1
|
Oh S, Kim S, Kim JP, Seo SW, Park BY, Park H. Association of APOC1 with cortical atrophy during conversion to Alzheimer's disease. GeroScience 2025:10.1007/s11357-025-01695-6. [PMID: 40369256 DOI: 10.1007/s11357-025-01695-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2025] [Accepted: 05/02/2025] [Indexed: 05/16/2025] Open
Abstract
Alzheimer's disease (AD) is a prevalent neurodegenerative disorder, with its progression influenced by aberrant gene expression and alterations in the brain network topology. Although APOE has been extensively studied in relation to AD, the role of APOC1 remains relatively underexplored. This study investigated the impact of APOC1 on changes in cortical thickness (CTh) during conversion to AD in a longitudinal setting. Using a normative modeling approach, we examined changes in CTh in patients with mild cognitive impairment (MCI). The spatial patterns of CTh changes were then correlated with APOC1 mRNA expression levels. We estimated the time to conversion to AD and compared progression rates between the low and high APOC1 expression groups. Finally, mediation analysis was performed to assess the indirect effects of APOC1 expression on memory function via CTh changes. In patients with MCI and AD, reduced CTh was observed in the limbic and default mode regions, with a notable impact on the entorhinal cortex, parahippocampus, and fusiform gyrus when comparing baseline and follow-up measurements. The degree of change in CTh was significantly associated with APOC1 expression, with the paralimbic regions identified as particularly vulnerable. Furthermore, the high APOC1 expression group demonstrated more rapid conversion to AD than that observed in the low expression group. Mediation analysis indicated a trend suggesting that APOC1 expression indirectly affected memory and cognitive function through its influence on CTh. These results highlight the potential of APOC1 as an additional focus of AD research, offering insights into the genetic influences on AD pathology.
Collapse
Affiliation(s)
- Sewook Oh
- Department of Electrical and Computer Engineering, Sungkyunkwan University, Jangan-gu, Suwon, Gyeonggi-do, 16419, Republic of Korea
- Center for Neuroscience Imaging Research, Institute for Basic Science, Jangan-gu, Suwon, Gyeonggi-do, 16419, Republic of Korea
| | - Sunghun Kim
- Department of Brain and Cognitive Engineering, Korea University, Seongbuk-gu, Seoul, 02841, Republic of Korea
- BK21 Four Institute of Precision Public Health, Seoul, Republic of Korea
| | - Jun Pyo Kim
- Alzheimer's Disease Convergence Research Center, Samsung Medical Center, Gangnam-gu, Seoul, 06351, Republic of Korea
- Department of Neurology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Gangnam-gu, Seoul, 06351, Republic of Korea
- Neuroscience Center, Samsung Medical Center, Gangnam-gu, Seoul, 06351, Republic of Korea
| | - Sang Won Seo
- Alzheimer's Disease Convergence Research Center, Samsung Medical Center, Gangnam-gu, Seoul, 06351, Republic of Korea
- Department of Neurology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Gangnam-gu, Seoul, 06351, Republic of Korea
- Neuroscience Center, Samsung Medical Center, Gangnam-gu, Seoul, 06351, Republic of Korea
| | - Bo-Yong Park
- Center for Neuroscience Imaging Research, Institute for Basic Science, Jangan-gu, Suwon, Gyeonggi-do, 16419, Republic of Korea.
- Department of Brain and Cognitive Engineering, Korea University, Seongbuk-gu, Seoul, 02841, Republic of Korea.
| | - Hyunjin Park
- Department of Electrical and Computer Engineering, Sungkyunkwan University, Jangan-gu, Suwon, Gyeonggi-do, 16419, Republic of Korea.
- Center for Neuroscience Imaging Research, Institute for Basic Science, Jangan-gu, Suwon, Gyeonggi-do, 16419, Republic of Korea.
| |
Collapse
|
2
|
OuYang C, Shi H, Lin Z. Identification of Alzheimer's disease susceptibility genes by the integration of genomics and transcriptomics. Neurol Res 2025:1-13. [PMID: 40331660 DOI: 10.1080/01616412.2025.2499890] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2025] [Accepted: 04/23/2025] [Indexed: 05/08/2025]
Abstract
BACKGROUND Alzheimer's disease (AD) is a progressive neurodegenerative disease. With the deepening of clinical and genomic research, a series of biomarkers and risk factors related to AD have been identified. However, the exact molecular mechanism of AD is not completely understood. METHODS By combining expression quantitative trait loci (eQTLs) analysis with the results of genome-wide association studies (GWAS), the candidate genes (CG) related to AD were screened out accurately. We identified that intersection genes of differentially expressed genes (DEGs) and CG are the key genes. Then, GO, KEGG, and GSEA were utilized for functional enrichment analysis. Finally, we predicted AD responses to immunotherapy by the single sample gene set enrichment analysis (ssGSEA). RESULTS A total of 253 DEGs were identified. The three key genes (VASP, SURF2, and TARBP1) were identified by taking the intersection of DEGs and CG. Through Mendelian randomization (MR) analysis, it was found that the risk of AD was significantly increased when VASP expression increased (OR = 0.1.046), while the risk of AD was significantly decreased when SURF2 (OR = 0.897) and TARBP1(OR = 0.920) expression increased. Subsequently, the functional analysis indicated that the core genes were mainly enriched in Leukocyte Transendothelial Migration, cGMP-PKG signaling pathway, and Rap1 signaling pathway. Through ssGSEA analysis showed that all three core genes were significantly related to M2 macrophages. CONCLUSIONS Three core genes were screened by integrating eQTLs data, GWAS data and transfer group data, and the potential mechanism of diagnosis and treatment of AD was revealed.
Collapse
Affiliation(s)
- Chenghong OuYang
- Jiangxi Provincial People's Hospital, The First Affiliated Hospital of Nanchang Medical College, Nanchang, China
| | - Hanping Shi
- School of Public Health, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
- Jiangxi Provincial Key Laboratory of Disease Prevention and Public Health, Nanchang University, Nanchang, Jiangxi, China
| | - Zhiying Lin
- Jiangxi Provincial People's Hospital, The First Affiliated Hospital of Nanchang Medical College, Nanchang, China
| |
Collapse
|
3
|
Liu S, Zhang Z, Gu Y, Hao J, Liu Y, Fu H, Guo X, Song H, Zhang S, Zhao Y. Beyond the eye: A relational model for early dementia detection using retinal OCTA images. Med Image Anal 2025; 102:103513. [PMID: 40022853 DOI: 10.1016/j.media.2025.103513] [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: 02/13/2025] [Accepted: 02/15/2025] [Indexed: 03/04/2025]
Abstract
Early detection of dementia, such as Alzheimer's disease (AD) or mild cognitive impairment (MCI), is essential to enable timely intervention and potential treatment. Accurate detection of AD/MCI is challenging due to the high complexity, cost, and often invasive nature of current diagnostic techniques, which limit their suitability for large-scale population screening. Given the shared embryological origins and physiological characteristics of the retina and brain, retinal imaging is emerging as a potentially rapid and cost-effective alternative for the identification of individuals with or at high risk of AD. In this paper, we present a novel PolarNet+ that uses retinal optical coherence tomography angiography (OCTA) to discriminate early-onset AD (EOAD) and MCI subjects from controls. Our method first maps OCTA images from Cartesian coordinates to polar coordinates, allowing approximate sub-region calculation to implement the clinician-friendly early treatment of diabetic retinopathy study (ETDRS) grid analysis. We then introduce a multi-view module to serialize and analyze the images along three dimensions for comprehensive, clinically useful information extraction. Finally, we abstract the sequence embedding into a graph, transforming the detection task into a general graph classification problem. A regional relationship module is applied after the multi-view module to explore the relationship between the sub-regions. Such regional relationship analyses validate known eye-brain links and reveal new discriminative patterns. The proposed model is trained, tested, and validated on four retinal OCTA datasets, including 1,671 participants with AD, MCI, and healthy controls. Experimental results demonstrate the performance of our model in detecting AD and MCI with an AUC of 88.69% and 88.02%, respectively. Our results provide evidence that retinal OCTA imaging, coupled with artificial intelligence, may serve as a rapid and non-invasive approach for large-scale screening of AD and MCI. The code is available at https://github.com/iMED-Lab/PolarNet-Plus-PyTorch, and the dataset is also available upon request.
Collapse
Affiliation(s)
- Shouyue Liu
- Ningbo Institute of Materials Technology and Engineering, Chinese Academy of Sciences, Ningbo, 315000, China
| | - Ziyi Zhang
- Department of Neurology, West China Hospital, Sichuan University, Chengdu, 610000, China
| | - Yuanyuan Gu
- Ningbo Institute of Materials Technology and Engineering, Chinese Academy of Sciences, Ningbo, 315000, China
| | - Jinkui Hao
- Ningbo Institute of Materials Technology and Engineering, Chinese Academy of Sciences, Ningbo, 315000, China
| | - Yonghuai Liu
- Department of Computer Science, Edge Hill University, Ormskirk, L39 4QP, United Kingdom
| | - Huazhu Fu
- Institute of High-Performance Computing, Agency for Science, Technology and Research, Singapore, 138632, Singapore
| | - Xinyu Guo
- Ningbo Institute of Materials Technology and Engineering, Chinese Academy of Sciences, Ningbo, 315000, China
| | - Hong Song
- School of Computer Science and Technology, Beijing Institute of Technology, Beijing, 100081, China.
| | - Shuting Zhang
- Department of Neurology, West China Hospital, Sichuan University, Chengdu, 610000, China.
| | - Yitian Zhao
- Ningbo Institute of Materials Technology and Engineering, Chinese Academy of Sciences, Ningbo, 315000, China.
| |
Collapse
|
4
|
Mieling M, Yousuf M, Bunzeck N. Predicting the progression of MCI and Alzheimer's disease on structural brain integrity and other features with machine learning. GeroScience 2025:10.1007/s11357-025-01626-5. [PMID: 40285975 DOI: 10.1007/s11357-025-01626-5] [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: 11/26/2024] [Accepted: 03/13/2025] [Indexed: 04/29/2025] Open
Abstract
Machine learning (ML) on structural MRI data shows high potential for classifying Alzheimer's disease (AD) progression, but the specific contribution of brain regions, demographics, and proteinopathy remains unclear. Using Alzheimer's Disease Neuroimaging Initiative (ADNI) data, we applied an extreme gradient-boosting algorithm and SHAP (SHapley Additive exPlanations) values to classify cognitively normal (CN) older adults, those with mild cognitive impairment (MCI) and AD dementia patients. Features included structural MRI, CSF status, demographics, and genetic data. Analyses comprised one cross-sectional multi-class classification (CN vs. MCI vs. AD dementia, n = 568) and two longitudinal binary-class classifications (CN-to-MCI converters vs. CN stable, n = 92; MCI-to-AD converters vs. MCI stable, n = 378). All classifications achieved 70-77% accuracy and 61-83% precision. Key features were CSF status, hippocampal volume, entorhinal thickness, and amygdala volume, with a clear dissociation: hippocampal properties contributed to the conversion to MCI, while the entorhinal cortex characterized the conversion to AD dementia. The findings highlight explainable, trajectory-specific insights into AD progression.
Collapse
Affiliation(s)
- Marthe Mieling
- Department of Psychology, University of Lübeck, Ratzeburger Allee 160, 23562, Lübeck, Germany.
| | - Mushfa Yousuf
- Department of Psychology, University of Lübeck, Ratzeburger Allee 160, 23562, Lübeck, Germany
| | - Nico Bunzeck
- Department of Psychology, University of Lübeck, Ratzeburger Allee 160, 23562, Lübeck, Germany.
- Center of Brain, Behavior and Metabolism (CBBM), University of Lübeck, Ratzeburger Allee 160, 23562, Lübeck, Germany.
| |
Collapse
|
5
|
Malik R, Martinez MR, So I, Finger E. Conversion to Mild Cognitive Impairment and Alzheimer's Disease Dementia Related to Apathy, APOE Genotype and Antidepressant Use. J Geriatr Psychiatry Neurol 2025:8919887251335002. [PMID: 40227643 DOI: 10.1177/08919887251335002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 04/15/2025]
Abstract
ObjectiveApathy and APOE ε4 genotype are risk factors for developing Alzheimer's disease dementia (ADD). Antidepressant use is known to induce apathy. This study aimed to examine associations between APOE ε4, apathy, and antidepressant use with progression from cognitively normal (CN) to mild cognitive impairments (MCI), and MCI to ADD.MethodsParticipants aged 55-90 were recruited from the Alzheimer's Disease Neuroimaging Initiative. Participants were CN or had MCI at baseline and had completed at least 3 consecutive study visits. The NPI and NPI-Q apathy subscales were used to index the presence of apathy. Antidepressants used by participants included SSRIs, SNRIs, and AYTADs. Cox proportional hazards analyses examined the combined effects of apathy, APOE ε4 genotype, and antidepressant use on conversion from CN to MCI and from MCI to ADD.ResultsApathy and APOE ε4 were associated with increased risk of conversion along the CN-MCI-ADD continuum. Antidepressant use was associated with progression from MCI to ADD, and progression from CN to MCI in non-apathetic APOE ε4 carriers.ConclusionOur findings support apathy and APOE ε4 as robust predictors of conversion to MCI and ADD, and demonstrate novel associations between antidepressant use and conversion. Future research should explore whether antidepressant use in MCI and ADD causes apathetic symptoms or serves to index apathy/depression severity.
Collapse
Affiliation(s)
- Rubina Malik
- Department of Clinical Neurological Sciences, Schulich School of Medicine & Dentistry, Western University, London, ON, Canada
| | - Miguel Restrepo Martinez
- Department of Clinical Neurological Sciences, Schulich School of Medicine & Dentistry, Western University, London, ON, Canada
- Department of Psychiatry, Clinica Las Americas AUNA, Medellin, Colombia
| | - Isis So
- Department of Clinical Neurological Sciences, Schulich School of Medicine & Dentistry, Western University, London, ON, Canada
| | - Elizabeth Finger
- Department of Clinical Neurological Sciences, Schulich School of Medicine & Dentistry, Western University, London, ON, Canada
| |
Collapse
|
6
|
Rabl M, Clark C, Dayon L, Popp J. Neuropsychiatric symptoms in cognitive decline and Alzheimer's disease: biomarker discovery using plasma proteomics. J Neurol Neurosurg Psychiatry 2025; 96:370-382. [PMID: 39288961 PMCID: PMC12015082 DOI: 10.1136/jnnp-2024-333819] [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: 03/13/2024] [Accepted: 08/07/2024] [Indexed: 09/19/2024]
Abstract
BACKGROUND AND OBJECTIVES Neuropsychiatric symptoms (NPS) are common in older people with cognitive impairment and Alzheimer's disease (AD). No biomarkers to detect the related pathology or predict the clinical evolution of NPS are available yet. This study aimed to identify plasma proteins that may serve as biomarkers for NPS and NPS-related clinical disease progression. METHODS A panel of 190 plasma proteins was quantified using Luminex xMAP in the Alzheimer's Disease Neuroimaging Initiative cohort. NPS and cognitive performance were assessed at baseline and after 1 and 2 years. Logistic regression, receiver operating characteristic analysis and cross-validation were used to address the relations of interest. RESULTS A total of 507 participants with mild cognitive impairment (n=396) or mild AD dementia (n=111) were considered. Selected plasma proteins improved the prediction of NPS (area under the curve (AUC) from 0.61 to 0.76, p<0.001) and future NPS (AUC from 0.63 to 0.80, p<0.001) when added to a reference model. Distinct protein panels were identified for single symptoms. Among the selected proteins, ANGT, CCL1 and IL3 were associated with NPS at all three time points while CCL1, serum glutamic oxaloacetic transaminase and complement factor H were also associated with cognitive decline. The associations were independent of the presence of cerebral AD pathology as assessed using cerebrospinal fluid biomarkers. CONCLUSIONS Plasma proteins are associated with NPS and improve prediction of future NPS.
Collapse
Affiliation(s)
- Miriam Rabl
- Department of Psychiatry, Psychotherapy and Psychosomatics, University of Zurich, Psychiatric University Hospital, Zurich, Switzerland
| | - Christopher Clark
- Department of Psychiatry, Psychotherapy and Psychosomatics, University of Zurich, Psychiatric University Hospital, Zurich, Switzerland
| | - Loïc Dayon
- Nestlé Institute of Food Safety & Analytical Sciences, Nestlé Research, Lausanne, Switzerland
- Institut des Sciences et Ingénierie Chimiques, Ecole Polytechnique Federale de Lausanne, Lausanne, Switzerland
| | - Julius Popp
- Department of Psychiatry, Psychotherapy and Psychosomatics, University of Zurich, Psychiatric University Hospital, Zurich, Switzerland
- Old-Age Psychiatry Service, Department of Psychiatry, Lausanne University Hospital, Lausanne, Switzerland
| |
Collapse
|
7
|
Li Y, Niu D, Qi K, Liang D, Long X. An imaging and genetic-based deep learning network for Alzheimer's disease diagnosis. Front Aging Neurosci 2025; 17:1532470. [PMID: 40191788 PMCID: PMC11968703 DOI: 10.3389/fnagi.2025.1532470] [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: 11/26/2024] [Accepted: 03/05/2025] [Indexed: 04/09/2025] Open
Abstract
Conventional computer-aided diagnostic techniques for Alzheimer's disease (AD) predominantly rely on magnetic resonance imaging (MRI) in isolation. Genetic imaging methods, by establishing the link between genes and brain structures in disease progression, facilitate early prediction of AD development. While deep learning methods based on MRI have demonstrated promising results for early AD diagnosis, the limited dataset size has led most AD studies to lean on statistical approaches within the realm of imaging genetics. Existing deep-learning approaches typically utilize pre-defined regions of interest and risk variants from known susceptibility genes, employing relatively straightforward feature fusion methods that fail to fully capture the relationship between images and genes. To address these limitations, we proposed a multi-modal deep learning classification network based on MRI and single nucleotide polymorphism (SNP) data for AD diagnosis and mild cognitive impairment (MCI) progression prediction. Our model leveraged a convolutional neural network (CNN) to extract whole-brain structural features, a Transformer network to capture genetic features, and employed a cross-transformer-based network for comprehensive feature fusion. Furthermore, we incorporated an attention-map-based interpretability method to analyze and elucidate the structural and risk variants associated with AD and their interrelationships. The proposed model was trained and evaluated using 1,541 subjects from the ADNI database. Experimental results underscored the superior performance of our model in effectively integrating and leveraging information from both modalities, thus enhancing the accuracy of AD diagnosis and prediction.
Collapse
Affiliation(s)
- Yuhan Li
- Research Centers for Medical AI, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Donghao Niu
- Research Centers for Medical AI, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Keying Qi
- Research Centers for Medical AI, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Dong Liang
- Research Centers for Medical AI, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
- The Key Laboratory of Biomedical Imaging Science and System, Chinese Academy of Sciences, Shenzhen, China
| | - Xiaojing Long
- Research Centers for Medical AI, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
- The Key Laboratory of Biomedical Imaging Science and System, Chinese Academy of Sciences, Shenzhen, China
| |
Collapse
|
8
|
Liu J, Wu S, Fu Q, Luo X, Luo Y, Qin S, Huang Y, Chen Z. Multimodal diagnosis of Alzheimer's disease based on resting-state electroencephalography and structural magnetic resonance imaging. Front Physiol 2025; 16:1515881. [PMID: 40144547 PMCID: PMC11937600 DOI: 10.3389/fphys.2025.1515881] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2024] [Accepted: 02/14/2025] [Indexed: 03/28/2025] Open
Abstract
Multimodal diagnostic methods for Alzheimer's disease (AD) have demonstrated remarkable performance. However, the inclusion of electroencephalography (EEG) in such multimodal studies has been relatively limited. Moreover, most multimodal studies on AD use convolutional neural networks (CNNs) to extract features from different modalities and perform fusion classification. Regrettably, this approach often lacks collaboration and fails to effectively enhance the representation ability of features. To address this issue and explore the collaborative relationship among multimodal EEG, this paper proposes a multimodal AD diagnosis model based on resting-state EEG and structural magnetic resonance imaging (sMRI). Specifically, this work designs corresponding feature extraction models for EEG and sMRI modalities to enhance the capability of extracting modality-specific features. Additionally, a multimodal joint attention mechanism (MJA) is developed to address the issue of independent modalities. The MJA promotes cooperation and collaboration between the two modalities, thereby enhancing the representation ability of multimodal fusion. Furthermore, a random forest classifier is introduced to enhance the classification ability. The diagnostic accuracy of the proposed model can achieve 94.7%, marking a noteworthy accomplishment. This research stands as the inaugural exploration into the amalgamation of deep learning and EEG multimodality for AD diagnosis. Concurrently, this work strives to bolster the use of EEG in multimodal AD research, thereby positioning itself as a hopeful prospect for future advancements in AD diagnosis.
