Retrospective. POPULATION A total of 2913 healthy controls (HC), with 1395 females; 331 multiple sclerosis (MS); 189 neuromyelitis optica spectrum disorder (NMOSD); 239 Alzheimer's disease (AD); 244 Parkinson's disease (PD); and 338 cerebral small vessel disease (cSVD). FIELD STRENGTH/SEQUENCE 3.0 T/Three-dimensional (3D) T1-weighted images. ASSESSMENT The brain age was estimated by our previously developed model, using a 3D convolutional neural network trained on 9794 3D T1-weighted images of healthy individuals. Brain age gap (BAG), the difference between chronological age and estimated brain age, was calculated to represent accelerated and resilient brain conditions. We compared MRI metrics between individuals with accelerated (BAG ≥ 5 years) and resilient brain age (BAG ≤ -5 years) in HC, and correlated BAG with MRI metrics, and cognitive and physical measures across neurological disorders. STATISTICAL TESTS Student's t test, Wilcoxon test, chi-square test or Fisher's exact test, and correlation analysis. P < 0.05 was considered statistically significant. RESULTS In HC, individuals with accelerated brain age exhibited significantly higher white matter hyperintensity (WMH) and lower regional brain volumes than those with resilient brain age. BAG was significantly higher in MS (10.30 ± 12.6 years), NMOSD (2.96 ± 7.8 years), AD (6.50 ± 6.6 years), PD (4.24 ± 4.8 years), and cSVD (3.24 ± 5.9 years) compared to HC. Increased BAG was significantly associated with regional brain atrophy, WMH burden, and cognitive impairment across neurological disorders. Increased BAG was significantly correlated with physical disability in MS (r = 0.17). DATA CONCLUSION Healthy individuals with accelerated brain age show high WMH burden and regional volume reduction. Neurological disorders exhibit distinct accelerated brain aging, correlated with impaired cognitive and physical function. LEVEL OF EVIDENCE 4 TECHNICAL EFFICACY: Stage 2.
Collapse
Affiliation(s)
- Li Chai
- Department of Radiology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Jun Sun
- Department of Radiology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Zhizheng Zhuo
- Department of Radiology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Ren Wei
- Department of Radiology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Xiaolu Xu
- Department of Radiology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Yunyun Duan
- Department of Radiology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Decai Tian
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Yutong Bai
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Ningnannan Zhang
- Department of Radiology and Tianjin Key Laboratory of Functional Imaging, Tianjin Medical University General Hospital, Tianjin, China
| | - Haiqing Li
- Radiology Department, Huashan Hospital, Fudan University, Shanghai, China
| | - Yuxin Li
- Radiology Department, Huashan Hospital, Fudan University, Shanghai, China
| | - Yongmei Li
- Department of Radiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Fuqing Zhou
- Department of Radiology, the First Affiliated Hospital, Nanchang University, Nanchang, China
| | - Jun Xu
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - James H Cole
- Centre for Medical Image Computing, University College London, London, UK
- Dementia Research Centre, Queen Square Institute of Neurology, University College London, London, UK
| | - Frederik Barkhof
- Dementia Research Centre, Queen Square Institute of Neurology, University College London, London, UK
- Department of Radiology and Nuclear Medicine, Neuroscience Campus Amsterdam, VU University Medical Center, Amsterdam, The Netherlands
| | - Jianguo Zhang
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Huaguang Zheng
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Yaou Liu
- Department of Radiology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
4
|
Pang H, Yu Z, Yu H, Li X, Bu S, Liu Y, Wang J, Zhao M, Fan G. Advanced Cognitive Patterns in Multiple System Atrophy Compared to Parkinson's Disease: An Individual Diffusion Tensor Imaging Study. Acad Radiol 2024; 31:2897-2909. [PMID: 38220569 DOI: 10.1016/j.acra.2024.01.006] [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: 11/09/2023] [Revised: 12/27/2023] [Accepted: 01/04/2024] [Indexed: 01/16/2024]
Abstract
RATIONALE AND OBJECTIVES Although both Multiple system atrophy (MSA) and Parkinson's disease (PD) belong to alpha-synucleinopathy, they have divergent clinical courses and prognoses. The degeneration of white matter has a considerable impact on cognitive performance, yet it is uncertain how PD and MSA affect its functioning in a similar or different manner. METHODS In this study, a total of 116 individuals (37 PD with mild cognitive impairment (PD-MCI), 37 MSA (parkinsonian variant) with mild cognitive impairment (MSA-MCI), and 42 healthy controls) underwent diffusion tensor imaging (DTI) and cognitive assessment. Utilizing probabilistic fiber tracking, association fibers, projection fibers, and thalamic fibers were reconstructed. Subsequently, regression, support vector machine, and SHAP (Shapley Addictive exPlanations) analyzes were conducted to evaluate the association between microstructural diffusion metrics and multiple cognitive domains, thus determining the white matter predictors of MCI. RESULTS MSA-MCI patients exhibited distinct white matter impairment extending to the middle cerebellar peduncle, corticospinal tract, and cingulum bundle. Furthermore, the fractional anisotropy (FA) and mean diffusivity (MD)values of the right anterior thalamic radiation were significantly associated with global efficiency (FA: B = 0.69, P < 0.001, VIF = 1.31; MD: B = -0.53, P = 0.02, VIF = 2.50). The diffusion metrics of white matter between PD-MCI and MSA-MCI proved to be an effective predictor of the MCI, with an accuracy of 0.73 (P < 0.01), and the most predictive factor being the MD of the anterior thalamic radiation. CONCLUSIONS Our results demonstrated that MSA-MCI had a more noticeable deterioration in white matter, which potentially linked to various cognitive domain connections. Diffusion MRI could be a useful tool in comprehending the neurological basis of cognitive impairment in Parkinsonian disorders.
Collapse
Affiliation(s)
- Huize Pang
- Department of Radiology, First Affiliated Hospital of China Medical University, Shenyang, Liaoning Province, China (H.P., Z.Y., X.L., S.B., Y.L., J.W., M.Z., G.F.)
| | - Ziyang Yu
- Department of Radiology, First Affiliated Hospital of China Medical University, Shenyang, Liaoning Province, China (H.P., Z.Y., X.L., S.B., Y.L., J.W., M.Z., G.F.)
| | - Hongmei Yu
- Department of Neurology, First Affiliated Hospital of China Medical University, Shenyang, Liaoning Province, China (H.Y.)
| | - Xiaolu Li
- Department of Radiology, First Affiliated Hospital of China Medical University, Shenyang, Liaoning Province, China (H.P., Z.Y., X.L., S.B., Y.L., J.W., M.Z., G.F.)
| | - Shuting Bu
- Department of Radiology, First Affiliated Hospital of China Medical University, Shenyang, Liaoning Province, China (H.P., Z.Y., X.L., S.B., Y.L., J.W., M.Z., G.F.)
| | - Yu Liu
- Department of Radiology, First Affiliated Hospital of China Medical University, Shenyang, Liaoning Province, China (H.P., Z.Y., X.L., S.B., Y.L., J.W., M.Z., G.F.)
| | - Juzhou Wang
- Department of Radiology, First Affiliated Hospital of China Medical University, Shenyang, Liaoning Province, China (H.P., Z.Y., X.L., S.B., Y.L., J.W., M.Z., G.F.)
| | - Mengwan Zhao
- Department of Radiology, First Affiliated Hospital of China Medical University, Shenyang, Liaoning Province, China (H.P., Z.Y., X.L., S.B., Y.L., J.W., M.Z., G.F.)
| | - Guoguang Fan
- Department of Radiology, First Affiliated Hospital of China Medical University, Shenyang, Liaoning Province, China (H.P., Z.Y., X.L., S.B., Y.L., J.W., M.Z., G.F.).