Collapse
Affiliation(s)
- Junxiu Liu
- Guangxi Key Laboratory of Brain-inspired Computing and Intelligent Chips, School of Electronic and Information Engineering, Guangxi Normal University, Guilin, China
- Key Laboratory of Nonlinear Circuits and Optical Communications, Education Department of Guangxi Zhuang Autonomous Region, Guangxi Normal University, Guilin, Guangxi, China
| | - Shangxiao Wu
- Guangxi Key Laboratory of Brain-inspired Computing and Intelligent Chips, School of Electronic and Information Engineering, Guangxi Normal University, Guilin, China
- Key Laboratory of Nonlinear Circuits and Optical Communications, Education Department of Guangxi Zhuang Autonomous Region, Guangxi Normal University, Guilin, Guangxi, China
- Xiangsihu College, Guangxi University for Nationalities, Nanning, China
| | - Qiang Fu
- Guangxi Key Laboratory of Brain-inspired Computing and Intelligent Chips, School of Electronic and Information Engineering, Guangxi Normal University, Guilin, China
- Key Laboratory of Nonlinear Circuits and Optical Communications, Education Department of Guangxi Zhuang Autonomous Region, Guangxi Normal University, Guilin, Guangxi, China
| | - Xiwen Luo
- Guangxi Key Laboratory of Brain-inspired Computing and Intelligent Chips, School of Electronic and Information Engineering, Guangxi Normal University, Guilin, China
- Key Laboratory of Nonlinear Circuits and Optical Communications, Education Department of Guangxi Zhuang Autonomous Region, Guangxi Normal University, Guilin, Guangxi, China
| | - Yuling Luo
- Guangxi Key Laboratory of Brain-inspired Computing and Intelligent Chips, School of Electronic and Information Engineering, Guangxi Normal University, Guilin, China
- Key Laboratory of Nonlinear Circuits and Optical Communications, Education Department of Guangxi Zhuang Autonomous Region, Guangxi Normal University, Guilin, Guangxi, China
| | - Sheng Qin
- Guangxi Key Laboratory of Brain-inspired Computing and Intelligent Chips, School of Electronic and Information Engineering, Guangxi Normal University, Guilin, China
- Key Laboratory of Nonlinear Circuits and Optical Communications, Education Department of Guangxi Zhuang Autonomous Region, Guangxi Normal University, Guilin, Guangxi, China
| | - Yiting Huang
- Guangxi Key Laboratory of Brain-inspired Computing and Intelligent Chips, School of Electronic and Information Engineering, Guangxi Normal University, Guilin, China
- Key Laboratory of Nonlinear Circuits and Optical Communications, Education Department of Guangxi Zhuang Autonomous Region, Guangxi Normal University, Guilin, Guangxi, China
| | - Zhaohui Chen
- Guangxi Key Laboratory of Brain-inspired Computing and Intelligent Chips, School of Electronic and Information Engineering, Guangxi Normal University, Guilin, China
- Key Laboratory of Nonlinear Circuits and Optical Communications, Education Department of Guangxi Zhuang Autonomous Region, Guangxi Normal University, Guilin, Guangxi, China
| |
Collapse
|
9
|
Wang B, Chibnik LB, Choi SH, Blacker D, DeStefano AL, Lin H. Association of genetic risk of Alzheimer's disease and cognitive function in two European populations. Sci Rep 2025; 15:6410. [PMID: 39984543 PMCID: PMC11845681 DOI: 10.1038/s41598-025-90277-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2024] [Accepted: 02/11/2025] [Indexed: 02/23/2025] Open
Abstract
Although there is some evidence of an association between Alzheimer's disease polygenic risk score (AD PRS) and cognitive function, limited validations have been performed in large populations. We investigated the relationship between AD PRS and cognitive function in the UK Biobank in over 276,000 participants and further validated the association in the Alzheimer's Disease Neuroimaging Initiative (ADNI) sample. We developed the AD PRS (excluded the APOE variants) in the middle age UK Biobank participants (age ranged 39-72, mean age 57 years) of European ancestries by LDpred2. To validate the association of AD PRS and cognitive function internally in the UK Biobank, we linearly regressed standardized cognitive function on continuous standardized AD PRS with age at cognitive test, sex, genotyping array, first 10 principal components of genotyping, smoking, education in years, body mass index, and apolipoprotein E gene ε4 (APOE4) risk allele dosages. To validate the associations externally, we ran the linear mixed effects model in the ADNI sample free of dementia (age ranged 55-91, mean age 73), including similar covariates as fixed effects and participants' IDs as the random effect. Stratification by age, APOE4 carrier status, and cognitive status (cognitively normal or mild cognitive impairment) was also investigated. Our study validated the associations of AD PRS and cognitive function in both midlife and late-life observational cohorts. Although not all of the cognitive measures were significantly associated with AD PRS, non-verbal fluid reasoning (matrix pattern completion, β = - 0.022, P = 0.003), processing speed (such as symbol digit substitution, β = - 0.017, P = 1.08E-05), short-term memory and attention (such as pairs matching, β = - 0.014, P = 1.66E-10), and reaction time (β = - 0.010, P = 1.19E-06) were inversely associated with increasing AD PRS in the UK Biobank. Higher likelihood of cognitive impairment was also associated with higher AD PRS in the ADNI cognitive normal individuals (AD assessment scale β = 0.079, P = 0.02). In summary, we confirmed that poorer cognitive function was associated with a higher polygenic AD risk, and suggested the potential utility of the AD PRS in identifying those who may be at risk for further cognitive decline.
Collapse
Affiliation(s)
- Biqi Wang
- Department of Medicine, University of Massachusetts Chan Medical School, Worcester, MA, USA.
| | - Lori B Chibnik
- Department of Neurology, Massachusetts General Hospital, Boston, MA, USA
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Seung Hoan Choi
- Department of Biostatistics, Boston University School of Public Health, Boston, MA, USA
| | - Deborah Blacker
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA
- Department of Psychiatry, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Anita L DeStefano
- Department of Biostatistics, Boston University School of Public Health, Boston, MA, USA
| | - Honghuang Lin
- Department of Medicine, University of Massachusetts Chan Medical School, Worcester, MA, USA
| |
Collapse
|
10
|
Sarma M, Chatterjee S. Machine Learning-Based Alzheimer's Disease Stage Diagnosis Utilizing Blood Gene Expression and Clinical Data: A Comparative Investigation. Diagnostics (Basel) 2025; 15:211. [PMID: 39857095 PMCID: PMC11765009 DOI: 10.3390/diagnostics15020211] [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: 12/01/2024] [Revised: 01/03/2025] [Accepted: 01/06/2025] [Indexed: 01/27/2025] Open
Abstract
Background/Objectives: This study presents a comparative analysis of the multistage diagnosis of Alzheimer's disease (AD), including mild cognitive impairment (MCI), utilizing two distinct types of biomarkers: blood gene expression and clinical biomarker samples. Both of these samples, obtained from participants in the Alzheimer's Disease Neuroimaging Initiative (ADNI), were independently analyzed utilizing machine learning (ML)-based multiclassifiers. This study applied novel machine learning-based data augmentation techniques to gene expression profile data that are high-dimensional, low-sample-size (HDLSS) and inherently highly imbalanced. The investigation obtained the highest multiclassification performance to date in the multistage diagnosis of Alzheimer's disease utilizing the blood gene expression profiles of Alzheimer's Disease Neuroimaging Initiative (ADNI) participants. Based on the performance results obtained, and other factors such as early prediction capabilities, this study compares the efficacies of the two types of biomarkers for multistage diagnosis. This study presents the sole investigation in which multiclassification-based AD stage diagnosis was conducted utilizing blood gene expression data. We obtained the best multiclassification result in both modalities of the ADNI data in terms of F1-score and were able to identify new genetic biomarkers. Methods: The combination of the XGBoost and SFBS (Sequential Floating Backward Selection) methods was used to select the features. We were able to select the 95 most effective gene probe sets out of 49,386. For the clinical study data, eight of the most effective biomarkers were selected using SFBS. A deep learning (DL) classifier was used to identify the stages-cognitive normal (CN), mild cognitive impairment (MCI), and Alzheimer's disease (AD)/dementia. DL, support vector machine (SVM), gradient boosting (GB), and random forest (RF) classifiers were used for the AD stage detection from gene expression profile data. Because of the high data imbalance in genomic data, borderline oversampling/data augmentation was applied in the model training and original samples for validation. Results: Utilizing clinical data, the highest ROC AUC scores attained were 0.989, 0.927, and 0.907 for the identification of the CN, MCI, and dementia stages, respectively. The highest F1 scores achieved were 0.971, 0.939, and 0.886. Employing gene expression data, we obtained ROC AUC scores of 0.763, 0.761, and 0.706 for the CN, MCI, and dementia stages, respectively, and F1 scores of 0.71, 0.77, and 0.53 for CN, MCI, and dementia, respectively. Conclusions: This represents the best outcome to date for AD stage diagnosis from ADNI blood gene expression profile data utilizing multiclassification techniques. The results indicated that our multiclassification model effectively manages the imbalanced data of a high-dimension, low-sample-size (HDLSS) nature to identify samples of the minority class. MAPK14, PLG, FZD2, FXYD6, and TEP1 are among the novel genes identified as being associated with AD risk.
Collapse
Affiliation(s)
- Manash Sarma
- Department of Computer Science and Engineering, Faculty of Engineering and Technology, Technology Campus (Peenya Campus), Ramaiah University of Applied Sciences, Bengaluru 560058, India
| | | |
Collapse
|
11
|
Gao M, Kong W, Liu K, Wen G, Yu Y, Zhu Y, Jiang Z, Wei K. Exploring Brain Imaging and Genetic Risk Factors in Different Progression States of Alzheimer's Disease Through OSnetNMF-Based Methods. J Mol Neurosci 2025; 75:7. [PMID: 39815147 DOI: 10.1007/s12031-024-02274-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2024] [Accepted: 09/29/2024] [Indexed: 01/18/2025]
Abstract
Alzheimer's disease (AD) is a neurodegenerative disease with no effective treatment, often preceded by mild cognitive impairment (MCI). Multimodal imaging genetics integrates imaging and genetic data to gain a deeper understanding of disease progression and individual variations. This study focuses on exploring the mechanisms that drive the transition from normal cognition to MCI and ultimately to AD. As an effective joint feature extraction and dimensionality reduction method, non-negative matrix factorization (NMF) and its improved variants, particularly the network-based non-negative matrix factorization (netNMF), have been widely used in multimodal analysis to mine brain imaging and genetic data by considering the interactions between different features. However, many of these methods overlook the importance of the coefficient matrix and do not address issues related to data accuracy and feature redundancy. To address these limitations, we propose an orthogonal sparse network non-negative matrix factorization (OSnetNMF) algorithm, which introduces orthogonal and sparse constraints based on netNMF. By establishing linear relationships between structural magnetic resonance imaging (sMRI) and corresponding gene expression data, OSnetNMF reduces feature redundancy and decreases correlation between data, resulting in more accurate and reliable biomarker extraction. Experiments demonstrate that the OSnetNMF algorithm can accurately identify risk regions of interest (ROIs) and key genes that characterize AD progression, revealing significant trends in ROI pairs such as l4thVen-HIF1A, rBst-MPO, and rBst-PTK2B. Comparative experiments show that the improved algorithm outperforms traditional methods, identifying more disease-related biomarkers and achieving better reconstruction performance.
Collapse
Affiliation(s)
- Min Gao
- College of Information Engineering, Shanghai Maritime University, 1550 Haigang Ave., Shanghai, 201306, P. R. China
| | - Wei Kong
- College of Information Engineering, Shanghai Maritime University, 1550 Haigang Ave., Shanghai, 201306, P. R. China.
| | - Kun Liu
- College of Information Engineering, Shanghai Maritime University, 1550 Haigang Ave., Shanghai, 201306, P. R. China
| | - Gen Wen
- Department of Orthopedic Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
| | - Yaling Yu
- Department of Orthopedic Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
- Institute of Microsurgery on Extremities, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
| | - Yuemin Zhu
- CREATIS, University of Lyon, INSA Lyon, CNRS UMR 5220, Inserm U1294, Lyon, 69621, France
| | - Zhihan Jiang
- College of Information Engineering, Shanghai Maritime University, 1550 Haigang Ave., Shanghai, 201306, P. R. China
| | - Kai Wei
- Bio-Med Big Data Center, CAS Key Laboratory of Computational Biology, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| |
Collapse
|
12
|
Patil S, Ayubcha C, Teichner E, Subtirelu R, Cho JH, Ghonim M, Ghonim M, Werner TJ, Høilund-Carlsen PF, Alavi A, Newberg AB. Clinical Applications of PET Imaging in Alzheimer's Disease. PET Clin 2025; 20:89-100. [PMID: 39547733 DOI: 10.1016/j.cpet.2024.09.015] [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] [Indexed: 11/17/2024]
Abstract
Alzheimer's disease (AD) involves a complex pathophysiology of neurodegeneration that leads to severe cognitive deficiencies. Understanding the molecular alterations that underlie this disease is fundamental to clinical management and therapeutic innovation. Functional imaging with positron emission tomography (PET) enables a visualization of these impaired pathways, such as cerebral hypometabolism, amyloid and tau accumulation, and neurotransmitter dysfunction. This review discusses the clinical applications of PET in AD and mild cognitive impairment, focusing on relevant original research studies for disease diagnosis, progression, and treatment response.
Collapse
Affiliation(s)
- Shiv Patil
- Department of Radiology, Hospital of the University of Pennsylvania, PA, USA; Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA, USA
| | - Cyrus Ayubcha
- Harvard Medical School, Boston, MA, USA; Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Eric Teichner
- Department of Radiology, Hospital of the University of Pennsylvania, PA, USA; Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA, USA
| | - Robert Subtirelu
- Department of Radiology, Hospital of the University of Pennsylvania, PA, USA
| | | | - Mohanad Ghonim
- Department of Radiology, Hospital of the University of Pennsylvania, PA, USA; Department of Radiology, Ain Shams University, Cairo, Egypt
| | - Mohamed Ghonim
- Department of Radiology, Hospital of the University of Pennsylvania, PA, USA; Department of Radiology, Ain Shams University, Cairo, Egypt
| | - Thomas J Werner
- Department of Radiology, Hospital of the University of Pennsylvania, PA, USA
| | | | - Abass Alavi
- Department of Radiology, Hospital of the University of Pennsylvania, PA, USA
| | - Andrew B Newberg
- Marcus Institute of Integrative Health, Thomas Jefferson University, Philadelphia, PA 19107, USA; Department of Radiology, Thomas Jefferson University, Philadelphia, PA, USA.
| |
Collapse
|
13
|
He B, Zhang S, Risacher SL, Saykin AJ, Yan J. Multi-modal Imaging-based Pseudotime Analysis of Alzheimer progression. PACIFIC SYMPOSIUM ON BIOCOMPUTING. PACIFIC SYMPOSIUM ON BIOCOMPUTING 2025; 30:664-674. [PMID: 39670403 PMCID: PMC12044618 DOI: 10.1142/9789819807024_0047] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 05/16/2025]
Abstract
Alzheimer's disease (AD) is a neurodegenerative disorder that results in progressive cognitive decline but without any clinically validated cures so far. Understanding the progression of AD is critical for early detection and risk assessment for AD in aging individuals, thereby enabling initiation of timely intervention and improved chance of success in AD trials. Recent pseudotime approach turns cross-sectional data into "faux" longitudinal data to understand how a complex process evolves over time. This is critical for Alzheimer, which unfolds over the course of decades, but the collected data offers only a snapshot. In this study, we tested several state-of-the-art pseudotime approaches to model the full spectrum of AD progression. Subsequently, we evaluated and compared the pseudotime progression score derived from individual imaging modalities and multi-modalities in the ADNI cohort. Our results showed that most existing pseudotime analysis tools do not generalize well to the imaging data, with either flipped progression score or poor separation of diagnosis groups. This is likely due to the underlying assumptions that only stand for single cell data. From the only tool with promising results, it was observed that all pseudotime, derived from either single imaging modalities or multi-modalities, captures the progressiveness of diagnosis groups. Pseudotime from multi-modality, but not the single modalities, confirmed the hypothetical temporal order of imaging phenotypes. In addition, we found that multi-modal pseudotime is mostly driven by amyloid and tau imaging, suggesting their continuous changes along the full spectrum of AD progression.
Collapse
Affiliation(s)
- Bing He
- Biomedical Engineering and Informatics, Indiana University Indianapolis, 535 W Michigan St., Indianapolis, Indiana 46202, USA,
| | - Shu Zhang
- Department of Computer Science, University of California Los Angeles 404 Westwood Plaza Engineering IV, Los Angeles, CA 90095, USA,
| | - Shannon L Risacher
- Department of Radiology and Imaging Sciences, Indiana University School of Medicine, 355 W 15th St., Indianapolis, Indiana 46202, USA,
| | - Andrew J Saykin
- Department of Radiology and Imaging Sciences, Indiana University School of Medicine, 355 W 15th St., Indianapolis, Indiana 46202, USA,
| | - Jingwen Yan
- Biomedical Engineering and Informatics, Indiana University Indianapolis, 535 W Michigan St., Indianapolis, Indiana 46202, USA,
| |
Collapse
|
14
|
Dagasso G, Wilms M, MacEachern SJ, Forkert ND. Application of a localized morphometrics approach to imaging-derived brain phenotypes for genotype-phenotype associations in pediatric mental health and neurodevelopmental disorders. Front Big Data 2024; 7:1429910. [PMID: 39722687 PMCID: PMC11668761 DOI: 10.3389/fdata.2024.1429910] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2024] [Accepted: 11/12/2024] [Indexed: 12/28/2024] Open
Abstract
Introduction Quantitative global or regional brain imaging measurements, known as imaging-specific or -derived phenotypes (IDPs), are commonly used in genotype-phenotype association studies to explore the genomic architecture of the brain and how it may be affected by neurological diseases (e.g., Alzheimer's disease), mental health (e.g., depression), and neurodevelopmental disorders (e.g., attention-deficit hyperactivity disorder [ADHD]). For this purpose, medical images have been used as IDPs using a voxel-wise or global approach via principal component analysis. However, these methods have limitations related to multiple testing or the inability to isolate high variation regions, respectively. Methods To address these limitations, this study investigates a localized, principal component analysis-like approach for dimensionality reduction of cross-sectional T1-weighted MRI datasets utilizing diffeomorphic morphometry. This approach can reduce the dimensionality of images while preserving spatial information and enables the inclusion of spatial locality in the analysis. In doing so, this method can be used to explore morphometric brain changes across specific components and spatial scales of interest and to identify associations with genome regions in a multivariate genome-wide association study. For a first clinical feasibility study, this method was applied to data from the Adolescent Brain Cognitive Development (ABCD) study, including adolescents with ADHD (n = 1,359), obsessive-compulsive disorder (n = 1,752), and depression (n = 1,766). Results Meaningful associations of specific morphometric features with genome regions were identified with the data and corresponded to previous found brain regions in the respective mental health and neurodevelopmental disorder cohorts. Discussion In summary, the localized, principal component analysis-like approach can reduce the dimensionality of medical images while still being able to identify meaningful local brain region alterations that are associated with genomic markers across multiple scales. The proposed method can be applied to various image types and can be easily integrated in many genotype-phenotype association study setups.