| |
Collapse
|
5
|
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
|
6
|
Chen CL, Cheng SY, Montaser-Kouhsari L, Wu WC, Hsu YC, Tai CH, Tseng WYI, Kuo MC, Wu RM. Advanced brain aging in Parkinson's disease with cognitive impairment. NPJ Parkinsons Dis 2024; 10:62. [PMID: 38493188 PMCID: PMC10944471 DOI: 10.1038/s41531-024-00673-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2023] [Accepted: 02/29/2024] [Indexed: 03/18/2024] Open
Abstract
Patients with Parkinson's disease and cognitive impairment (PD-CI) deteriorate faster than those without cognitive impairment (PD-NCI), suggesting an underlying difference in the neurodegeneration process. We aimed to verify brain age differences in PD-CI and PD-NCI and their clinical significance. A total of 94 participants (PD-CI, n = 27; PD-NCI, n = 34; controls, n = 33) were recruited. Predicted age difference (PAD) based on gray matter (GM) and white matter (WM) features were estimated to represent the degree of brain aging. Patients with PD-CI showed greater GM-PAD (7.08 ± 6.64 years) and WM-PAD (8.82 ± 7.69 years) than those with PD-NCI (GM: 1.97 ± 7.13, Padjusted = 0.011; WM: 4.87 ± 7.88, Padjusted = 0.049) and controls (GM: -0.58 ± 7.04, Padjusted = 0.004; WM: 0.88 ± 7.45, Padjusted = 0.002) after adjusting demographic factors. In patients with PD, GM-PAD was negatively correlated with MMSE (Padjusted = 0.011) and MoCA (Padjusted = 0.013) and positively correlated with UPDRS Part II (Padjusted = 0.036). WM-PAD was negatively correlated with logical memory of immediate and delayed recalls (Padjusted = 0.003 and Padjusted < 0.001). Also, altered brain regions in PD-CI were identified and significantly correlated with brain age measures, implicating the neuroanatomical underpinning of neurodegeneration in PD-CI. Moreover, the brain age metrics can improve the classification between PD-CI and PD-NCI. The findings suggest that patients with PD-CI had advanced brain aging that was associated with poor cognitive functions. The identified neuroimaging features and brain age measures can serve as potential biomarkers of PD-CI.
Collapse
Affiliation(s)
- Chang-Le Chen
- Institute of Medical Device and Imaging, National Taiwan University College of Medicine, Taipei, Taiwan
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, USA
| | - Shao-Ying Cheng
- Department of Neurology, National Taiwan University Hospital Bei-Hu Branch, Taipei, Taiwan
| | | | - Wen-Chao Wu
- Institute of Medical Device and Imaging, National Taiwan University College of Medicine, Taipei, Taiwan
| | | | - Chun-Hwei Tai
- Department of Neurology, National Taiwan University Hospital, Taipei, Taiwan
| | - Wen-Yih Isaac Tseng
- Institute of Medical Device and Imaging, National Taiwan University College of Medicine, Taipei, Taiwan.
- Acroviz Inc, Taipei, Taiwan.
- Department of Medical Imaging, National Taiwan University Hospital, Taipei, Taiwan.
| | - Ming-Che Kuo
- Department of Neurology, National Taiwan University Hospital, Taipei, Taiwan.
- Department of Medicine, National Taiwan University Cancer Center, Taipei, Taiwan.
- Neurobiology and Cognitive Science Center, National Taiwan University, Taipei, Taiwan.
| | - Ruey-Meei Wu
- Department of Neurology, National Taiwan University Hospital, Taipei, Taiwan
| |
Collapse
|
7
|
Liu L, Lin L, Sun S, Wu S. Elucidating Multimodal Imaging Patterns in Accelerated Brain Aging: Heterogeneity through a Discriminant Analysis Approach Using the UK Biobank Dataset. Bioengineering (Basel) 2024; 11:124. [PMID: 38391610 PMCID: PMC10886122 DOI: 10.3390/bioengineering11020124] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Revised: 01/17/2024] [Accepted: 01/24/2024] [Indexed: 02/24/2024] Open
Abstract
Accelerated brain aging (ABA) intricately links with age-associated neurodegenerative and neuropsychiatric diseases, emphasizing the critical need for a nuanced exploration of heterogeneous ABA patterns. This investigation leveraged data from the UK Biobank (UKB) for a comprehensive analysis, utilizing structural magnetic resonance imaging (sMRI), diffusion magnetic resonance imaging (dMRI), and resting-state functional magnetic resonance imaging (rsfMRI) from 31,621 participants. Pre-processing employed tools from the FMRIB Software Library (FSL, version 5.0.10), FreeSurfer, DTIFIT, and MELODIC, seamlessly integrated into the UKB imaging processing pipeline. The Lasso algorithm was employed for brain-age prediction, utilizing derived phenotypes obtained from brain imaging data. Subpopulations of accelerated brain aging (ABA) and resilient brain aging (RBA) were delineated based on the error between actual age and predicted brain age. The ABA subgroup comprised 1949 subjects (experimental group), while the RBA subgroup comprised 3203 subjects (control group). Semi-supervised heterogeneity through discriminant analysis (HYDRA) refined and characterized the ABA subgroups based on distinctive neuroimaging features. HYDRA systematically stratified ABA subjects into three subtypes: SubGroup 2 exhibited extensive gray-matter atrophy, distinctive white-matter patterns, and unique connectivity features, displaying lower cognitive performance; SubGroup 3 demonstrated minimal atrophy, superior cognitive performance, and higher physical activity; and SubGroup 1 occupied an intermediate position. This investigation underscores pronounced structural and functional heterogeneity in ABA, revealing three subtypes and paving the way for personalized neuroprotective treatments for age-related neurological, neuropsychiatric, and neurodegenerative diseases.