Collapse
Affiliation(s)
- Gabrielle Dagasso
- Department of Radiology, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
- Hotchkiss Brain Institute, University of Calgary, Calgary, AB, Canada
- Biomedical Engineering Graduate Program, University of Calgary, Calgary, AB, Canada
- Alberta Children's Hospital Research Institute, University of Calgary, Calgary, AB, Canada
| | - Matthias Wilms
- Hotchkiss Brain Institute, University of Calgary, Calgary, AB, Canada
- Alberta Children's Hospital Research Institute, University of Calgary, Calgary, AB, Canada
- Department of Pediatrics, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
- Department of Community Health Sciences, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Sarah J. MacEachern
- Hotchkiss Brain Institute, University of Calgary, Calgary, AB, Canada
- Alberta Children's Hospital Research Institute, University of Calgary, Calgary, AB, Canada
- Department of Pediatrics, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Nils D. Forkert
- Department of Radiology, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
- Hotchkiss Brain Institute, University of Calgary, Calgary, AB, Canada
- Alberta Children's Hospital Research Institute, University of Calgary, Calgary, AB, Canada
- Department of Clinical Neurosciences, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| |
Collapse
|
15
|
Andrews SJ, Boeriu AI, Belloy ME, Renton AE, Fulton‐Howard B, Brenowitz WD, Yaffe K, for the Alzheimer's Disease Neuroimaging Initiative. Dementia risk scores, apolipoprotein E, and risk of Alzheimer's disease: One size does not fit all. Alzheimers Dement 2024; 20:8595-8604. [PMID: 39428957 PMCID: PMC11667490 DOI: 10.1002/alz.14300] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2024] [Revised: 08/23/2024] [Accepted: 09/10/2024] [Indexed: 10/22/2024]
Abstract
INTRODUCTION Evaluating the generalizability of dementia risk scores, primarily developed in non-Latinx White (NLW) participants, and interactions with genetic risk factors in diverse populations is crucial for addressing health disparities. METHODS We analyzed the association of the Cardiovascular Risk Factors, Aging, and Incidence of Dementia (CAIDE) and modified CAIDE (mCAIDE) scores with dementia risk using logistic regression models stratified by race/ethnicity in National Alzheimer's Coordinating Center (NACC) and Alzheimer's Disease Neuroimaging Initiative (ADNI), and assessed their interaction with apolipoprotein E (APOE). RESULTS Higher CAIDE scores were associated with an increased risk of dementia in Asian, Latinx, and NLW participants but not in Black participants. In contrast, higher mCAIDE scores were also associated with an increased risk of dementia in Black participants. Unfavorable mCAIDE risk profiles exacerbated the apolipoprotein E*ε4 (APOE*ε4) risk effect and attenuated the APOE*ε2 protective effect. DISCUSSION Our findings underscore the importance of evaluating the validity of dementia risk scores in diverse populations for their use in personalized medicine approaches to promote brain health. HIGHLIGHTS Dementia risk scores demonstrate race/ethnic-specific effects on dementia risk. Unfavorable modifiable risk profiles moderate the effect of APOE on dementia risk. Dementia risk scores need to be validated in diverse populations.
Collapse
Affiliation(s)
- Shea J. Andrews
- Department of Psychiatry and Behavioral SciencesUniversity of California San FranciscoSan FranciscoCaliforniaUSA
| | - Ana I. Boeriu
- Department of Psychiatry and Behavioral SciencesUniversity of California San FranciscoSan FranciscoCaliforniaUSA
| | - Michael E. Belloy
- Department of Neurology and Neurological SciencesStanford UniversityStanfordCaliforniaUSA
- NeuroGenomics and Informatics CenterWashington University School of MedicineSt.LouisMissouriUSA
- Department of NeurologyWashington University School of MedicineSt.LouisMissouriUSA
| | - Alan E. Renton
- Department of Genetics and Genomic SciencesIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
| | - Brian Fulton‐Howard
- Department of Genetics and Genomic SciencesIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
| | - Willa D. Brenowitz
- Kaiser Permanente Center for Health ResearchPortlandOregonUSA
- Department of Epidemiology and BiostatisticsUniversity of California San FranciscoSan FranciscoCaliforniaUSA
| | - Kristine Yaffe
- Department of Psychiatry and Behavioral SciencesUniversity of California San FranciscoSan FranciscoCaliforniaUSA
- Department of Epidemiology and BiostatisticsUniversity of California San FranciscoSan FranciscoCaliforniaUSA
- Department of NeurologyUniversity of California San FranciscoSan FranciscoCaliforniaUSA
| | | |
Collapse
|
16
|
Wang J, Wen S, Liu W, Meng X, Jiao Z. Deep joint learning diagnosis of Alzheimer's disease based on multimodal feature fusion. BioData Min 2024; 17:48. [PMID: 39501294 PMCID: PMC11536794 DOI: 10.1186/s13040-024-00395-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2024] [Accepted: 10/08/2024] [Indexed: 11/09/2024] Open
Abstract
Alzheimer's disease (AD) is an advanced and incurable neurodegenerative disease. Genetic variations are intrinsic etiological factors contributing to the abnormal expression of brain function and structure in AD patients. A new multimodal feature fusion called "magnetic resonance imaging (MRI)-p value" was proposed to construct 3D fusion images by introducing genes as a priori knowledge. Moreover, a new deep joint learning diagnostic model was constructed to fully learn images features. One branch trained a residual network (ResNet) to learn the features of local pathological regions. The other branch learned the position information of brain regions with different changes in the different categories of subjects' brains by introducing attention convolution, and then obtained the discriminative probability information from locations via convolution and global average pooling. The feature and position information of the two branches were linearly interacted to acquire the diagnostic basis for classifying the different categories of subjects. The diagnoses of AD and health control (HC), AD and mild cognitive impairment (MCI), HC and MCI were performed with data from the Alzheimer's Disease Neuroimaging Initiative (ADNI). The results showed that the proposed method achieved optimal results in AD-related diagnosis. The classification accuracy (ACC) and area under the curve (AUC) of the three experimental groups were 93.44% and 96.67%, 89.06% and 92%, and 84% and 81.84%, respectively. Moreover, a total of six novel genes were found to be significantly associated with AD, namely NTM, MAML2, NAALADL2, FHIT, TMEM132D and PCSK5, which provided new targets for the potential treatment of neurodegenerative diseases.
Collapse
Affiliation(s)
- Jingru Wang
- School of Computer Science and Artificial Intelligence, Changzhou University, Changzhou, 213164, China
| | - Shipeng Wen
- School of Computer Science and Artificial Intelligence, Changzhou University, Changzhou, 213164, China
| | - Wenjie Liu
- School of Computer Information and Engineering, Changzhou Institute of Technology, Changzhou, 213032, China
| | - Xianglian Meng
- School of Computer Information and Engineering, Changzhou Institute of Technology, Changzhou, 213032, China.
- Wangzheng School of Microelectronics, Changzhou University, Changzhou, 213164, China.
| | - Zhuqing Jiao
- School of Computer Science and Artificial Intelligence, Changzhou University, Changzhou, 213164, China.
- Wangzheng School of Microelectronics, Changzhou University, Changzhou, 213164, China.
| |
Collapse
|
17
|
He B, Wu R, Sangani N, Pugalenthi PV, Patania A, Risacher SL, Nho K, Apostolova LG, Shen L, Saykin AJ, Yan J, for the Alzheimer's Disease Neuroimaging Initiative. Integrating amyloid imaging and genetics for early risk stratification of Alzheimer's disease. Alzheimers Dement 2024; 20:7819-7830. [PMID: 39285750 PMCID: PMC11567859 DOI: 10.1002/alz.14244] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2024] [Revised: 07/24/2024] [Accepted: 08/15/2024] [Indexed: 09/21/2024]
Abstract
INTRODUCTION Alzheimer's disease (AD) initiates years prior to symptoms, underscoring the importance of early detection. While amyloid accumulation starts early, individuals with substantial amyloid burden may remain cognitively normal, implying that amyloid alone is not sufficient for early risk assessment. METHODS Given the genetic susceptibility of AD, a multi-factorial pseudotime approach was proposed to integrate amyloid imaging and genotype data for estimating a risk score. Validation involved association with cognitive decline and survival analysis across risk-stratified groups, focusing on patients with mild cognitive impairment (MCI). RESULTS Our risk score outperformed amyloid composite standardized uptake value ratio in correlation with cognitive scores. MCI subjects with lower pseudotime risk score showed substantial delayed onset of AD and slower cognitive decline. Moreover, pseudotime risk score demonstrated strong capability in risk stratification within traditionally defined subgroups such as early MCI, apolipoprotein E (APOE) ε4+ MCI, APOE ε4- MCI, and amyloid+ MCI. DISCUSSION Our risk score holds great potential to improve the precision of early risk assessment. HIGHLIGHTS Accurate early risk assessment is critical for the success of clinical trials. A new risk score was built from integrating amyloid imaging and genetic data. Our risk score demonstrated improved capability in early risk stratification.
Collapse
Affiliation(s)
- Bing He
- Department of Biomedical Engineering and InformaticsIndiana University Luddy School of Informatics, Computing and EngineeringIndianapolisIndianaUSA
| | - Ruiming Wu
- Department of Biomedical Engineering and Informatics, Perelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
| | - Neel Sangani
- Department of Radiology and Imaging SciencesIndiana University School of MedicineIndianapolisIndianaUSA
| | - Pradeep Varathan Pugalenthi
- Department of Biomedical Engineering and InformaticsIndiana University Luddy School of Informatics, Computing and EngineeringIndianapolisIndianaUSA
| | - Alice Patania
- Department of Mathematics StatisticsUniversity of VermontBurlingtonVermontUSA
| | - Shannon L. Risacher
- Department of Radiology and Imaging SciencesIndiana University School of MedicineIndianapolisIndianaUSA
| | - Kwangsik Nho
- Department of Radiology and Imaging SciencesIndiana University School of MedicineIndianapolisIndianaUSA
| | - Liana G. Apostolova
- Department of Biomedical Engineering and Informatics, Perelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
| | - Li Shen
- Department of Biomedical Engineering and Informatics, Perelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
| | - Andrew J. Saykin
- Department of Radiology and Imaging SciencesIndiana University School of MedicineIndianapolisIndianaUSA
| | - Jingwen Yan
- Department of Biomedical Engineering and InformaticsIndiana University Luddy School of Informatics, Computing and EngineeringIndianapolisIndianaUSA
| | | |
Collapse
|
18
|
Paschali M, Jiang YH, Siegel S, Gonzalez C, Pohl KM, Chaudhari A, Zhao Q. Spectral Graph Sample Weighting for Interpretable Sub-cohort Analysis in Predictive Models for Neuroimaging. PREDICTIVE INTELLIGENCE IN MEDICINE. PRIME (WORKSHOP) 2024; 15155:24-34. [PMID: 39525051 PMCID: PMC11549025 DOI: 10.1007/978-3-031-74561-4_3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/16/2024]
Abstract
Recent advancements in medicine have confirmed that brain disorders often comprise multiple subtypes of mechanisms, developmental trajectories, or severity levels. Such heterogeneity is often associated with demographic aspects (e.g., sex) or disease-related contributors (e.g., genetics). Thus, the predictive power of machine learning models used for symptom prediction varies across subjects based on such factors. To model this heterogeneity, one can assign each training sample a factor-dependent weight, which modulates the subject's contribution to the overall objective loss function. To this end, we propose to model the subject weights as a linear combination of the eigenbases of a spectral population graph that captures the similarity of factors across subjects. In doing so, the learned weights smoothly vary across the graph, highlighting sub-cohorts with high and low predictability. Our proposed sample weighting scheme is evaluated on two tasks. First, we predict initiation of heavy alcohol drinking in young adulthood from imaging and neuropsychological measures from the National Consortium on Alcohol and NeuroDevelopment in Adolescence (NCANDA). Next, we detect Dementia vs. Mild Cognitive Impairment (MCI) using imaging and demographic measurements in subjects from the Alzheimer's Disease Neuroimaging Initiative (ADNI). Compared to existing sample weighting schemes, our sample weights improve interpretability and highlight sub-cohorts with distinct characteristics and varying model accuracy.
Collapse
Affiliation(s)
| | - Yu Hang Jiang
- Department of Statistics, Stanford University, Stanford, USA
| | - Spencer Siegel
- Department of Statistics, Stanford University, Stanford, USA
| | - Camila Gonzalez
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA, USA
| | - Kilian M Pohl
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA, USA
| | - Akshay Chaudhari
- Department of Radiology, Stanford University, Stanford, CA, USA
- Department of Biomedical Data Science, Stanford University, Stanford, CA, USA
| | - Qingyu Zhao
- Department of Radiology, Weill Cornell Medicine, New York, NY, USA
| |
Collapse
|
19
|
Reus LM, Jansen IE, Tijms BM, Visser PJ, Tesi N, van der Lee SJ, Vermunt L, Peeters CFW, De Groot LA, Hok-A-Hin YS, Chen-Plotkin A, Irwin DJ, Hu WT, Meeter LH, van Swieten JC, Holstege H, Hulsman M, Lemstra AW, Pijnenburg YAL, van der Flier WM, Teunissen CE, del Campo Milan M. Connecting dementia risk loci to the CSF proteome identifies pathophysiological leads for dementia. Brain 2024; 147:3522-3533. [PMID: 38527854 PMCID: PMC11449142 DOI: 10.1093/brain/awae090] [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: 09/04/2023] [Revised: 01/29/2024] [Accepted: 02/23/2024] [Indexed: 03/27/2024] Open
Abstract
Genome-wide association studies have successfully identified many genetic risk loci for dementia, but exact biological mechanisms through which genetic risk factors contribute to dementia remains unclear. Integrating CSF proteomic data with dementia risk loci could reveal intermediate molecular pathways connecting genetic variance to the development of dementia. We tested to what extent effects of known dementia risk loci can be observed in CSF levels of 665 proteins [proximity extension-based (PEA) immunoassays] in a deeply-phenotyped mixed memory clinic cohort [n = 502, mean age (standard deviation, SD) = 64.1 (8.7) years, 181 female (35.4%)], including patients with Alzheimer's disease (AD, n = 213), dementia with Lewy bodies (DLB, n = 50) and frontotemporal dementia (FTD, n = 93), and controls (n = 146). Validation was assessed in independent cohorts (n = 99 PEA platform, n = 198, mass reaction monitoring-targeted mass spectroscopy and multiplex assay). We performed additional analyses stratified according to diagnostic status (AD, DLB, FTD and controls separately), to explore whether associations between CSF proteins and genetic variants were specific to disease or not. We identified four AD risk loci as protein quantitative trait loci (pQTL): CR1-CR2 (rs3818361, P = 1.65 × 10-8), ZCWPW1-PILRB (rs1476679, P = 2.73 × 10-32), CTSH-CTSH (rs3784539, P = 2.88 × 10-24) and HESX1-RETN (rs186108507, P = 8.39 × 10-8), of which the first three pQTLs showed direct replication in the independent cohorts. We identified one AD-specific association between a rare genetic variant of TREM2 and CSF IL6 levels (rs75932628, P = 3.90 × 10-7). DLB risk locus GBA showed positive trans effects on seven inter-related CSF levels in DLB patients only. No pQTLs were identified for FTD loci, either for the total sample as for analyses performed within FTD only. Protein QTL variants were involved in the immune system, highlighting the importance of this system in the pathophysiology of dementia. We further identified pQTLs in stratified analyses for AD and DLB, hinting at disease-specific pQTLs in dementia. Dissecting the contribution of risk loci to neurobiological processes aids in understanding disease mechanisms underlying dementia.
Collapse
Affiliation(s)
- Lianne M Reus
- Alzheimer Center Amsterdam, Neurology, Vrije Universiteit Amsterdam, Amsterdam UMC location VUmc, 1081 HZ Amsterdam, The Netherlands
- Amsterdam Neuroscience, Neurodegeneration, Amsterdam, 1081 HV Amsterdam, The Netherlands
- Center for Neurobehavioral Genetics, University of California Los Angeles, Los Angeles, CA 90095 CA, USA
| | - Iris E Jansen
- Alzheimer Center Amsterdam, Neurology, Vrije Universiteit Amsterdam, Amsterdam UMC location VUmc, 1081 HZ Amsterdam, The Netherlands
- Amsterdam Neuroscience, Neurodegeneration, Amsterdam, 1081 HV Amsterdam, The Netherlands
- Department of Complex Trait Genetics, Center for Neurogenomics and Cognitive Research, VU University Amsterdam, 1081 HV Amsterdam, The Netherlands
| | - Betty M Tijms
- Alzheimer Center Amsterdam, Neurology, Vrije Universiteit Amsterdam, Amsterdam UMC location VUmc, 1081 HZ Amsterdam, The Netherlands
- Amsterdam Neuroscience, Neurodegeneration, Amsterdam, 1081 HV Amsterdam, The Netherlands
| | - Pieter Jelle Visser
- Alzheimer Center Amsterdam, Neurology, Vrije Universiteit Amsterdam, Amsterdam UMC location VUmc, 1081 HZ Amsterdam, The Netherlands
- Amsterdam Neuroscience, Neurodegeneration, Amsterdam, 1081 HV Amsterdam, The Netherlands
- Department of Psychiatry, Maastricht University, 6229 ET Maastricht, The Netherlands
| | - Niccoló Tesi
- Alzheimer Center Amsterdam, Neurology, Vrije Universiteit Amsterdam, Amsterdam UMC location VUmc, 1081 HZ Amsterdam, The Netherlands
- Amsterdam Neuroscience, Neurodegeneration, Amsterdam, 1081 HV Amsterdam, The Netherlands
- Genomics of Neurodegenerative Diseases and Aging, Department of Human Genetics, Vrije Universiteit Amsterdam, Amsterdam UMC, 1081 HZ Amsterdam, The Netherlands
| | - Sven J van der Lee
- Alzheimer Center Amsterdam, Neurology, Vrije Universiteit Amsterdam, Amsterdam UMC location VUmc, 1081 HZ Amsterdam, The Netherlands
- Amsterdam Neuroscience, Neurodegeneration, Amsterdam, 1081 HV Amsterdam, The Netherlands
- Genomics of Neurodegenerative Diseases and Aging, Department of Human Genetics, Vrije Universiteit Amsterdam, Amsterdam UMC, 1081 HZ Amsterdam, The Netherlands
| | - Lisa Vermunt
- Alzheimer Center Amsterdam, Neurology, Vrije Universiteit Amsterdam, Amsterdam UMC location VUmc, 1081 HZ Amsterdam, The Netherlands
- Amsterdam Neuroscience, Neurodegeneration, Amsterdam, 1081 HV Amsterdam, The Netherlands
- Neurochemistry Laboratory, Department of Clinical Chemistry, Amsterdam Neuroscience, Amsterdam University Medical Centers, Location VUmc, 1081 HZ Amsterdam, The Netherlands
| | - Carel F W Peeters
- Mathematical and Statistical Methods group (Biometris), Wageningen University and Research, Wageningen, 6708 PB Wageningen, The Netherlands
| | - Lisa A De Groot
- Alzheimer Center Amsterdam, Neurology, Vrije Universiteit Amsterdam, Amsterdam UMC location VUmc, 1081 HZ Amsterdam, The Netherlands
- Amsterdam Neuroscience, Neurodegeneration, Amsterdam, 1081 HV Amsterdam, The Netherlands
| | - Yanaika S Hok-A-Hin
- Neurochemistry Laboratory, Department of Clinical Chemistry, Amsterdam Neuroscience, Amsterdam University Medical Centers, Location VUmc, 1081 HZ Amsterdam, The Netherlands
| | - Alice Chen-Plotkin
- Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - David J Irwin
- Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - William T Hu
- Department of Neurology, Emory University School of Medicine, Atlanta, GA 30322, USA
- Rutgers-RWJ Medical School, Institute for Health, Health Care Policy, and Aging Research, Rutgers Biomedical and Health Sciences, New Brunswick, NJ 08901, USA
| | - Lieke H Meeter
- Department of Neurology and Alzheimer Center, Erasmus Medical Center Rotterdam, Rotterdam, 3015 GD, The Netherlands
| | - John C van Swieten
- Department of Neurology and Alzheimer Center, Erasmus Medical Center Rotterdam, Rotterdam, 3015 GD, The Netherlands
| | - Henne Holstege
- Alzheimer Center Amsterdam, Neurology, Vrije Universiteit Amsterdam, Amsterdam UMC location VUmc, 1081 HZ Amsterdam, The Netherlands
- Amsterdam Neuroscience, Neurodegeneration, Amsterdam, 1081 HV Amsterdam, The Netherlands
- Genomics of Neurodegenerative Diseases and Aging, Department of Human Genetics, Vrije Universiteit Amsterdam, Amsterdam UMC, 1081 HZ Amsterdam, The Netherlands
| | - Marc Hulsman
- Alzheimer Center Amsterdam, Neurology, Vrije Universiteit Amsterdam, Amsterdam UMC location VUmc, 1081 HZ Amsterdam, The Netherlands
- Amsterdam Neuroscience, Neurodegeneration, Amsterdam, 1081 HV Amsterdam, The Netherlands
- Genomics of Neurodegenerative Diseases and Aging, Department of Human Genetics, Vrije Universiteit Amsterdam, Amsterdam UMC, 1081 HZ Amsterdam, The Netherlands
| | - Afina W Lemstra
- Alzheimer Center Amsterdam, Neurology, Vrije Universiteit Amsterdam, Amsterdam UMC location VUmc, 1081 HZ Amsterdam, The Netherlands
- Amsterdam Neuroscience, Neurodegeneration, Amsterdam, 1081 HV Amsterdam, The Netherlands
| | - Yolande A L Pijnenburg
- Alzheimer Center Amsterdam, Neurology, Vrije Universiteit Amsterdam, Amsterdam UMC location VUmc, 1081 HZ Amsterdam, The Netherlands
- Amsterdam Neuroscience, Neurodegeneration, Amsterdam, 1081 HV Amsterdam, The Netherlands
| | - Wiesje M van der Flier
- Alzheimer Center Amsterdam, Neurology, Vrije Universiteit Amsterdam, Amsterdam UMC location VUmc, 1081 HZ Amsterdam, The Netherlands
- Amsterdam Neuroscience, Neurodegeneration, Amsterdam, 1081 HV Amsterdam, The Netherlands
| | - Charlotte E Teunissen
- Neurochemistry Laboratory, Department of Clinical Chemistry, Amsterdam Neuroscience, Amsterdam University Medical Centers, Location VUmc, 1081 HZ Amsterdam, The Netherlands
| | - Marta del Campo Milan
- Neurochemistry Laboratory, Department of Clinical Chemistry, Amsterdam Neuroscience, Amsterdam University Medical Centers, Location VUmc, 1081 HZ Amsterdam, The Netherlands
- Departamento de Ciencias Farmacéuticas y de la Salud, Facultad de Farmacia, Universidad San Pablo-CEU, CEU Universities, Madrid, 28003 Madrid, Spain
- Barcelonaβeta Brain Research Center (BBRC), Pasqual Maragall Foundation, Barcelona, 08005 Barcelona, Spain
| |
Collapse
|
20
|
Howe MD, Caruso MR, Manoochehri M, Kunicki ZJ, Emrani S, Rudolph JL, Huey ED, Salloway SP, Oh H, for the Alzheimer's Disease Neuroimaging Initiative. Utility of cerebrovascular imaging biomarkers to detect cerebral amyloidosis. Alzheimers Dement 2024; 20:7220-7231. [PMID: 39219209 PMCID: PMC11485066 DOI: 10.1002/alz.14207] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2024] [Revised: 07/18/2024] [Accepted: 07/27/2024] [Indexed: 09/04/2024]
Abstract
INTRODUCTION The relationship between cerebrovascular disease (CVD) and amyloid beta (Aβ) in Alzheimer's disease (AD) is understudied. We hypothesized that magnetic resonance imaging (MRI)-based CVD biomarkers-including cerebral microbleeds (CMBs), lacunar infarction, and white matter hyperintensities (WMHs)-would correlate with Aβ positivity on positron emission tomography (Aβ-PET). METHODS We cross-sectionally analyzed data from the Alzheimer's Disease Neuroimaging Initiative (ADNI, N = 1352). Logistic regression was used to calculate odds ratios (ORs), with Aβ-PET positivity as the standard-of-truth. RESULTS Following adjustment, WMHs (OR = 1.25) and superficial CMBs (OR = 1.45) remained positively associated with Aβ-PET positivity (p < 0.001). Deep CMBs and lacunes exhibited a varied relationship with Aβ-PET in cognitive subgroups. The combined diagnostic model, which included CVD biomarkers and other accessible measures, significantly predicted Aβ-PET (pseudo-R2 = 0.41). DISCUSSION The study highlights the translational value of CVD biomarkers in diagnosing AD, and underscores the need for more research on their inclusion in diagnostic criteria. CLINICALTRIALS gov: ADNI-2 (NCT01231971), ADNI-3 (NCT02854033). HIGHLIGHTS Cerebrovascular biomarkers linked to amyloid beta (Aβ) in Alzheimer's disease (AD). White matter hyperintensities and cerebral microbleeds reliably predict Aβ-PET positivity. Relationships with Aβ-PET vary by cognitive stage. Novel accessible model predicts Aβ-PET status. Study supports multimodal diagnostic approaches.