Collapse
Affiliation(s)
- Lingyu Liu
- Department of Biomedical Engineering, College of Chemistry and Life Science, Beijing University of Technology, Beijing 100124, China
| | - Lan Lin
- Department of Biomedical Engineering, College of Chemistry and Life Science, Beijing University of Technology, Beijing 100124, China
- Intelligent Physiological Measurement and Clinical Translation, Beijing International Base for Scientific and Technological Cooperation, Beijing University of Technology, Beijing 100124, China
| | - Shen Sun
- Department of Biomedical Engineering, College of Chemistry and Life Science, Beijing University of Technology, Beijing 100124, China
- 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
- Intelligent Physiological Measurement and Clinical Translation, Beijing International Base for Scientific and Technological Cooperation, Beijing University of Technology, Beijing 100124, China
| |
Collapse
|
8
|
Tseng WYI, Hsu YC, Huang LK, Hong CT, Lu YH, Chen JH, Fu CK, Chan L. Brain Age Is Associated with Cognitive Outcomes of Cholinesterase Inhibitor Treatment in Patients with Mild Cognitive Impairment. J Alzheimers Dis 2024; 98:1095-1106. [PMID: 38517785 DOI: 10.3233/jad-231109] [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] [Indexed: 03/24/2024]
Abstract
Background The effect of cholinesterase inhibitor (ChEI) on mild cognitive impairment (MCI) is controversial. Brain age has been shown to predict Alzheimer's disease conversion from MCI. Objective The study aimed to show that brain age is related to cognitive outcomes of ChEI treatment in MCI. Methods Brain MRI, the Clinical Dementia Rating (CDR) and Mini-Mental State Exam (MMSE) scores were retrospectively retrieved from a ChEI treatment database. Patients who presented baseline CDR of 0.5 and received ChEI treatment for at least 2 years were selected. Patients with stationary or improved cognition as verified by the CDR and MMSE were categorized to the ChEI-responsive group, and those with worsened cognition were assigned to the ChEI-unresponsive group. A gray matter brain age model was built with a machine learning algorithm by training T1-weighted MRI data of 362 healthy participants. The model was applied to each patient to compute predicted age difference (PAD), i.e. the difference between brain age and chronological age. The PADs were compared between the two groups. Results 58 patients were found to fit the ChEI-responsive criteria in the patient data, and 58 matched patients that fit the ChEI-unresponsive criteria were compared. ChEI-unresponsive patients showed significantly larger PAD than ChEI-responsive patients (8.44±8.78 years versus 3.87±9.02 years, p = 0.0067). Conclusions Gray matter brain age is associated with cognitive outcomes after 2 years of ChEI treatment in patients with the CDR of 0.5. It might facilitate the clinical trials of novel therapeutics for MCI.
Collapse
Affiliation(s)
| | | | - Li-Kai Huang
- Department of Neurology, Taipei Medical University-Shuang Ho Hospital, Ministry of Health and Welfare, Taipei Medical University, New Taipei City, Taiwan (R.O.C.)
- Taipei Neuroscience Institute, Taipei Medical University, Taipei, Taiwan (R.O.C.)
| | - Chien-Tai Hong
- Department of Neurology, Taipei Medical University-Shuang Ho Hospital, Ministry of Health and Welfare, Taipei Medical University, New Taipei City, Taiwan (R.O.C.)
- Taipei Neuroscience Institute, Taipei Medical University, Taipei, Taiwan (R.O.C.)
- Department of Neurology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan (R.O.C.)
| | - Yueh-Hsun Lu
- Taipei Neuroscience Institute, Taipei Medical University, Taipei, Taiwan (R.O.C.)
- Department of Radiology, Shuang-Ho Hospital, Taipei Medical University, New Taipei City, Taiwan (R.O.C.)
- Department of Radiology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan (R.O.C.)
| | - Jia-Hung Chen
- Department of Neurology, Taipei Medical University-Shuang Ho Hospital, Ministry of Health and Welfare, Taipei Medical University, New Taipei City, Taiwan (R.O.C.)