Collapse
Affiliation(s)
- Matthew D. Howe
- Butler Hospital Memory & Aging ProgramProvidenceRhode IslandUSA
- Department of Psychiatry and Human BehaviorBrown UniversityProvidenceRhode IslandUSA
| | - Megan R. Caruso
- Butler Hospital Memory & Aging ProgramProvidenceRhode IslandUSA
| | | | - Zachary J. Kunicki
- Department of Psychiatry and Human BehaviorBrown UniversityProvidenceRhode IslandUSA
| | - Sheina Emrani
- University of Pennsylvania Perelman School of MedicinePhiladelphiaPennsylvaniaUSA
| | - James L. Rudolph
- Center of Innovation in Long‐Term Services and Supports, Providence VA Medical CenterProvidenceRhode IslandUSA
- Department of MedicineThe Warren Alpert Medical School of Brown UniversityProvidenceRhode IslandUSA
| | - Edward D. Huey
- Butler Hospital Memory & Aging ProgramProvidenceRhode IslandUSA
- Department of Psychiatry and Human BehaviorBrown UniversityProvidenceRhode IslandUSA
| | - Stephen P. Salloway
- Butler Hospital Memory & Aging ProgramProvidenceRhode IslandUSA
- Department of Psychiatry and Human BehaviorBrown UniversityProvidenceRhode IslandUSA
| | - Hwamee Oh
- Department of Psychiatry and Human BehaviorBrown UniversityProvidenceRhode IslandUSA
| | | |
Collapse
|
21
|
Puerta R, de Rojas I, García-González P, Olivé C, Sotolongo-Grau O, García-Sánchez A, García-Gutiérrez F, Montrreal L, Pablo Tartari J, Sanabria Á, Pytel V, Lage C, Quintela I, Aguilera N, Rodriguez-Rodriguez E, Alarcón-Martín E, Orellana A, Pastor P, Pérez-Tur J, Piñol-Ripoll G, de Munian AL, García-Alberca JM, Royo JL, Bullido MJ, Álvarez V, Real LM, Anchuelo AC, Gómez-Garre D, Larrad MTM, Franco-Macías E, Mir P, Medina M, Sánchez-Valle R, Dols-Icardo O, Sáez ME, Carracedo Á, Tárraga L, Alegret M, Valero S, Marquié M, Boada M, Juan PS, Cavazos JE, Cabrera A, Cano A, Alzheimer’s Disease Neuroimaging Initiative.. Connecting genomic and proteomic signatures of amyloid burden in the brain. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.09.06.24313124. [PMID: 39281766 PMCID: PMC11398581 DOI: 10.1101/2024.09.06.24313124] [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: 09/18/2024]
Abstract
Background Alzheimer's disease (AD) has a high heritable component characteristic of complex diseases, yet many of the genetic risk factors remain unknown. We combined genome-wide association studies (GWAS) on amyloid endophenotypes measured in cerebrospinal fluid (CSF) and positron emission tomography (PET) as surrogates of amyloid pathology, which may be helpful to understand the underlying biology of the disease. Methods We performed a meta-analysis of GWAS of CSF Aβ42 and PET measures combining six independent cohorts (n=2,076). Due to the opposite effect direction of Aβ phenotypes in CSF and PET measures, only genetic signals in the opposite direction were considered for analysis (n=376,599). Polygenic risk scores (PRS) were calculated and evaluated for AD status and amyloid endophenotypes. We then searched the CSF proteome signature of brain amyloidosis using SOMAscan proteomic data (Ace cohort, n=1,008) and connected it with GWAS results of loci modulating amyloidosis. Finally, we compared our results with a large meta-analysis using publicly available datasets in CSF (n=13,409) and PET (n=13,116). This combined approach enabled the identification of overlapping genes and proteins associated with amyloid burden and the assessment of their biological significance using enrichment analyses. Results After filtering the meta-GWAS, we observed genome-wide significance in the rs429358-APOE locus and nine suggestive hits were annotated. We replicated the APOE loci using the large CSF-PET meta-GWAS and identified multiple AD-associated genes as well as the novel GADL1 locus. Additionally, we found a significant association between the AD PRS and amyloid levels, whereas no significant association was found between any Aβ PRS with AD risk. CSF SOMAscan analysis identified 1,387 FDR-significant proteins associated with CSF Aβ42 levels. The overlap among GWAS loci and proteins associated with amyloid burden was very poor (n=35). The enrichment analysis of overlapping hits strongly suggested several signalling pathways connecting amyloidosis with the anchored component of the plasma membrane, synapse physiology and mental disorders that were replicated in the large CSF-PET meta-analysis. Conclusions The strategy of combining CSF and PET amyloid endophenotypes GWAS with CSF proteome analyses might be effective for identifying signals associated with the AD pathological process and elucidate causative molecular mechanisms behind the amyloid mobilization in AD.
Collapse
Affiliation(s)
- Raquel Puerta
- Ace Alzheimer Center Barcelona – Universitat Internacional de Catalunya, Spain
- Universitat de Barcelona (UB)
| | - Itziar de Rojas
- Ace Alzheimer Center Barcelona – Universitat Internacional de Catalunya, Spain
- CIBERNED, Network Center for Biomedical Research in Neurodegenerative Diseases, National Institute of Health Carlos III, Madrid, Spain
| | - Pablo García-González
- Ace Alzheimer Center Barcelona – Universitat Internacional de Catalunya, Spain
- CIBERNED, Network Center for Biomedical Research in Neurodegenerative Diseases, National Institute of Health Carlos III, Madrid, Spain
| | - Clàudia Olivé
- Ace Alzheimer Center Barcelona – Universitat Internacional de Catalunya, Spain
| | | | | | | | - Laura Montrreal
- Ace Alzheimer Center Barcelona – Universitat Internacional de Catalunya, Spain
| | - Juan Pablo Tartari
- Ace Alzheimer Center Barcelona – Universitat Internacional de Catalunya, Spain
| | - Ángela Sanabria
- Ace Alzheimer Center Barcelona – Universitat Internacional de Catalunya, Spain
- CIBERNED, Network Center for Biomedical Research in Neurodegenerative Diseases, National Institute of Health Carlos III, Madrid, Spain
| | - Vanesa Pytel
- Ace Alzheimer Center Barcelona – Universitat Internacional de Catalunya, Spain
- CIBERNED, Network Center for Biomedical Research in Neurodegenerative Diseases, National Institute of Health Carlos III, Madrid, Spain
| | - Carmen Lage
- CIBERNED, Network Center for Biomedical Research in Neurodegenerative Diseases, National Institute of Health Carlos III, Madrid, Spain
- Neurology Service, Marqués de Valdecilla University Hospital (University of Cantabria and IDIVAL), Santander, Spain
| | - Inés Quintela
- Grupo de Medicina Xenómica, Centro Nacional de Genotipado (CEGEN-PRB3-ISCIII). Universidade de Santiago de Compostela, Santiago de Compostela, Spain
| | - Nuria Aguilera
- Ace Alzheimer Center Barcelona – Universitat Internacional de Catalunya, Spain
| | - Eloy Rodriguez-Rodriguez
- CIBERNED, Network Center for Biomedical Research in Neurodegenerative Diseases, National Institute of Health Carlos III, Madrid, Spain
- Neurology Service, Marqués de Valdecilla University Hospital (University of Cantabria and IDIVAL), Santander, Spain
| | | | - Adelina Orellana
- Ace Alzheimer Center Barcelona – Universitat Internacional de Catalunya, Spain
- CIBERNED, Network Center for Biomedical Research in Neurodegenerative Diseases, National Institute of Health Carlos III, Madrid, Spain
| | - Pau Pastor
- Unit of Neurodegenerative diseases, Department of Neurology, University Hospital Germans Trias i Pujol, Badalona, Barcelona, Spain
- The Germans Trias i Pujol Research Institute (IGTP), Badalona, Barcelona, Spain
| | - Jordi Pérez-Tur
- CIBERNED, Network Center for Biomedical Research in Neurodegenerative Diseases, National Institute of Health Carlos III, Madrid, Spain
- Unitat de Genètica Molecular, Institut de Biomedicina de València-CSIC, Valencia, Spain
- Unidad Mixta de Neurologia Genètica, Instituto de Investigación Sanitaria La Fe, Valencia, Spain
| | - Gerard Piñol-Ripoll
- Unitat Trastorns Cognitius, Hospital Universitari Santa Maria de Lleida, Lleida, Spain
- Institut de Recerca Biomedica de Lleida (IRBLLeida), Lleida, Spain
| | - Adolfo López de Munian
- CIBERNED, Network Center for Biomedical Research in Neurodegenerative Diseases, National Institute of Health Carlos III, Madrid, Spain
- Department of Neurology. Hospital Universitario Donostia. San Sebastian, Spain
- Department of Neurosciences. Faculty of Medicine and Nursery. University of the Basque Country, San Sebastián, Spain
- Neurosciences Area. Instituto Biodonostia. San Sebastian, Spain
| | - Jose María García-Alberca
- CIBERNED, Network Center for Biomedical Research in Neurodegenerative Diseases, National Institute of Health Carlos III, Madrid, Spain
- Alzheimer Research Center & Memory Clinic, Andalusian Institute for Neuroscience, Málaga, Spain
| | - Jose Luís Royo
- Departamento de Especialidades Quirúrgicas, Bioquímica e Inmunología. School of Medicine. University of Malaga. Málaga, Spain
| | - María Jesús Bullido
- CIBERNED, Network Center for Biomedical Research in Neurodegenerative Diseases, National Institute of Health Carlos III, Madrid, Spain
- Centro de Biología Molecular Severo Ochoa (UAM-CSIC)
- Instituto de Investigacion Sanitaria ‘Hospital la Paz’ (IdIPaz), Madrid, Spain
- Universidad Autónoma de Madrid
| | - Victoria Álvarez
- Laboratorio de Genética. Hospital Universitario Central de Asturias, Oviedo, Spain
- Instituto de Investigación Sanitaria del Principado de Asturias (ISPA)
| | - Luis Miguel Real
- Departamento de Especialidades Quirúrgicas, Bioquímica e Inmunología. School of Medicine. University of Malaga. Málaga, Spain
- Unidad Clínica de Enfermedades Infecciosas y Microbiología.Hospital Universitario de Valme, Sevilla, Spain
| | - Arturo Corbatón Anchuelo
- Instituto de Investigación Sanitaria del Hospital Clínico San Carlos (IdISSC), Hospital Clínico San Carlos
| | - Dulcenombre Gómez-Garre
- Instituto de Investigación Sanitaria del Hospital Clínico San Carlos (IdISSC), Hospital Clínico San Carlos
- Laboratorio de Riesgo Cardiovascular y Microbiota, Hospital Clínico San Carlos; Departamento de Fisiología, Facultad de Medicina, Universidad Complutense de Madrid (UCM)
- Biomedical Research Networking Center in Cardiovascular Diseases (CIBERCV), Madrid, Spain
| | - María Teresa Martínez Larrad
- Instituto de Investigación Sanitaria del Hospital Clínico San Carlos (IdISSC), Hospital Clínico San Carlos
- Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM)
| | - Emilio Franco-Macías
- Dementia Unit, Department of Neurology, Hospital Universitario Virgen del Rocío, Instituto de Biomedicina de Sevilla (IBiS), Sevilla, Spain
| | - Pablo Mir
- CIBERNED, Network Center for Biomedical Research in Neurodegenerative Diseases, National Institute of Health Carlos III, Madrid, Spain
- Unidad de Trastornos del Movimiento, Servicio de Neurología y Neurofisiología. Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Seville, Spain
| | - Miguel Medina
- CIBERNED, Network Center for Biomedical Research in Neurodegenerative Diseases, National Institute of Health Carlos III, Madrid, Spain
- CIEN Foundation/Queen Sofia Foundation Alzheimer Center
| | - Raquel Sánchez-Valle
- Alzheimer’s disease and other cognitive disorders unit. Service of Neurology. Hospital Clínic of Barcelona. Institut d’Investigacions Biomèdiques August Pi i Sunyer, University of Barcelona, Barcelona, Spain
| | - Oriol Dols-Icardo
- Department of Neurology, Sant Pau Memory Unit, Sant Pau Biomedical Research Institute, Hospital de la Santa Creu i Sant Pau, Universitat Autònoma de Barcelona, Barcelona, Spain
| | | | - Ángel Carracedo
- Grupo de Medicina Xenómica, Centro Nacional de Genotipado (CEGEN-PRB3-ISCIII). Universidade de Santiago de Compostela, Santiago de Compostela, Spain
- Fundación Pública Galega de Medicina Xenómica – CIBERER-IDIS, Santiago de Compostela, Spain
| | - Lluís Tárraga
- Ace Alzheimer Center Barcelona – Universitat Internacional de Catalunya, Spain
- CIBERNED, Network Center for Biomedical Research in Neurodegenerative Diseases, National Institute of Health Carlos III, Madrid, Spain
| | - Montse Alegret
- Ace Alzheimer Center Barcelona – Universitat Internacional de Catalunya, Spain
- CIBERNED, Network Center for Biomedical Research in Neurodegenerative Diseases, National Institute of Health Carlos III, Madrid, Spain
| | - Sergi Valero
- Ace Alzheimer Center Barcelona – Universitat Internacional de Catalunya, Spain
- CIBERNED, Network Center for Biomedical Research in Neurodegenerative Diseases, National Institute of Health Carlos III, Madrid, Spain
| | - Marta Marquié
- Ace Alzheimer Center Barcelona – Universitat Internacional de Catalunya, Spain
- CIBERNED, Network Center for Biomedical Research in Neurodegenerative Diseases, National Institute of Health Carlos III, Madrid, Spain
| | - Mercè Boada
- Ace Alzheimer Center Barcelona – Universitat Internacional de Catalunya, Spain
- CIBERNED, Network Center for Biomedical Research in Neurodegenerative Diseases, National Institute of Health Carlos III, Madrid, Spain
| | - Pascual Sánchez Juan
- CIBERNED, Network Center for Biomedical Research in Neurodegenerative Diseases, National Institute of Health Carlos III, Madrid, Spain
- Neurology Service, Marqués de Valdecilla University Hospital (University of Cantabria and IDIVAL), Santander, Spain
| | - Jose Enrique Cavazos
- South Texas Medical Science Training Program, University of Texas Health San Antonio, San Antonio
- Glenn Biggs Institute for Alzheimer’s & Neurodegenerative Diseases, University of Texas Health Science Center at San Antonio, 7703 Floyd Curl Dr, San Antonio, TX 78229 USA
| | - Alfredo Cabrera
- Neuroscience Therapeutic Area, Janssen Research & Development, Turnhoutseweg 30, 2340 Beerse, Belgium
| | - Amanda Cano
- Ace Alzheimer Center Barcelona – Universitat Internacional de Catalunya, Spain
- CIBERNED, Network Center for Biomedical Research in Neurodegenerative Diseases, National Institute of Health Carlos III, Madrid, Spain
| | - Alzheimer’s Disease Neuroimaging Initiative.
- Ace Alzheimer Center Barcelona – Universitat Internacional de Catalunya, Spain
- CIBERNED, Network Center for Biomedical Research in Neurodegenerative Diseases, National Institute of Health Carlos III, Madrid, Spain
- Glenn Biggs Institute for Alzheimer’s & Neurodegenerative Diseases, University of Texas Health Science Center at San Antonio, 7703 Floyd Curl Dr, San Antonio, TX 78229 USA
| |
Collapse
|
22
|
Qian S, Zheng Y, Jiang T, Hou J, Cao R, Cai J, Ma E, Wang W, Song W, Xie C. A Risk Variant rs6922617 in TREM Is Discrepantly Associated With Defining Neuropathological Hallmarks in the Alzheimer's Continuum. J Gerontol A Biol Sci Med Sci 2024; 79:glae185. [PMID: 39051708 DOI: 10.1093/gerona/glae185] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2024] [Indexed: 07/27/2024] Open
Abstract
The single nucleotide polymorphism (SNP)-rs6922617 in the triggering receptor expressed on myeloid cells (TREM) gene cluster is a potential risk factor for Alzheimer's disease (AD). Here, we examined whether rs6922617 is associated with AD-defining neuropathological hallmarks and memory performance. We assessed the interaction between the variant rs6922617 and levels of beta-amyloid (Aβ), tau pathology, neurodegeneration, namely amyloid-tau-neurodegeneration framework, and cognition functions in 660 healthy controls, 794 mild cognitively impaired, and 272 subjects with AD. We employed linear regression and linear mixed models to examine the association. Here we find that the SNP-rs6922617 in the TREM gene cluster is associated with a higher global amyloid-ligands positron emission tomography (Aβ-PET) burden and lower fluorodeoxyglucose positron emission tomography (FDG-PET) load. Interestingly, rs6922617 risk allele carriers exhibit a significantly reduced tau accumulation compared to the non-carriers, indicating a discrepant association with Aβ and tau pathologies. Though the participants carrying the rs6922617 risk allele do not show a correlation with poorer cognitive performance, stronger neuropathological phenotypes, and memory impairments are evident in ApoE ε4 carriers with the rs6922617 risk allele. These results support the notion that the SNP-rs6922617 in the TREM gene cluster is associated with AD-related neuropathological hallmarks, such as Aβ and FDG-mediated neurodegeneration, rather than tau accumulation. Although the direct association with memory impairment in the Alzheimer's continuum remains inconclusive, our findings suggest a potential role of rs6922617 in facilitating neuropathology hallmarks.
Collapse
Affiliation(s)
- Shuangjie Qian
- Department of Neurology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Yi Zheng
- Department of Neurology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Tao Jiang
- Department of Neurology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Jialong Hou
- Department of Neurology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Ruixue Cao
- The Second Affiliated Hospital and Yuying Children's Hospital, Wenzhou Medical University, Wenzhou, China
- Key Laboratory of Alzheimer's Disease of Zhejiang Province, Institute of Aging, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Jinlai Cai
- Department of Neurology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Enzi Ma
- Department of Neurology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Wenwen Wang
- The Center of Traditional Chinese Medicine, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| | - Weihong Song
- Key Laboratory of Alzheimer's Disease of Zhejiang Province, Institute of Aging, Wenzhou Medical University, Wenzhou, Zhejiang, China
- Oujiang Laboratory, Wenzhou, Zhejiang, China
| | - Chenglong Xie
- Department of Neurology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
- Key Laboratory of Alzheimer's Disease of Zhejiang Province, Institute of Aging, Wenzhou Medical University, Wenzhou, Zhejiang, China
| |
Collapse
|
23
|
Oh S, Kim S, Lee JE, Park BY, Hye Won J, Park H. Multimodal analysis of disease onset in Alzheimer's disease using Connectome, Molecular, and genetics data. Neuroimage Clin 2024; 43:103660. [PMID: 39197213 PMCID: PMC11393605 DOI: 10.1016/j.nicl.2024.103660] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2024] [Revised: 08/23/2024] [Accepted: 08/23/2024] [Indexed: 09/01/2024]
Abstract
Alzheimer's disease (AD) and its related age at onset (AAO) are highly heterogeneous, due to the inherent complexity of the disease. They are affected by multiple factors, such as neuroimaging and genetic predisposition. Multimodal integration of various data types is necessary; however, it has been nontrivial due to the high dimensionality of each modality. We aimed to identify multimodal biomarkers of AAO in AD using an extended version of sparse canonical correlation analysis, in which we integrated two imaging modalities, functional magnetic resonance imaging (fMRI) and positron emission tomography (PET), and genetic data in the form of single-nucleotide polymorphisms (SNPs) obtained from the Alzheimer's disease neuroimaging initiative database. These three modalities cover low-to-high-level complementary information and offer multiscale insights into the AAO. We identified multivariate markers of AAO in AD using fMRI, PET, and SNP. Furthermore, the markers identified were largely consistent with those reported in the existing literature. In particular, our serial mediation analysis suggests that genetic variants influence the AAO in AD by indirectly affecting brain connectivity by mediation of amyloid-beta protein accumulation, supporting a plausible path in existing research. Our approach provides comprehensive biomarkers related to AAO in AD and offers novel multimodal insights into AD.
Collapse
Affiliation(s)
- Sewook Oh
- Department of Electrical and Computer Engineering, Sungkyunkwan University, Suwon, Republic of Korea
| | - Sunghun Kim
- Department of Electrical and Computer Engineering, Sungkyunkwan University, Suwon, Republic of Korea; Department of Artificial Intelligence, Sungkyunkwan University, Suwon, Republic of Korea; Center for Neuroscience Imaging Research, Institute for Basic Science, Suwon, Republic of Korea
| | - Jong-Eun Lee
- Department of Electrical and Computer Engineering, Sungkyunkwan University, Suwon, Republic of Korea; Center for Neuroscience Imaging Research, Institute for Basic Science, Suwon, Republic of Korea
| | - Bo-Yong Park
- Center for Neuroscience Imaging Research, Institute for Basic Science, Suwon, Republic of Korea; Department of Brain and Cognitive Engineering, Korea University, Seoul, Republic of Korea
| | - Ji Hye Won
- Department of Computer Engineering, Pukyong National University, Busan, Republic of Korea
| | - Hyunjin Park
- Department of Electrical and Computer Engineering, Sungkyunkwan University, Suwon, Republic of Korea; Department of Artificial Intelligence, Sungkyunkwan University, Suwon, Republic of Korea; Center for Neuroscience Imaging Research, Institute for Basic Science, Suwon, Republic of Korea.
| |
Collapse
|
24
|
Andrews SJ, Boeriu AI, Belloy ME, Renton AE, Fulton-Howard B, Brenowitz WD, Yaffe K, Alzheimer’s Disease Neuroimaging Initiative. Dementia Risk Scores, APOE, and risk of Alzheimer disease: one size does not fit all. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.04.27.24306486. [PMID: 38746455 PMCID: PMC11092714 DOI: 10.1101/2024.04.27.24306486] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/16/2024]
Abstract
Introduction Evaluating the generalizability of dementia risk scores, primarily developed in non-Latinx White (NLW) participants, and interactions with genetic risk factors in diverse populations is crucial for addressing health disparities. Methods We analyzed the association of the Cardiovascular Risk Factors, Aging, and Incidence of Dementia (CAIDE) and modified CAIDE (mCAIDE) scores with dementia risk using logistic regression models stratified by race/ethnicity in NACC and ADNI, and assessed their interaction with APOE. Results Higher CAIDE scores were associated with an increased risk of dementia in Asian, Latinx, and NLW participants but not in Black participants. In contrast, higher mCAIDE scores were also associated with an increased risk of dementia in Black participants. Unfavorable mCAIDE risk profiles exacerbated the APOE*ε4 risk effect and attenuated the APOE*ε2 protective effect. Discussion Our findings underscore the importance of evaluating the validity of dementia risk scores in diverse populations for their use in personalized medicine approaches to promote brain health.
Collapse
Affiliation(s)
- Shea J. Andrews
- Department of Psychiatry and Behavioral Sciences, University of California San Francisco, 505 Parnassus Ave, San Francisco, CA, USA, 94143
| | - Ana I. Boeriu
- Department of Psychiatry and Behavioral Sciences, University of California San Francisco, 505 Parnassus Ave, San Francisco, CA, USA, 94143
| | - Michael E. Belloy
- Department of Neurology and Neurological Sciences, Stanford University, Stanford, CA, USA, 94304
- NeuroGenomics and Informatics Center, Washington University School of Medicine, St.Louis, MO, USA, 63108
- Department of Neurology, Washington University School of Medicine, St.Louis, MO, USA, 63108
| | - Alan E. Renton
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, 1428 Madison Ave, New York, NY, USA, 10029
| | - Brian Fulton-Howard
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, 1428 Madison Ave, New York, NY, USA, 10029
| | - Willa D. Brenowitz
- Kaiser Permanente Center for Health Research, 3800 N Interstate Ave, Portland, OR, USA, 97227
- Department of Epidemiology and Biostatistics, University of California San Francisco, 505 Parnassus Ave, San Francisco, CA, USA, 94143
| | - Kristine Yaffe
- Department of Psychiatry and Behavioral Sciences, University of California San Francisco, 505 Parnassus Ave, San Francisco, CA, USA, 94143
- Department of Epidemiology and Biostatistics, University of California San Francisco, 505 Parnassus Ave, San Francisco, CA, USA, 94143
- Department of Neurology, University of California San Francisco, 505 Parnassus Ave, San Francisco, CA, USA, 94143
| | | |
Collapse
|
25
|
Altmann A, Aksman LM, Oxtoby NP, Young AL, Alexander DC, Barkhof F, Shoai M, Hardy J, Schott JM. Towards cascading genetic risk in Alzheimer's disease. Brain 2024; 147:2680-2690. [PMID: 38820112 PMCID: PMC11292901 DOI: 10.1093/brain/awae176] [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: 12/21/2023] [Revised: 04/22/2024] [Accepted: 05/06/2024] [Indexed: 06/02/2024] Open
Abstract
Alzheimer's disease typically progresses in stages, which have been defined by the presence of disease-specific biomarkers: amyloid (A), tau (T) and neurodegeneration (N). This progression of biomarkers has been condensed into the ATN framework, in which each of the biomarkers can be either positive (+) or negative (-). Over the past decades, genome-wide association studies have implicated ∼90 different loci involved with the development of late-onset Alzheimer's disease. Here, we investigate whether genetic risk for Alzheimer's disease contributes equally to the progression in different disease stages or whether it exhibits a stage-dependent effect. Amyloid (A) and tau (T) status was defined using a combination of available PET and CSF biomarkers in the Alzheimer's Disease Neuroimaging Initiative cohort. In 312 participants with biomarker-confirmed A-T- status, we used Cox proportional hazards models to estimate the contribution of APOE and polygenic risk scores (beyond APOE) to convert to A+T- status (65 conversions). Furthermore, we repeated the analysis in 290 participants with A+T- status and investigated the genetic contribution to conversion to A+T+ (45 conversions). Both survival analyses were adjusted for age, sex and years of education. For progression from A-T- to A+T-, APOE-e4 burden showed a significant effect [hazard ratio (HR) = 2.88; 95% confidence interval (CI): 1.70-4.89; P < 0.001], whereas polygenic risk did not (HR = 1.09; 95% CI: 0.84-1.42; P = 0.53). Conversely, for the transition from A+T- to A+T+, the contribution of APOE-e4 burden was reduced (HR = 1.62; 95% CI: 1.05-2.51; P = 0.031), whereas the polygenic risk showed an increased contribution (HR = 1.73; 95% CI: 1.27-2.36; P < 0.001). The marginal APOE effect was driven by e4 homozygotes (HR = 2.58; 95% CI: 1.05-6.35; P = 0.039) as opposed to e4 heterozygotes (HR = 1.74; 95% CI: 0.87-3.49; P = 0.12). The genetic risk for late-onset Alzheimer's disease unfolds in a disease stage-dependent fashion. A better understanding of the interplay between disease stage and genetic risk can lead to a more mechanistic understanding of the transition between ATN stages and a better understanding of the molecular processes leading to Alzheimer's disease, in addition to opening therapeutic windows for targeted interventions.
Collapse
Affiliation(s)
- Andre Altmann
- UCL Centre for Medical Image Computing, Department of Medical Physics and Biomedical Engineering, University College London, London, WC1E 6BT, UK
| | - Leon M Aksman
- Stevens Neuroimaging and Informatics Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Neil P Oxtoby
- UCL Centre for Medical Image Computing, Department of Computer Science, University College London, London, WC1E 6BT, UK
| | - Alexandra L Young
- UCL Centre for Medical Image Computing, Department of Computer Science, University College London, London, WC1E 6BT, UK
| | - Daniel C Alexander
- UCL Centre for Medical Image Computing, Department of Computer Science, University College London, London, WC1E 6BT, UK
| | - Frederik Barkhof
- UCL Centre for Medical Image Computing, Department of Medical Physics and Biomedical Engineering, University College London, London, WC1E 6BT, UK
- UCL Queen Square Institute of Neurology, University College London, London, WC1N 3BG, UK
- Department of Radiology and Nuclear Medicine, Amsterdam University Medical Center, Amsterdam, 1081 HV, The Netherlands
| | - Maryam Shoai
- UCL Queen Square Institute of Neurology, University College London, London, WC1N 3BG, UK
- UK Dementia Research Institute, University College London, London, WC1E 6BT, UK
| | - John Hardy
- UCL Queen Square Institute of Neurology, University College London, London, WC1N 3BG, UK
- UK Dementia Research Institute, University College London, London, WC1E 6BT, UK
| | - Jonathan M Schott
- UK Dementia Research Institute, University College London, London, WC1E 6BT, UK
- Dementia Research Centre, UCL Queen Square Institute of Neurology, University College London, London, WC1N 3AR, UK
| |
Collapse
|
26
|
Barrett JS. Artificial Intelligence Opportunities to Guide Precision Dosing Strategies. J Pediatr Pharmacol Ther 2024; 29:434-440. [PMID: 39144390 PMCID: PMC11321806 DOI: 10.5863/1551-6776-29.4.434] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2024] [Accepted: 03/12/2024] [Indexed: 08/16/2024]
|
27
|
Al Olaimat M, Bozdag S, for the Alzheimer’s Disease Neuroimaging Initiative. TA-RNN: an attention-based time-aware recurrent neural network architecture for electronic health records. Bioinformatics 2024; 40:i169-i179. [PMID: 38940180 PMCID: PMC11211851 DOI: 10.1093/bioinformatics/btae264] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/29/2024] Open
Abstract
MOTIVATION Electronic health records (EHRs) represent a comprehensive resource of a patient's medical history. EHRs are essential for utilizing advanced technologies such as deep learning (DL), enabling healthcare providers to analyze extensive data, extract valuable insights, and make precise and data-driven clinical decisions. DL methods such as recurrent neural networks (RNN) have been utilized to analyze EHR to model disease progression and predict diagnosis. However, these methods do not address some inherent irregularities in EHR data such as irregular time intervals between clinical visits. Furthermore, most DL models are not interpretable. In this study, we propose two interpretable DL architectures based on RNN, namely time-aware RNN (TA-RNN) and TA-RNN-autoencoder (TA-RNN-AE) to predict patient's clinical outcome in EHR at the next visit and multiple visits ahead, respectively. To mitigate the impact of irregular time intervals, we propose incorporating time embedding of the elapsed times between visits. For interpretability, we propose employing a dual-level attention mechanism that operates between visits and features within each visit. RESULTS The results of the experiments conducted on Alzheimer's Disease Neuroimaging Initiative (ADNI) and National Alzheimer's Coordinating Center (NACC) datasets indicated the superior performance of proposed models for predicting Alzheimer's Disease (AD) compared to state-of-the-art and baseline approaches based on F2 and sensitivity. Additionally, TA-RNN showed superior performance on the Medical Information Mart for Intensive Care (MIMIC-III) dataset for mortality prediction. In our ablation study, we observed enhanced predictive performance by incorporating time embedding and attention mechanisms. Finally, investigating attention weights helped identify influential visits and features in predictions. AVAILABILITY AND IMPLEMENTATION https://github.com/bozdaglab/TA-RNN.
Collapse
Affiliation(s)
- Mohammad Al Olaimat
- Department of Computer Science and Engineering, University of North Texas, Denton, TX 76203, United States
- BioDiscovery Institute, University of North Texas, Denton, TX 76203, United States
| | - Serdar Bozdag
- Department of Computer Science and Engineering, University of North Texas, Denton, TX 76203, United States
- BioDiscovery Institute, University of North Texas, Denton, TX 76203, United States
- Department of Mathematics, University of North Texas, Denton, TX 76203, United States
| | | |
Collapse
|
28
|
Howe MD, Caruso MR, Manoochehri M, Kunicki ZJ, Emrani S, Rudolph JL, Huey ED, Salloway SP, Oh H. Utility of cerebrovascular imaging biomarkers to detect cerebral amyloidosis. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.05.28.24308056. [PMID: 38853879 PMCID: PMC11160821 DOI: 10.1101/2024.05.28.24308056] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2024]
Abstract
INTRODUCTION The relationship between cerebrovascular disease (CVD) and amyloid-β (Aβ) in Alzheimer disease (AD) is understudied. We hypothesized that magnetic resonance imaging (MRI)-based CVD biomarkers, including cerebral microbleeds (CMBs), ischemic infarction, and white matter hyperintensities (WMH), would correlate with Aβ positivity on positron emission tomography (Aβ-PET). METHODS We cross-sectionally analyzed data from the Alzheimer's Disease Neuroimaging Initiative (ADNI, N=1,352). Logistic regression was used to calculate odds ratios (ORs), with Aβ-PET positivity as the standard-of-truth. RESULTS Following adjustment, WMH (OR=1.25) and superficial CMBs (OR=1.45) remained positively associated with Aβ-PET positivity (p<.001). Deep CMBs and infarcts exhibited a varied relationship with Aβ-PET in cognitive subgroups. The combined diagnostic model, which included CVD biomarkers and other accessible measures, significantly predicted Aβ-PET (pseudo-R 2 =.41). DISCUSSION The study highlights the translational value of CVD biomarkers in diagnosing AD, and underscores the need for more research on their inclusion in diagnostic criteria. ClinicalTrials.gov: ADNI-2 ( NCT01231971 ), ADNI-3 ( NCT02854033 ).
Collapse
|
29
|
Zhang S, Zhou Y, Geng P, Lu Q. Functional Neural Networks for High-Dimensional Genetic Data Analysis. IEEE/ACM TRANSACTIONS ON COMPUTATIONAL BIOLOGY AND BIOINFORMATICS 2024; 21:383-393. [PMID: 38507390 PMCID: PMC11301578 DOI: 10.1109/tcbb.2024.3364614] [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] [Indexed: 03/22/2024]
Abstract
Artificial intelligence (AI) is a thriving research field with many successful applications in areas such as computer vision and speech recognition. Machine learning methods, such as artificial neural networks (ANN), play a central role in modern AI technology. While ANN also holds great promise for human genetic research, the high-dimensional genetic data and complex genetic structure bring tremendous challenges. The vast majority of genetic variants on the genome have small or no effects on diseases, and fitting ANN on a large number of variants without considering the underlying genetic structure (e.g., linkage disequilibrium) could bring a serious overfitting issue. Furthermore, while a single disease phenotype is often studied in a classic genetic study, in emerging research fields (e.g., imaging genetics), researchers need to deal with different types of disease phenotypes. To address these challenges, we propose a functional neural networks (FNN) method. FNN uses a series of basis functions to model high-dimensional genetic data and a variety of phenotype data and further builds a multi-layer functional neural network to capture the complex relationships between genetic variants and disease phenotypes. Through simulations, we demonstrate the advantages of FNN for high-dimensional genetic data analysis in terms of robustness and accuracy. The real data applications also showed that FNN attained higher accuracy than the existing methods.
Collapse
|
30
|
Cheek CL, Lindner P, Grigorenko EL. Statistical and Machine Learning Analysis in Brain-Imaging Genetics: A Review of Methods. Behav Genet 2024; 54:233-251. [PMID: 38336922 DOI: 10.1007/s10519-024-10177-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2023] [Accepted: 01/24/2024] [Indexed: 02/12/2024]
Abstract
Brain-imaging-genetic analysis is an emerging field of research that aims at aggregating data from neuroimaging modalities, which characterize brain structure or function, and genetic data, which capture the structure and function of the genome, to explain or predict normal (or abnormal) brain performance. Brain-imaging-genetic studies offer great potential for understanding complex brain-related diseases/disorders of genetic etiology. Still, a combined brain-wide genome-wide analysis is difficult to perform as typical datasets fuse multiple modalities, each with high dimensionality, unique correlational landscapes, and often low statistical signal-to-noise ratios. In this review, we outline the progress in brain-imaging-genetic methodologies starting from early massive univariate to current deep learning approaches, highlighting each approach's strengths and weaknesses and elongating it with the field's development. We conclude by discussing selected remaining challenges and prospects for the field.