- Taipei Neuroscience Institute, Taipei Medical University, Taipei, Taiwan (R.O.C.)
| | | | - Lung Chan
- Department of Neurology, Taipei Medical University-Shuang Ho Hospital, Ministry of Health and Welfare, Taipei Medical University, New Taipei City, Taiwan (R.O.C.)
- Taipei Neuroscience Institute, Taipei Medical University, Taipei, Taiwan (R.O.C.)
- Department of Neurology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan (R.O.C.)
| |
Collapse
|
9
|
Liang T, Wang X, Wang Y, Ma W. IFN-γ Triggered IFITM2 Expression to Induce Malignant Phenotype in Elderly GBM. J Mol Neurosci 2023; 73:946-955. [PMID: 37889394 DOI: 10.1007/s12031-023-02156-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2023] [Accepted: 09/07/2023] [Indexed: 10/28/2023]
Abstract
Advanced age is an important risk factor for the worse clinical presentation of gliomas, especially glioblastoma (GBM). The tumor microenvironment (TME) in elderly GBM (eGBM) patients is considerably different from that in young ones, which causes the inferior clinical outcome. Based on the data from the Chinese Glioma Genome Atlas RNA sequence (CGGA RNA-seq), the Cancer Genome Atlas RNA array (TCGA RNA-array), and gene set enrichment (GSE) 16011 array sets, the differential genes and function between eGBM (≥ 60 years old) and young GBM (yGBM, 20-60 years old) groups were explored. Immunohistochemistry (IHC) was utilized to depict the abundance of CD8+ cells (the main resource of IFN-γ) and IFITM2 protein expression in GBM samples. Furthermore, reverse transcription-polymerase chain reaction (RT-PCR) and Western blotting (WB) were performed to verify the link between IFN-γ and IFITM2. Moreover, the small-interfering RNA (siRNA) of IFITM2 was used to explore the function of IFITM2 in GBM. Characterized by inflammatory TME and higher IFITM2 expression, eGBM harbored a shorter survival time. Chemotaxis and inflammatory cytokine-related genes were enriched in the eGBM group, with more infiltrative CD8+ T cells. The IHC of CD8 and IFITM2-staining could demonstrate these results. In addition, the IFN-γ response pathway was activated in eGBM and resulted in a dismal outcome. Next, it was found that IFITM2 triggered by IFN-γ played a key role in IFN-γ-induced malignant phenotype in eGBM.
Collapse
Affiliation(s)
- Tingyu Liang
- Department of Neurosurgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xiaoxuan Wang
- Department of Pathology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Yu Wang
- Department of Neurosurgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
| | - Wenbin Ma
- Department of Neurosurgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
| |
Collapse
|
10
|
Bigi A, Cascella R, Cecchi C. α-Synuclein oligomers and fibrils: partners in crime in synucleinopathies. Neural Regen Res 2023; 18:2332-2342. [PMID: 37282450 PMCID: PMC10360081 DOI: 10.4103/1673-5374.371345] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/08/2023] Open
Abstract
The misfolding and aggregation of α-synuclein is the general hallmark of a group of devastating neurodegenerative pathologies referred to as synucleinopathies, such as Parkinson's disease, dementia with Lewy bodies, and multiple system atrophy. In such conditions, a range of different misfolded aggregates, including oligomers, protofibrils, and fibrils, are present both in neurons and glial cells. Growing experimental evidence supports the proposition that soluble oligomeric assemblies, formed during the early phases of the aggregation process, are the major culprits of neuronal toxicity; at the same time, fibrillar conformers appear to be the most efficient at propagating among interconnected neurons, thus contributing to the spreading of α-synuclein pathology. Moreover, α-synuclein fibrils have been recently reported to release soluble and highly toxic oligomeric species, responsible for an immediate dysfunction in the recipient neurons. In this review, we discuss the current knowledge about the plethora of mechanisms of cellular dysfunction caused by α-synuclein oligomers and fibrils, both contributing to neurodegeneration in synucleinopathies.
Collapse
Affiliation(s)
- Alessandra Bigi
- Department of Experimental and Clinical Biomedical Sciences, Section of Biochemistry, University of Florence, Florence, Italy
| | - Roberta Cascella
- Department of Experimental and Clinical Biomedical Sciences, Section of Biochemistry, University of Florence, Florence, Italy
| | - Cristina Cecchi
- Department of Experimental and Clinical Biomedical Sciences, Section of Biochemistry, University of Florence, Florence, Italy
| |
Collapse
|