Collapse
Affiliation(s)
- Connor L Cheek
- Texas Institute for Evaluation, Measurement, and Statistics, University of Houston, Houston, TX, USA.
- Department of Physics, University of Houston, Houston, TX, USA.
| | - Peggy Lindner
- Texas Institute for Evaluation, Measurement, and Statistics, University of Houston, Houston, TX, USA
- Department of Information Science Technology, University of Houston, Houston, TX, USA
| | - Elena L Grigorenko
- Texas Institute for Evaluation, Measurement, and Statistics, University of Houston, Houston, TX, USA
- Department of Psychology, University of Houston, Houston, TX, USA
- Baylor College of Medicine, Houston, TX, USA
- Sirius University of Science and Technology, Sochi, Russia
| |
Collapse
|
31
|
Lacey C, Paterson T, Gawryluk JR, for the Alzheimer’s Disease Neuroimaging Initiative. Impact of APOE-ε alleles on brain structure and cognitive function in healthy older adults: A VBM and DTI replication study. PLoS One 2024; 19:e0292576. [PMID: 38635499 PMCID: PMC11025752 DOI: 10.1371/journal.pone.0292576] [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: 05/18/2023] [Accepted: 09/22/2023] [Indexed: 04/20/2024] Open
Abstract
BACKGROUND The Apolipoprotein E (APOE) gene has been established in the Alzheimer's disease (AD) literature to impact brain structure and function and may also show congruent effects in healthy older adults, although findings in this population are much less consistent. The current study aimed to replicate and expand the multimodal approach employed by Honea et al. Structural magnetic resonance imaging (MRI), diffusion tensor imaging (DTI), and neuropsychological measures were used to investigate the impact of APOE-ε status on grey matter structure, white matter integrity, and cognitive functioning. METHODS Data were obtained from the Alzheimer's Disease Initiative Phase 3 (ADNI3) database. Baseline MRI, DTI and cognitive composite scores for memory (ADNI-Mem) and executive function (ADNI-EF) were acquired from 116 healthy controls. Participants were grouped according to APOE allele presence (APOE-ε2+ N = 17, APOE-ε3ε3 N = 64, APOE-ε4+ N = 35). Voxel-based morphometry (VBM) and tract-based spatial statistics (TBSS) were used to compare grey matter volume (GMV) and white matter integrity, respectively, between APOE-ε2+ and APOE-ε3ε3 controls, and again between APOE-ε4+ and APOE-ε3ε3 controls. Multivariate analysis of covariance (MANCOVA) was used to examine the effects of APOE polymorphism on memory and EF across all APOE groups with age, sex and education as regressors of no interest. Cognitive scores were correlated (Pearson r) with imaging metrics within groups. RESULTS No significant differences were seen across groups, within groups in MRI metrics, or cognitive performance (p>0.05, corrected for multiple comparisons). CONCLUSIONS The current study partially replicated and extended previous findings from an earlier multimodal study (Honea 2009). Future studies should clarify APOE mechanisms in healthy ageing by adding other imaging, cognitive, and lifestyle metrics and longitudinal design in larger sample sizes.
Collapse
Affiliation(s)
- Colleen Lacey
- Department of Psychology, University of Victoria, Victoria, British Columbia, Canada
- Institute on Aging and Lifelong Health, University of Victoria, Victoria, British Columbia, Canada
| | - Theone Paterson
- Department of Psychology, University of Victoria, Victoria, British Columbia, Canada
- Institute on Aging and Lifelong Health, University of Victoria, Victoria, British Columbia, Canada
| | - Jodie R. Gawryluk
- Department of Psychology, University of Victoria, Victoria, British Columbia, Canada
- Institute on Aging and Lifelong Health, University of Victoria, Victoria, British Columbia, Canada
- Division of Medical Sciences, University of Victoria, Victoria, British Columbia, Canada
| | | |
Collapse
|
32
|
Cai Y, Shi D, Lan G, Chen L, Jiang Y, Zhou L, Guo T. Association of β-Amyloid, Microglial Activation, Cortical Thickness, and Metabolism in Older Adults Without Dementia. Neurology 2024; 102:e209205. [PMID: 38489560 DOI: 10.1212/wnl.0000000000209205] [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: 11/06/2023] [Accepted: 12/13/2023] [Indexed: 03/17/2024] Open
Abstract
BACKGROUND AND OBJECTIVES Plasma β-amyloid42 (Aβ42)/Aβ40 levels have shown promise in identifying Aβ-PET positive individuals. This study explored the concordance and discordance of plasma Aβ42/Aβ40 positivity (Plasma±) with CSF Aβ42/Aβ40 positivity (CSF±) and Aβ-PET positivity (PET±) in older adults without dementia. Associations of Aβ deposition, cortical thickness, glucose metabolism, and microglial activation were also investigated. METHODS We selected participants without dementia who had concurrent plasma Aβ42/Aβ40 and Aβ-PET scans from the Alzheimer's Disease Neuroimaging Initiative cohort. Participants were categorized into Plasma±/PET± based on thresholds of composite 18F-florbetapir (FBP) standardized uptake value ratio (SUVR) ≥1.11 and plasma Aβ42/Aβ40 ≤0.1218. Aβ-PET-negative individuals were further divided into Plasma±/CSF± (CSF Aβ42/Aβ40 ≤0.138), and the concordance and discordance of Aβ42/Aβ40 in the plasma and CSF were investigated. Baseline and slopes of regional FBP SUVR were compared among Plasma±/PET± groups, and associations of regional FBP SUVR, FDG SUVR, cortical thickness, and CSF soluble Triggering Receptor Expressed on Myeloid Cell 2 (sTREM2) levels were analyzed. RESULTS One hundred eighty participants (mean age 72.7 years, 51.4% female, 96 cognitively unimpaired, and 84 with mild cognitive impairment) were included. We found that the proportion of Plasma+/PET- individuals was 6.14 times higher (odds ratio (OR) = 6.143, 95% confidence interval (CI) 2.740-16.185, p < 0.001) than that of Plasma-/PET+ individuals, and Plasma+/CSF- individuals showed 8.5 times larger percentage (OR = 8.5, 95% CI: 3.031-32.974, p < 0.001) than Plasma-/CSF+ individuals in Aβ-PET-negative individuals. Besides, Plasma+/PET- individuals exhibited faster (p < 0.05) Aβ accumulation predominantly in bilateral banks of superior temporal sulcus (BANKSSTS) and supramarginal, and superior parietal cortices compared with Plasma-/PET- individuals, despite no difference in baseline FBP SUVRs. In Plasma+/PET+ individuals, higher CSF sTREM2 levels correlated with slower BANKSSTS Aβ accumulation (standardized β (βstd) = -0.418, 95% CI -0.681 to -0.154, p = 0.002). Conversely, thicker cortical thickness and higher glucose metabolism in supramarginal and superior parietal cortices were associated with faster (p < 0.05) CSF sTREM2 increase in Plasma+/PET- individuals rather than in Plasma+/PET+ individuals. DISCUSSION These findings suggest that plasma Aβ42/Aβ40 abnormalities may predate CSF Aβ42/Aβ40 and Aβ-PET abnormalities. Higher sTREM2-related microglial activation is linked to thicker cortical thickness and higher metabolism in early amyloidosis stages but tends to mitigate Aβ accumulation primarily at relatively advanced stages.
Collapse
Affiliation(s)
- Yue Cai
- From the Institute of Biomedical Engineering (Y.C., G.L., L.C., T.G.), Shenzhen Bay Laboratory; Neurology Medicine Center (D.S., L.Z.), The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, China; Department of Psychology (Y.J.), University of Texas at Austin; and Institute of Biomedical Engineering (T.G.), Peking University Shenzhen Graduate School, China
| | - Dai Shi
- From the Institute of Biomedical Engineering (Y.C., G.L., L.C., T.G.), Shenzhen Bay Laboratory; Neurology Medicine Center (D.S., L.Z.), The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, China; Department of Psychology (Y.J.), University of Texas at Austin; and Institute of Biomedical Engineering (T.G.), Peking University Shenzhen Graduate School, China
| | - Guoyu Lan
- From the Institute of Biomedical Engineering (Y.C., G.L., L.C., T.G.), Shenzhen Bay Laboratory; Neurology Medicine Center (D.S., L.Z.), The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, China; Department of Psychology (Y.J.), University of Texas at Austin; and Institute of Biomedical Engineering (T.G.), Peking University Shenzhen Graduate School, China
| | - Linting Chen
- From the Institute of Biomedical Engineering (Y.C., G.L., L.C., T.G.), Shenzhen Bay Laboratory; Neurology Medicine Center (D.S., L.Z.), The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, China; Department of Psychology (Y.J.), University of Texas at Austin; and Institute of Biomedical Engineering (T.G.), Peking University Shenzhen Graduate School, China
| | - Yanni Jiang
- From the Institute of Biomedical Engineering (Y.C., G.L., L.C., T.G.), Shenzhen Bay Laboratory; Neurology Medicine Center (D.S., L.Z.), The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, China; Department of Psychology (Y.J.), University of Texas at Austin; and Institute of Biomedical Engineering (T.G.), Peking University Shenzhen Graduate School, China
| | - Liemin Zhou
- From the Institute of Biomedical Engineering (Y.C., G.L., L.C., T.G.), Shenzhen Bay Laboratory; Neurology Medicine Center (D.S., L.Z.), The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, China; Department of Psychology (Y.J.), University of Texas at Austin; and Institute of Biomedical Engineering (T.G.), Peking University Shenzhen Graduate School, China
| | - Tengfei Guo
- From the Institute of Biomedical Engineering (Y.C., G.L., L.C., T.G.), Shenzhen Bay Laboratory; Neurology Medicine Center (D.S., L.Z.), The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, China; Department of Psychology (Y.J.), University of Texas at Austin; and Institute of Biomedical Engineering (T.G.), Peking University Shenzhen Graduate School, China
| |
Collapse
|
33
|
Sun W, Jon K, Zhu W. Multiple phenotype association tests based on sliced inverse regression. BMC Bioinformatics 2024; 25:144. [PMID: 38575890 PMCID: PMC10996256 DOI: 10.1186/s12859-024-05731-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Accepted: 03/05/2024] [Indexed: 04/06/2024] Open
Abstract
BACKGROUND Joint analysis of multiple phenotypes in studies of biological systems such as Genome-Wide Association Studies is critical to revealing the functional interactions between various traits and genetic variants, but growth of data in dimensionality has become a very challenging problem in the widespread use of joint analysis. To handle the excessiveness of variables, we consider the sliced inverse regression (SIR) method. Specifically, we propose a novel SIR-based association test that is robust and powerful in testing the association between multiple predictors and multiple outcomes. RESULTS We conduct simulation studies in both low- and high-dimensional settings with various numbers of Single-Nucleotide Polymorphisms and consider the correlation structure of traits. Simulation results show that the proposed method outperforms the existing methods. We also successfully apply our method to the genetic association study of ADNI dataset. Both the simulation studies and real data analysis show that the SIR-based association test is valid and achieves a higher efficiency compared with its competitors. CONCLUSION Several scenarios with low- and high-dimensional responses and genotypes are considered in this paper. Our SIR-based method controls the estimated type I error at the pre-specified level α .
Collapse
Affiliation(s)
- Wenyuan Sun
- Key Laboratory for Applied Statistics of MOE, School of Mathematics and Statistics, Northeast Normal University, Changchun, 130024, Jilin, China
- Department of Mathematics, College of Science, Yanbian University, Yanji, 133002, Jilin, China
| | - Kyongson Jon
- Key Laboratory for Applied Statistics of MOE, School of Mathematics and Statistics, Northeast Normal University, Changchun, 130024, Jilin, China
- Faculty of Mathematics, Kim Il Sung University, Pyongyan , 999093, Democratic People's Republic of Korea
| | - Wensheng Zhu
- Key Laboratory for Applied Statistics of MOE, School of Mathematics and Statistics, Northeast Normal University, Changchun, 130024, Jilin, China.
- School of Mathematical Sciences, Harbin Normal University, Harbin, 150025, Heilongjiang, China.
| |
Collapse
|
34
|
Punzi M, Sestieri C, Picerni E, Chiarelli AM, Padulo C, Delli Pizzi A, Tullo MG, Tosoni A, Granzotto A, Della Penna S, Onofrj M, Ferretti A, Delli Pizzi S, Sensi SL, for the Alzheimer's Disease Neuroimaging Initiative. Atrophy of hippocampal subfields and amygdala nuclei in subjects with mild cognitive impairment progressing to Alzheimer's disease. Heliyon 2024; 10:e27429. [PMID: 38509925 PMCID: PMC10951508 DOI: 10.1016/j.heliyon.2024.e27429] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2023] [Revised: 02/27/2024] [Accepted: 02/28/2024] [Indexed: 03/22/2024] Open
Abstract
The hippocampus and amygdala are the first brain regions to show early signs of Alzheimer's Disease (AD) pathology. AD is preceded by a prodromal stage known as Mild Cognitive Impairment (MCI), a crucial crossroad in the clinical progression of the disease. The topographical development of AD has been the subject of extended investigation. However, it is still largely unknown how the transition from MCI to AD affects specific hippocampal and amygdala subregions. The present study is set to answer that question. We analyzed data from 223 subjects: 75 healthy controls, 52 individuals with MCI, and 96 AD patients obtained from the ADNI. The MCI group was further divided into two subgroups depending on whether individuals in the 48 months following the diagnosis either remained stable (N = 21) or progressed to AD (N = 31). A MANCOVA test evaluated group differences in the volume of distinct amygdala and hippocampal subregions obtained from magnetic resonance images. Subsequently, a stepwise linear discriminant analysis (LDA) determined which combination of magnetic resonance imaging parameters was most effective in predicting the conversion from MCI to AD. The predictive performance was assessed through a Receiver Operating Characteristic analysis. AD patients displayed widespread subregional atrophy. MCI individuals who progressed to AD showed selective atrophy of the hippocampal subiculum and tail compared to stable MCI individuals, who were undistinguishable from healthy controls. Converter MCI showed atrophy of the amygdala's accessory basal, central, and cortical nuclei. The LDA identified the hippocampal subiculum and the amygdala's lateral and accessory basal nuclei as significant predictors of MCI conversion to AD. The analysis returned a sensitivity value of 0.78 and a specificity value of 0.62. These findings highlight the importance of targeted assessments of distinct amygdala and hippocampus subregions to help dissect the clinical and pathophysiological development of the MCI to AD transition.
Collapse
Affiliation(s)
- Miriam Punzi
- Department of Neuroscience, Imaging, and Clinical Sciences, University “G. D'Annunzio of Chieti-Pescara”, Chieti, 66100, Italy
- Molecular Neurology Unit, Center for Advanced Studies and Technology (CAST), University “G. D'Annunzio of Chieti-Pescara”, Chieti, 66100, Italy
| | - Carlo Sestieri
- Department of Neuroscience, Imaging, and Clinical Sciences, University “G. D'Annunzio of Chieti-Pescara”, Chieti, 66100, Italy
- Institute for Advanced Biomedical Technologies (ITAB), “G. D'Annunzio of Chieti-Pescara”, Chieti, 66100, Italy
| | - Eleonora Picerni
- Department of Neuroscience, Imaging, and Clinical Sciences, University “G. D'Annunzio of Chieti-Pescara”, Chieti, 66100, Italy
| | - Antonio Maria Chiarelli
- Department of Neuroscience, Imaging, and Clinical Sciences, University “G. D'Annunzio of Chieti-Pescara”, Chieti, 66100, Italy
| | - Caterina Padulo
- Department of Neuroscience, Imaging, and Clinical Sciences, University “G. D'Annunzio of Chieti-Pescara”, Chieti, 66100, Italy
- Department of Humanities, University of Naples Federico II, Naples, 80133, Italy
| | - Andrea Delli Pizzi
- Department of Neuroscience, Imaging, and Clinical Sciences, University “G. D'Annunzio of Chieti-Pescara”, Chieti, 66100, Italy
| | - Maria Giulia Tullo
- Department of Neuroscience, Imaging, and Clinical Sciences, University “G. D'Annunzio of Chieti-Pescara”, Chieti, 66100, Italy
| | - Annalisa Tosoni
- Department of Neuroscience, Imaging, and Clinical Sciences, University “G. D'Annunzio of Chieti-Pescara”, Chieti, 66100, Italy
| | - Alberto Granzotto
- Department of Neuroscience, Imaging, and Clinical Sciences, University “G. D'Annunzio of Chieti-Pescara”, Chieti, 66100, Italy
- Molecular Neurology Unit, Center for Advanced Studies and Technology (CAST), University “G. D'Annunzio of Chieti-Pescara”, Chieti, 66100, Italy
| | - Stefania Della Penna
- Department of Neuroscience, Imaging, and Clinical Sciences, University “G. D'Annunzio of Chieti-Pescara”, Chieti, 66100, Italy
- Institute for Advanced Biomedical Technologies (ITAB), “G. D'Annunzio of Chieti-Pescara”, Chieti, 66100, Italy
| | - Marco Onofrj
- Department of Neuroscience, Imaging, and Clinical Sciences, University “G. D'Annunzio of Chieti-Pescara”, Chieti, 66100, Italy
| | - Antonio Ferretti
- Department of Neuroscience, Imaging, and Clinical Sciences, University “G. D'Annunzio of Chieti-Pescara”, Chieti, 66100, Italy
- Institute for Advanced Biomedical Technologies (ITAB), “G. D'Annunzio of Chieti-Pescara”, Chieti, 66100, Italy
- UdA-TechLab, Research Center, University “G. D’Annunzio” of Chieti-Pescara, 66100, Chieti, Italy
| | - Stefano Delli Pizzi
- Department of Neuroscience, Imaging, and Clinical Sciences, University “G. D'Annunzio of Chieti-Pescara”, Chieti, 66100, Italy
- Institute for Advanced Biomedical Technologies (ITAB), “G. D'Annunzio of Chieti-Pescara”, Chieti, 66100, Italy
- Molecular Neurology Unit, Center for Advanced Studies and Technology (CAST), University “G. D'Annunzio of Chieti-Pescara”, Chieti, 66100, Italy
| | - Stefano L. Sensi
- Department of Neuroscience, Imaging, and Clinical Sciences, University “G. D'Annunzio of Chieti-Pescara”, Chieti, 66100, Italy
- Institute for Advanced Biomedical Technologies (ITAB), “G. D'Annunzio of Chieti-Pescara”, Chieti, 66100, Italy
- Molecular Neurology Unit, Center for Advanced Studies and Technology (CAST), University “G. D'Annunzio of Chieti-Pescara”, Chieti, 66100, Italy
| | | |
Collapse
|
35
|
Das Adhikari S, Cui Y, Wang J. BayesKAT: bayesian optimal kernel-based test for genetic association studies reveals joint genetic effects in complex diseases. Brief Bioinform 2024; 25:bbae182. [PMID: 38653490 PMCID: PMC11036342 DOI: 10.1093/bib/bbae182] [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: 11/22/2023] [Revised: 03/10/2024] [Accepted: 04/05/2024] [Indexed: 04/25/2024] Open
Abstract
Genome-wide Association Studies (GWAS) methods have identified individual single-nucleotide polymorphisms (SNPs) significantly associated with specific phenotypes. Nonetheless, many complex diseases are polygenic and are controlled by multiple genetic variants that are usually non-linearly dependent. These genetic variants are marginally less effective and remain undetected in GWAS analysis. Kernel-based tests (KBT), which evaluate the joint effect of a group of genetic variants, are therefore critical for complex disease analysis. However, choosing different kernel functions in KBT can significantly influence the type I error control and power, and selecting the optimal kernel remains a statistically challenging task. A few existing methods suffer from inflated type 1 errors, limited scalability, inferior power or issues of ambiguous conclusions. Here, we present a new Bayesian framework, BayesKAT (https://github.com/wangjr03/BayesKAT), which overcomes these kernel specification issues by selecting the optimal composite kernel adaptively from the data while testing genetic associations simultaneously. Furthermore, BayesKAT implements a scalable computational strategy to boost its applicability, especially for high-dimensional cases where other methods become less effective. Based on a series of performance comparisons using both simulated and real large-scale genetics data, BayesKAT outperforms the available methods in detecting complex group-level associations and controlling type I errors simultaneously. Applied on a variety of groups of functionally related genetic variants based on biological pathways, co-expression gene modules and protein complexes, BayesKAT deciphers the complex genetic basis and provides mechanistic insights into human diseases.
Collapse
Affiliation(s)
- Sikta Das Adhikari
- Department of Statistics and Probability, Michigan State University, East Lansing, MI 48824, USA
- Department of Computational Mathematics, Science and Engineering, Michigan State University, East Lansing, MI 48824, USA
| | - Yuehua Cui
- Department of Statistics and Probability, Michigan State University, East Lansing, MI 48824, USA
| | - Jianrong Wang
- Department of Computational Mathematics, Science and Engineering, Michigan State University, East Lansing, MI 48824, USA
| |
Collapse
|
36
|
Kim RT, Zhou L, Li Y, Krieger AC, Nordvig AS, Butler T, de Leon MJ, Chiang GC. Impaired sleep is associated with tau deposition on 18F-flortaucipir PET and accelerated cognitive decline, accounting for medications that affect sleep. J Neurol Sci 2024; 458:122927. [PMID: 38341949 PMCID: PMC10947806 DOI: 10.1016/j.jns.2024.122927] [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: 10/23/2023] [Revised: 01/06/2024] [Accepted: 02/06/2024] [Indexed: 02/13/2024]
Abstract
BACKGROUND Impaired sleep is commonly associated with Alzheimer's disease (AD), although the underlying mechanisms remain unclear. Furthermore, the moderating effects of sleep-affecting medications, which have been linked to AD pathology, are incompletely characterized. Using data from the Alzheimer's Disease Neuroimaging Initiative, we investigated whether a medical history of impaired sleep, informant-reported nighttime behaviors, and sleep-affecting medications are associated with beta-amyloid and tau deposition on PET and cognitive change, cross-sectionally and longitudinally. METHODS We included 964 subjects with 18F-florbetapir PET scans. Measures of sleep impairment and medication use were obtained from medical histories and the Neuropsychiatric Inventory Questionnaire. Multivariate models, adjusted for covariates, were used to assess associations among sleep-related features, beta-amyloid and tau, and cognition. Cortical tau deposition, categorized by Braak stage, was assessed using the standardized uptake value peak alignment (SUVP) method on 18F-flortaucipir PET. RESULTS Medical history of sleep impairment was associated with greater baseline tau in the meta-temporal, Braak 1, and Braak 4 regions (p = 0.04, p < 0.001, p = 0.025, respectively). Abnormal nighttime behaviors were also associated with greater baseline tau in the meta-temporal region (p = 0.024), and greater cognitive impairment, cross-sectionally (p = 0.007) and longitudinally (p < 0.001). Impaired sleep was not associated with baseline beta-amyloid (p > 0.05). Short-term use of selective serotonin reuptake inhibitors and benzodiazepines slightly weakened the sleep-tau relationship. CONCLUSIONS Sleep impairment was associated with tauopathy and cognitive decline, which could be linked to increased tau secretion from neuronal hyperactivity. Clinically, our results help identify high-risk individuals who could benefit from sleep-related interventions aimed to delay cognitive decline and AD.
Collapse
Affiliation(s)
- Ryan T Kim
- From the Department of Stem Cell and Regenerative Biology, Harvard University, Bauer-Sherman Fairchild Complex 7 Divinity Avenue, Cambridge, MA 02138, United States of America.
| | - Liangdong Zhou
- From the Department of Radiology, Brain Health Imaging Institute, Weill Cornell Medicine, NewYork-Presbyterian Hospital, 407 E 61(st) Street, New York, NY 10065, United States of America.
| | - Yi Li
- From the Department of Radiology, Brain Health Imaging Institute, Weill Cornell Medicine, NewYork-Presbyterian Hospital, 407 E 61(st) Street, New York, NY 10065, United States of America.
| | - Ana C Krieger
- From the Departments of Medicine and Neurology, Division of Sleep Neurology, Weill Cornell Medicine, NewYork-Presbyterian Hospital, 425 E 61st St., 5th Floor, New York, NY 10065, United States of America.
| | - Anna S Nordvig
- From the Department of Neurology, Alzheimer's Disease and Memory Disorders Program, Weill Cornell Medicine, NewYork-Presbyterian Hospital, 428 East 72(nd) Street Suite 500, New York, NY 10021, United States of America.
| | - Tracy Butler
- From the Department of Radiology, Brain Health Imaging Institute, Weill Cornell Medicine, NewYork-Presbyterian Hospital, 407 E 61(st) Street, New York, NY 10065, United States of America.
| | - Mony J de Leon
- From the Department of Radiology, Brain Health Imaging Institute, Weill Cornell Medicine, NewYork-Presbyterian Hospital, 407 E 61(st) Street, New York, NY 10065, United States of America.
| | - Gloria C Chiang
- From the Department of Radiology, Brain Health Imaging Institute, Weill Cornell Medicine, NewYork-Presbyterian Hospital, 407 E 61(st) Street, New York, NY 10065, United States of America; From the Department of Radiology, Division of Neuroradiology, Weill Cornell Medicine, NewYork-Presbyterian Hospital, 525 East 68th Street, Starr Pavilion, Box 141, New York, NY 10065, United States of America.
| |
Collapse
|
37
|
Levin F, Grothe MJ, Dyrba M, Franzmeier N, Teipel SJ. Longitudinal trajectories of cognitive reserve in hypometabolic subtypes of Alzheimer's disease. Neurobiol Aging 2024; 135:26-38. [PMID: 38157587 DOI: 10.1016/j.neurobiolaging.2023.12.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Revised: 11/16/2023] [Accepted: 12/13/2023] [Indexed: 01/03/2024]
Abstract
Previous studies have demonstrated resilience to AD-related neuropathology in a form of cognitive reserve (CR). In this study we investigated a relationship between CR and hypometabolic subtypes of AD, specifically the typical and the limbic-predominant subtypes. We analyzed data from 59 Aβ-positive cognitively normal (CN), 221 prodromal Alzheimer's disease (AD) and 174 AD dementia participants from the Alzheimer's Disease Neuroimaging Initiative (ADNI) from ADNI and ADNIGO/2 phases. For replication, we analyzed data from 5 Aβ-positive CN, 89 prodromal AD and 43 AD dementia participants from ADNI3. CR was estimated as standardized residuals in a model predicting cognition from temporoparietal grey matter volumes and covariates. Higher CR estimates predicted slower cognitive decline. Typical and limbic-predominant hypometabolic subtypes demonstrated similar baseline CR, but the results suggested a faster decline of CR in the typical subtype. These findings support the relationship between subtypes and CR, specifically longitudinal trajectories of CR. Results also underline the importance of longitudinal analyses in research on CR.
Collapse
Affiliation(s)
- Fedor Levin
- Deutsches Zentrum für Neurodegenerative Erkrankungen (DZNE), Rostock, Germany.
| | - Michel J Grothe
- Unidad de Trastornos del Movimiento, Servicio de Neurología y Neurofisiología Clínica, Instituto de Biomedicina de Sevilla, Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Seville, Spain
| | - Martin Dyrba
- Deutsches Zentrum für Neurodegenerative Erkrankungen (DZNE), Rostock, Germany
| | - Nicolai Franzmeier
- Institute for Stroke and Dementia Research, Klinikum der Universität München, Ludwig-Maximilians-Universität LMU, Munich, Germany; Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Sweden; Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Stefan J Teipel
- Deutsches Zentrum für Neurodegenerative Erkrankungen (DZNE), Rostock, Germany; Department of Psychosomatic Medicine, Rostock University Medical Center, Rostock, Germany
| |
Collapse
|
38
|
Kikuchi M, Miyashita A, Hara N, Kasuga K, Saito Y, Murayama S, Kakita A, Akatsu H, Ozaki K, Niida S, Kuwano R, Iwatsubo T, Nakaya A, Ikeuchi T. Polygenic effects on the risk of Alzheimer's disease in the Japanese population. Alzheimers Res Ther 2024; 16:45. [PMID: 38414085 PMCID: PMC10898021 DOI: 10.1186/s13195-024-01414-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2023] [Accepted: 02/11/2024] [Indexed: 02/29/2024]
Abstract
BACKGROUND Polygenic effects have been proposed to account for some disease phenotypes; these effects are calculated as a polygenic risk score (PRS). This score is correlated with Alzheimer's disease (AD)-related phenotypes, such as biomarker abnormalities and brain atrophy, and is associated with conversion from mild cognitive impairment (MCI) to AD. However, the AD PRS has been examined mainly in Europeans, and owing to differences in genetic structure and lifestyle, it is unclear whether the same relationships between the PRS and AD-related phenotypes exist in non-European populations. In this study, we calculated and evaluated the AD PRS in Japanese individuals using genome-wide association study (GWAS) statistics from Europeans. METHODS In this study, we calculated the AD PRS in 504 Japanese participants (145 cognitively unimpaired (CU) participants, 220 participants with late mild cognitive impairment (MCI), and 139 patients with mild AD dementia) enrolled in the Japanese Alzheimer's Disease Neuroimaging Initiative (J-ADNI) project. In order to evaluate the clinical value of this score, we (1) determined the polygenic effects on AD in the J-ADNI and validated it using two independent cohorts (a Japanese neuropathology (NP) cohort (n = 565) and the North American ADNI (NA-ADNI) cohort (n = 617)), (2) examined the AD-related phenotypes associated with the PRS, and (3) tested whether the PRS helps predict the conversion of MCI to AD. RESULTS The PRS using 131 SNPs had an effect independent of APOE. The PRS differentiated between CU participants and AD patients with an area under the curve (AUC) of 0.755 when combined with the APOE variants. Similar AUC was obtained when PRS calculated by the NP and NA-ADNI cohorts was applied. In MCI patients, the PRS was associated with cerebrospinal fluid phosphorylated-tau levels (β estimate = 0.235, p value = 0.026). MCI with a high PRS showed a significantly increased conversion to AD in APOE ε4 noncarriers with a hazard rate of 2.22. In addition, we also developed a PRS model adjusted for LD and observed similar results. CONCLUSIONS We showed that the AD PRS is useful in the Japanese population, whose genetic structure is different from that of the European population. These findings suggest that the polygenicity of AD is partially common across ethnic differences.
Collapse
Affiliation(s)
- Masataka Kikuchi
- Department of Computational Biology and Medical Sciences, Graduate School of Frontier Science, The University of Tokyo, 6-2-3 Kashiwanoha, Kashiwa, Chiba, 277-0882, Japan.
- Department of Medical Informatics, Graduate School of Medicine, Osaka University, Osaka, Japan.
| | - Akinori Miyashita
- Department of Molecular Genetics, Brain Research Institute, Niigata University, 1-757 Asahimachi, Niigata, 951-8585, Japan
| | - Norikazu Hara
- Department of Molecular Genetics, Brain Research Institute, Niigata University, 1-757 Asahimachi, Niigata, 951-8585, Japan
| | - Kensaku Kasuga
- Department of Molecular Genetics, Brain Research Institute, Niigata University, 1-757 Asahimachi, Niigata, 951-8585, Japan
| | - Yuko Saito
- Brain Bank for Aging Research (Department of Neuropathology), Tokyo Metropolitan Institute of Geriatrics and Gerontology, Tokyo, Japan
| | - Shigeo Murayama
- Brain Bank for Aging Research (Department of Neuropathology), Tokyo Metropolitan Institute of Geriatrics and Gerontology, Tokyo, Japan
- Brain Bank for Neurodevelopmental, Neurological and Psychiatric Disorders, United Graduate School of Child Development, Osaka University, Osaka, Japan
| | - Akiyoshi Kakita
- Department of Pathology, Brain Research Institute, Niigata University, Niigata, Japan
| | - Hiroyasu Akatsu
- Department of General Medicine & General Internal Medicine, Nagoya City University Graduate School of Medicine, Nagoya, Japan
| | - Kouichi Ozaki
- Medical Genome Center, National Center for Geriatrics and Gerontology, Research Institute, Aichi, Japan
- RIKEN Center for Integrative Medical Sciences, Kanagawa, Japan
| | - Shumpei Niida
- Core Facility Administration, National Center for Geriatrics and Gerontology, Research Institute, Aichi, Japan
| | - Ryozo Kuwano
- Social Welfare Corporation Asahigawaso, Asahigawaso Research Institute, Okayama, Japan
| | - Takeshi Iwatsubo
- Department of Neuropathology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Akihiro Nakaya
- Department of Computational Biology and Medical Sciences, Graduate School of Frontier Science, The University of Tokyo, 6-2-3 Kashiwanoha, Kashiwa, Chiba, 277-0882, Japan
| | - Takeshi Ikeuchi
- Department of Molecular Genetics, Brain Research Institute, Niigata University, 1-757 Asahimachi, Niigata, 951-8585, Japan.
| |
Collapse
|
39
|
Guo F, Tan MS, Hu H, Ou YN, Zhang MZ, Sheng ZH, Chi HC, Tan L. sTREM2 Mediates the Correlation Between BIN1 Gene Polymorphism and Tau Pathology in Alzheimer's Disease. J Alzheimers Dis 2024; 101:693-704. [PMID: 39240638 DOI: 10.3233/jad-240372] [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: 09/07/2024]
Abstract
Background Bridging integrator 1 (BIN1) gene polymorphism has been reported to play a role in the pathological processes of Alzheimer's disease (AD). Objective To explore the association of BIN1 loci with neuroinflammation and AD pathology. Methods Alzheimer's Disease Neuroimaging Initiative (ADNI, N = 495) was the discovery cohort, and Chinese Alzheimer's Biomarker and LifestylE (CABLE, N = 619) study was used to replicate the results. Two BIN1 gene polymorphism (rs7561528 and rs744373) were included in the analysis. Multiple linear regression model and causal mediation analysis conducted through 10,000 bootstrapped iterations were used to examine the BIN1 loci relationship with cerebrospinal fluid (CSF) AD biomarkers and alternative biomarker of microglial activation microglia-soluble triggering receptor expressed on myeloid cells 2 (sTREM2). Results In ADNI database, we found a significant association between BIN1 loci (rs7561528 and rs744373) and levels of CSF phosphorylated-tau (P-tau) (pc = 0.017; 0.010, respectively) and total-tau (T-tau) (pc = 0.011; 0.013, respectively). The BIN1 loci were also correlated with CSF sTREM2 levels (pc = 0.010; 0.008, respectively). Mediation analysis demonstrated that CSF sTREM2 partially mediated the association of BIN1 loci with P-tau (Proportion of rs7561528 : 20.8%; Proportion of rs744373 : 24.8%) and T-tau (Proportion of rs7561528 : 36.5%; Proportion of rs744373 : 43.9%). The analysis in CABLE study replicated the mediation role of rs7561528. Conclusions This study demonstrated the correlation between BIN1 loci and CSF AD biomarkers as well as microglia biomarkers. Additionally, the link between BIN1 loci and tau pathology was partially mediated by CSF sTREM2.
Collapse
Affiliation(s)
- Fan Guo
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, Qingdao, China
| | - Meng-Shan Tan
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, Qingdao, China
- Department of Neurology, Qingdao Hospital, University of Health and Rehabilitation Sciences, Qingdao, China
- Department of Neurology, School of Clinical Medicine, Shandong Second Medical University, Weifang, China
| | - Hao Hu
- Department of Neurology, Qingdao Hospital, University of Health and Rehabilitation Sciences, Qingdao, China
| | - Ya-Nan Ou
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, Qingdao, China
| | - Ming-Zhan Zhang
- Department of Neurology, School of Clinical Medicine, Shandong Second Medical University, Weifang, China
| | - Ze-Hu Sheng
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, Qingdao, China
| | - Hao-Chen Chi
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, Qingdao, China
| | - Lan Tan
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, Qingdao, China
- Department of Neurology, Qingdao Hospital, University of Health and Rehabilitation Sciences, Qingdao, China
- Department of Neurology, School of Clinical Medicine, Shandong Second Medical University, Weifang, China
| |
Collapse
|
40
|
Rehman H, Ang TFA, Tao Q, Espenilla AL, Au R, Farrer LA, Zhang X, Qiu WQ. Comparison of Commonly Measured Plasma and Cerebrospinal Fluid Proteins and Their Significance for the Characterization of Cognitive Impairment Status. J Alzheimers Dis 2024; 97:621-633. [PMID: 38143358 DOI: 10.3233/jad-230837] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2023]
Abstract
BACKGROUND Although cerebrospinal fluid (CSF) amyloid-β42 peptide (Aβ42) and phosphorylated tau (p-tau) and blood p-tau are valuable for differential diagnosis of Alzheimer's disease (AD) from cognitively normal (CN) there is a lack of validated biomarkers for mild cognitive impairment (MCI). OBJECTIVE This study sought to determine how plasma and CSF protein markers compared in the characterization of MCI and AD status. METHODS This cohort study included Alzheimer's Disease Neuroimaging Initiative (ADNI) participants who had baseline levels of 75 proteins measured commonly in plasma and CSF (257 total, 46 CN, 143 MCI, and 68 AD). Logistic regression, least absolute shrinkage and selection operator (LASSO) and Random Forest (RF) methods were used to identify the protein candidates for the disease classification. RESULTS We observed that six plasma proteins panel (APOE, AMBP, C3, IL16, IGFBP2, APOD) outperformed the seven CSF proteins panel (VEGFA, HGF, PRL, FABP3, FGF4, CD40, RETN) as well as AD markers (CSF p-tau and Aβ42) to distinguish the MCI from AD [area under the curve (AUC) = 0.75 (plasma proteins), AUC = 0.60 (CSF proteins) and AUC = 0.56 (CSF p-tau and Aβ42)]. Also, these six plasma proteins performed better than the CSF proteins and were in line with CSF p-tau and Aβ42 in differentiating CN versus MCI subjects [AUC = 0.89 (plasma proteins), AUC = 0.85 (CSF proteins) and AUC = 0.89 (CSF p-tau and Aβ42)]. These results were adjusted for age, sex, education, and APOEϵ4 genotype. CONCLUSIONS This study suggests that the combination of 6 plasma proteins can serve as an effective marker for differentiating MCI from AD and CN.
Collapse
Affiliation(s)
- Habbiburr Rehman
- Department of Medicine (Biomedical Genetics), Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
| | - Ting Fang Alvin Ang
- Department of Anatomy & Neurobiology, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
- Department of Neurology, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
| | - Qiushan Tao
- Department of Pharmacology & Experimental Therapeutics, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
| | - Arielle Lauren Espenilla
- Department of Biostatistics and Boston University School of Public Health, Boston, MA, USA
- Department of Epidemiology, Boston University School of Public Health, Boston, MA, USA
| | - Rhoda Au
- Department of Medicine (Biomedical Genetics), Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
- Department of Anatomy & Neurobiology, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
- Department of Neurology, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
- Department of Epidemiology, Boston University School of Public Health, Boston, MA, USA
- Framingham Heart Study, Boston University School of Medicine, Framingham, MA, USA
- Alzheimer's Disease Research Center, Boston University School of Medicine, Boston, MA, USA
| | - Lindsay A Farrer
- Department of Medicine (Biomedical Genetics), Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
- Department of Neurology, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
- Department of Ophthalmology, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
- Department of Biostatistics and Boston University School of Public Health, Boston, MA, USA
- Department of Epidemiology, Boston University School of Public Health, Boston, MA, USA
- Framingham Heart Study, Boston University School of Medicine, Framingham, MA, USA
- Alzheimer's Disease Research Center, Boston University School of Medicine, Boston, MA, USA
| | - Xiaoling Zhang
- Department of Medicine (Biomedical Genetics), Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
- Department of Biostatistics and Boston University School of Public Health, Boston, MA, USA
- Department of Epidemiology, Boston University School of Public Health, Boston, MA, USA
- Framingham Heart Study, Boston University School of Medicine, Framingham, MA, USA
- Alzheimer's Disease Research Center, Boston University School of Medicine, Boston, MA, USA
| | - Wei Qiao Qiu
- Department of Anatomy & Neurobiology, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
- Department of Neurology, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
- Department of Pharmacology & Experimental Therapeutics, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
- Department of Psychiatry, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
- Alzheimer's Disease Research Center, Boston University School of Medicine, Boston, MA, USA
| |
Collapse
|
41
|
Albala B, Appelmans E, Burress R, De Santi S, Devins T, Klein G, Logovinsky V, Novak GP, Ribeiro K, Schmidt ME, Schwarz AJ, Scott D, Shcherbinin S, Siemers E, Travaglia A, Weber CJ, White L, Wolf‐Rodda J, Vasanthakumar A. The Alzheimer's Disease Neuroimaging Initiative and the role and contributions of the Private Partners Scientific Board (PPSB). Alzheimers Dement 2024; 20:695-708. [PMID: 37774088 PMCID: PMC10843521 DOI: 10.1002/alz.13483] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2023] [Revised: 08/24/2023] [Accepted: 08/27/2023] [Indexed: 10/01/2023]
Abstract
The Alzheimer's Disease Neuroimaging Initiative (ADNI) Private Partners Scientific Board (PPSB) encompasses members from industry, biotechnology, diagnostic, and non-profit organizations that have until recently been managed by the Foundation for the National Institutes of Health (FNIH) and provided financial and scientific support to ADNI programs. In this article, we review some of the major activities undertaken by the PPSB, focusing on those supporting the most recently completed National Institute on Aging grant, ADNI3, and the impact it has had on streamlining biomarker discovery and validation in Alzheimer's disease. We also provide a perspective on the gaps that may be filled with future PPSB activities as part of ADNI4 and beyond. HIGHLIGHTS: The Private Partners Scientific board (PPSB) continues to play a key role in enabling several Alzheimer's Disease Neuroimaging Initiative (ADNI) activities. PPSB working groups have led landscape assessments to provide valuable feedback on new technologies, platforms, and methods that may be taken up by ADNI in current or future iterations.
Collapse
Affiliation(s)
- Bruce Albala
- Eisai Inc.NutleyNew JerseyUSA
- Present address:
Program in Public HealthIrvine and Department of NeurologyUCI School of MedicineUniversity of California856 Health Sciences QuadIrvineCalifornia92697‐3957USA
| | - Eline Appelmans
- Foundation for the National Institutes of HealthNorth BethesdaMarylandUSA
| | - Ramona Burress
- Janssen Research & Development, LLCTitusvilleNew JerseyUSA
- Present address:
Takeda95, Hayden AvenueLexingtonMassachusetts02421USA
| | - Susan De Santi
- Eisai Inc.NutleyNew JerseyUSA
- Life Molecular ImagingBerlinGermany
- Present address:
Eisai Inc.NutleyNew JerseyUSA
| | - Theresa Devins
- Eisai Inc.NutleyNew JerseyUSA
- Present address:
Cognition Therapeutics2500 Westchester AvenuePurchaseNew York10577USA
| | | | - Veronika Logovinsky
- Eisai Inc.NutleyNew JerseyUSA
- Present address:
Lundbeck6 Parkway NDeerfieldIllinois60015USA
| | | | | | | | | | | | | | | | - Alessio Travaglia
- Foundation for the National Institutes of HealthNorth BethesdaMarylandUSA
| | | | - Leah White
- Foundation for the National Institutes of HealthNorth BethesdaMarylandUSA
- Present address:
Veranex5420 Wade Park Blvd Suite 204RaleighNorth Carolina27607USA
| | - Julie Wolf‐Rodda
- Foundation for the National Institutes of HealthNorth BethesdaMarylandUSA
| | | |
Collapse
|
42
|
Luo X, Hong H, Li K, Zeng Q, Liu X, Hong L, Li J, Zhang X, Zhong S, Xu X, Chen Y, Zhang M, Huang P. Association Between Small Vessel Disease and Financial Capacity: A Study Based on Cognitively Normal Older Adults. J Alzheimers Dis 2024; 98:897-906. [PMID: 38461505 DOI: 10.3233/jad-231089] [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: 03/12/2024]
Abstract
Background Financial capacity is vital for the elderly, who possess a substantial share of global wealth but are vulnerable to financial fraud. Objective We explored the link between small vessel disease (SVD) and financial capacity in cognitively unimpaired (CU) older adults via both cross-sectional and longitudinal analyses. Methods 414 CU participants underwent MRI and completed the Financial Capacity Instrument-Short Form (FCI-SF). Subsequent longitudinal FCI-SF data were obtained from 104, 240, and 141 participants at one, two, and four years, respectively. SVD imaging markers, encompassing white matter hyperintensities (WMH), cerebral microbleeds (CMB), and lacune were evaluated. We used linear regression analyses to cross-sectionally explore the association between FCI-SF and SVD severity, and linear mixed models to assess how baseline SVD severity impacted longitudinal FCI-SF change. The false discovery rate method was used to adjust multiple comparisons. Results Cross-sectional analysis revealed a significant association between baseline WMH and Bank Statement (BANK, β=-0.194), as well as between lacune number and Financial Conceptual Knowledge (FC, β= -0.171). These associations were stronger in APOE ɛ4 carriers, with β= -0.282 for WMH and BANK, and β= -0.366 for lacune number and FC. Longitudinally, higher baseline SVD total score was associated with severe FCI-SF total score decrease (β= -0.335). Additionally, baseline WMH burden predicted future decreases in Single Checkbook/Register Task (SNG, β= -0.137) and FC (β= -0.052). Notably, the association between baseline WMH and SNG changes was amplified in APOE ɛ4 carriers (β= -0.187). Conclusions Severe SVD was associated with worse FCI-SF and could predict the decline of financial capacity in CU older adults.
Collapse
Affiliation(s)
- Xiao Luo
- Department of Radiology,The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Hui Hong
- Department of Radiology,The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
- Department of Clinical Neurosciences, Cambridge Biomedical Campus, University of Cambridge, Addenbrooke's Hospital, Cambridge, UK
| | - Kaicheng Li
- Department of Radiology,The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Qingze Zeng
- Department of Radiology,The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Xiaocao Liu
- Department of Radiology,The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Luwei Hong
- Department of Radiology,The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Jixuan Li
- Department of Radiology,The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Xinyi Zhang
- Department of Neurology, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Siyan Zhong
- Department of Neurology, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Xiaopei Xu
- Department of Radiology,The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Yanxing Chen
- Department of Neurology, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Minming Zhang
- Department of Radiology,The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Peiyu Huang
- Department of Radiology,The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| |
Collapse
|
43
|
Rosewood TJ, Nho K, Risacher SL, Gao S, Shen L, Foroud T, Saykin AJ, on behalf of the Alzheimer’s Disease Neuroimaging Initiative. Genome-Wide Association Analysis across Endophenotypes in Alzheimer's Disease: Main Effects and Disease Stage-Specific Interactions. Genes (Basel) 2023; 14:2010. [PMID: 38002954 PMCID: PMC10671827 DOI: 10.3390/genes14112010] [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: 09/11/2023] [Revised: 10/23/2023] [Accepted: 10/23/2023] [Indexed: 11/26/2023] Open
Abstract
The underlying genetic susceptibility for Alzheimer's disease (AD) is not yet fully understood. The heterogeneous nature of the disease challenges genetic association studies. Endophenotype approaches can help to address this challenge by more direct interrogation of biological traits related to the disease. AD endophenotypes based on amyloid-β, tau, and neurodegeneration (A/T/N) biomarkers and cognitive performance were selected from the Alzheimer's Disease Neuroimaging Initiative (ADNI) cohort (N = 1565). A genome-wide association study (GWAS) of quantitative phenotypes was performed using an SNP main effect and an SNP by Diagnosis interaction (SNP × DX) model to identify disease stage-specific genetic effects. Nine loci were identified as study-wide significant with one or more A/T/N endophenotypes in the main effect model, as well as additional findings significantly associated with cognitive measures. These nine loci include SNPs in or near the genes APOE, SRSF10, HLA-DQB1, XKR3, and KIAA1671. The SNP × DX model identified three study-wide significant genetic loci (BACH2, EP300, and PACRG-AS1) with a neuroprotective effect in later AD stage endophenotypes. An endophenotype approach identified novel genetic associations and provided insight into the molecular mechanisms underlying the genetic associations that may otherwise be missed using conventional case-control study designs.
Collapse
Affiliation(s)
- Thea J. Rosewood
- Indiana Alzheimer’s Disease Research Center, Indianapolis, IN 46202, USA; (T.J.R.); (S.L.R.); (S.G.); (T.F.)
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN 46202, USA
- Department of Radiology and Imaging Sciences, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Kwangsik Nho
- Indiana Alzheimer’s Disease Research Center, Indianapolis, IN 46202, USA; (T.J.R.); (S.L.R.); (S.G.); (T.F.)
- Department of Radiology and Imaging Sciences, Indiana University School of Medicine, Indianapolis, IN 46202, USA
- School of Informatics and Computing, Indiana University, Indianapolis, IN 46202, USA
| | - Shannon L. Risacher
- Indiana Alzheimer’s Disease Research Center, Indianapolis, IN 46202, USA; (T.J.R.); (S.L.R.); (S.G.); (T.F.)
- Department of Radiology and Imaging Sciences, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Sujuan Gao
- Indiana Alzheimer’s Disease Research Center, Indianapolis, IN 46202, USA; (T.J.R.); (S.L.R.); (S.G.); (T.F.)
- Department of Biostatistics, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Li Shen
- Department of Biostatistics, Epidemiology and Informatics, The Perelman School of Medicine, Philadelphia, PA 19104, USA;
| | - Tatiana Foroud
- Indiana Alzheimer’s Disease Research Center, Indianapolis, IN 46202, USA; (T.J.R.); (S.L.R.); (S.G.); (T.F.)
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Andrew J. Saykin
- Indiana Alzheimer’s Disease Research Center, Indianapolis, IN 46202, USA; (T.J.R.); (S.L.R.); (S.G.); (T.F.)
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN 46202, USA
- Department of Radiology and Imaging Sciences, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | | |
Collapse
|
44
|
Das Adhikari S, Cui Y, Wang J. BayesKAT: Bayesian Optimal Kernel-based Test for genetic association studies reveals joint genetic effects in complex diseases. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.10.18.562824. [PMID: 37905124 PMCID: PMC10614916 DOI: 10.1101/2023.10.18.562824] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/02/2023]
Abstract
GWAS methods have identified individual SNPs significantly associated with specific phenotypes. Nonetheless, many complex diseases are polygenic and are controlled by multiple genetic variants that are usually non-linearly dependent. These genetic variants are marginally less effective and remain undetected in GWAS analysis. Kernel-based tests (KBT), which evaluate the joint effect of a group of genetic variants, are therefore critical for complex disease analysis. However, choosing different kernel functions in KBT can significantly influence the type I error control and power, and selecting the optimal kernel remains a statistically challenging task. A few existing methods suffer from inflated type 1 errors, limited scalability, inferior power, or issues of ambiguous conclusions. Here, we present a new Bayesian framework, BayesKAT( https://github.com/wangjr03/BayesKAT ), which overcomes these kernel specification issues by selecting the optimal composite kernel adaptively from the data while testing genetic associations simultaneously. Furthermore, BayesKAT implements a scalable computational strategy to boost its applicability, especially for high-dimensional cases where other methods become less effective. Based on a series of performance comparisons using both simulated and real large-scale genetics data, BayesKAT outperforms the available methods in detecting complex group-level associations and controlling type I errors simultaneously. Applied on a variety of groups of functionally related genetic variants based on biological pathways, co-expression gene modules, and protein complexes, BayesKAT deciphers the complex genetic basis and provides mechanistic insights into human diseases.
Collapse
|
45
|
Strain JF, Phuah CL, Adeyemo B, Cheng K, Womack KB, McCarthy J, Goyal M, Chen Y, Sotiras A, An H, Xiong C, Scharf A, Newsom-Stewart C, Morris JC, Benzinger TLS, Lee JM, Ances BM. White matter hyperintensity longitudinal morphometric analysis in association with Alzheimer disease. Alzheimers Dement 2023; 19:4488-4497. [PMID: 37563879 PMCID: PMC10592317 DOI: 10.1002/alz.13377] [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: 11/29/2022] [Revised: 05/30/2023] [Accepted: 06/02/2023] [Indexed: 08/12/2023]
Abstract
INTRODUCTION Vascular damage in Alzheimer's disease (AD) has shown conflicting findings particularly when analyzing longitudinal data. We introduce white matter hyperintensity (WMH) longitudinal morphometric analysis (WLMA) that quantifies WMH expansion as the distance from lesion voxels to a region of interest boundary. METHODS WMH segmentation maps were derived from 270 longitudinal fluid-attenuated inversion recovery (FLAIR) ADNI images. WLMA was performed on five data-driven WMH patterns with distinct spatial distributions. Amyloid accumulation was evaluated with WMH expansion across the five WMH patterns. RESULTS The preclinical group had significantly greater expansion in the posterior ventricular WM compared to controls. Amyloid significantly associated with frontal WMH expansion primarily within AD individuals. WLMA outperformed WMH volume changes for classifying AD from controls primarily in periventricular and posterior WMH. DISCUSSION These data support the concept that localized WMH expansion continues to proliferate with amyloid accumulation throughout the entirety of the disease in distinct spatial locations.
Collapse
Affiliation(s)
- Jeremy Fuller Strain
- Department of Neurology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Chia-Ling Phuah
- Department of Neurology, Washington University School of Medicine, St. Louis, Missouri, USA
- NeuroGenomics and Informatics Center, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Babatunde Adeyemo
- Department of Neurology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Kathleen Cheng
- Department of Neurology, Washington University School of Medicine, St. Louis, Missouri, USA
- Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, Missouri, USA
| | - Kyle B Womack
- Department of Neurology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - John McCarthy
- Department of Mathematics, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Manu Goyal
- Department of Radiology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Yasheng Chen
- Department of Neurology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Aristeidis Sotiras
- Department of Radiology, Washington University School of Medicine, St. Louis, Missouri, USA
- Institute for Informatics, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Hongyu An
- Department of Radiology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Chengjie Xiong
- Division of Biostatics, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Andrea Scharf
- Department of Biological Sciences, Missouri University for Science and Technology, Rolla, Missouri, USA
| | - Catherine Newsom-Stewart
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - John Carl Morris
- Department of Neurology, Washington University School of Medicine, St. Louis, Missouri, USA
- Knight Alzheimer Disease Research Center, St. Louis, Missouri, USA
| | - Tammie Lee Smith Benzinger
- Department of Radiology, Washington University School of Medicine, St. Louis, Missouri, USA
- Knight Alzheimer Disease Research Center, St. Louis, Missouri, USA
| | - Jin-Moo Lee
- Department of Neurology, Washington University School of Medicine, St. Louis, Missouri, USA
- Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, Missouri, USA
- Department of Radiology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Beau M Ances
- Department of Neurology, Washington University School of Medicine, St. Louis, Missouri, USA
- Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, Missouri, USA
- Department of Radiology, Washington University School of Medicine, St. Louis, Missouri, USA
- Knight Alzheimer Disease Research Center, St. Louis, Missouri, USA
| |
Collapse
|
46
|
Quattrini G, Ferrari C, Pievani M, Geviti A, Ribaldi F, Scheffler M, Frisoni GB, Garibotto V, Marizzoni M. Unsupervised [ 18F]Flortaucipir cutoffs for tau positivity and staging in Alzheimer's disease. Eur J Nucl Med Mol Imaging 2023; 50:3265-3275. [PMID: 37272955 PMCID: PMC10542510 DOI: 10.1007/s00259-023-06280-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Accepted: 05/19/2023] [Indexed: 06/06/2023]
Abstract
PURPOSE Several [18F]Flortaucipir cutoffs have been proposed for tau PET positivity (T+) in Alzheimer's disease (AD), but none were data-driven. The aim of this study was to establish and validate unsupervised T+ cutoffs by applying Gaussian mixture models (GMM). METHODS Amyloid negative (A-) cognitively normal (CN) and amyloid positive (A+) AD-related dementia (ADRD) subjects from ADNI (n=269) were included. ADNI (n=475) and Geneva Memory Clinic (GMC) cohorts (n=98) were used for validation. GMM-based cutoffs were extracted for the temporal meta-ROI, and validated against previously published cutoffs and visual rating. RESULTS GMM-based cutoffs classified less subjects as T+, mainly in the A- CN (<3.4% vs >28.5%) and A+ CN (<14.5% vs >42.9%) groups and showed higher agreement with visual rating (ICC=0.91 vs ICC<0.62) than published cutoffs. CONCLUSION We provided reliable data-driven [18F]Flortaucipir cutoffs for in vivo T+ detection in AD. These cutoffs might be useful to select participants in clinical and research studies.
Collapse
Affiliation(s)
- Giulia Quattrini
- Laboratory of Alzheimer's Neuroimaging and Epidemiology (LANE), IRCCS Istituto Centro San Giovanni di Dio Fatebenefratelli, 25125, Brescia, Italy
- Department of Molecular and Translational Medicine, University of Brescia, 25123, Brescia, Italy
| | - Clarissa Ferrari
- FONDAZIONE POLIAMBULANZA ISTITUTO OSPEDALIERO via Bissolati, 57, 25124, Brescia, Italy
- Unit of Statistics, IRCCS Istituto Centro San Giovanni di Dio Fatebenefratelli, 25125, Brescia, Italy
| | - Michela Pievani
- Laboratory of Alzheimer's Neuroimaging and Epidemiology (LANE), IRCCS Istituto Centro San Giovanni di Dio Fatebenefratelli, 25125, Brescia, Italy
| | - Andrea Geviti
- Unit of Statistics, IRCCS Istituto Centro San Giovanni di Dio Fatebenefratelli, 25125, Brescia, Italy
| | - Federica Ribaldi
- LANVIE - Laboratory of Neuroimaging of Aging, University Hospitals and University of Geneva, 1205, Geneva, Switzerland
- Geneva Memory Center, Department of Rehabilitation and Geriatrics, Geneva University Hospitals, 1205, Geneva, Switzerland
| | - Max Scheffler
- Division of Radiology, Geneva University Hospitals, Geneva, Switzerland
| | - Giovanni B Frisoni
- LANVIE - Laboratory of Neuroimaging of Aging, University Hospitals and University of Geneva, 1205, Geneva, Switzerland
- Geneva Memory Center, Department of Rehabilitation and Geriatrics, Geneva University Hospitals, 1205, Geneva, Switzerland
| | - Valentina Garibotto
- Laboratory of Neuroimaging and Innovative Molecular Tracers (NIMTlab), Geneva University Neurocentre, Faculty of Medicine, University of Geneva, 1205, Geneva, Switzerland
- Division of Nuclear Medicine and Molecular Imaging, University Hospitals of Geneva, 1205, Geneva, Switzerland
- Centre for Biomedical Imaging (CIBM), 1205, Geneva, Switzerland
| | - Moira Marizzoni
- Laboratory of Alzheimer's Neuroimaging and Epidemiology (LANE), IRCCS Istituto Centro San Giovanni di Dio Fatebenefratelli, 25125, Brescia, Italy.
- Biological Psychiatric Unit, IRCCS Istituto Centro San Giovanni di Dio Fatebenefratelli, 25125, Brescia, Italy.
| |
Collapse
|
47
|
S R M, S S. Identification of Mild Cognitive Impairment Subtypes using an Interpretable Neural Network based Clustering of Gene Expression Data and Neuroimaging Markers. ANNUAL INTERNATIONAL CONFERENCE OF THE IEEE ENGINEERING IN MEDICINE AND BIOLOGY SOCIETY. IEEE ENGINEERING IN MEDICINE AND BIOLOGY SOCIETY. ANNUAL INTERNATIONAL CONFERENCE 2023; 2023:1-4. [PMID: 38082666 DOI: 10.1109/embc40787.2023.10340584] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2023]
Abstract
In this study, an attempt is made to cluster the gene expression data and neuroimaging markers using an interpretable neural network model to identify Mild Cognitive Impairment (MCI) subtypes. For this, structural Magnetic Resonance (MR) brain images and gene expression data of early and late MCI subjects are considered from a public database. A neural network model is employed to cluster the gene expression data and regional MR volumes. To evaluate the performance of model, clustering metrics are employed and model is explained using perturbation-based method. Results indicate that the developed model is able to identify MCI subtypes. The network learns latent embeddings of disease-specific genes and MR images markers. The clustering metrics are found to be highest when both the imaging and genetic markers are employed. Volumes of lateral ventricles, hippocampus, amygdala and thalamus are found to be associated with late MCI. Significant scores suggest that genes such as StAR, CCDC108, APOO, TRMT13, RASAL2 and ZNF43 play a key role in identifying the MCI subtypes.Clinical Relevance-Identifying distinct MCI subtypes offer potential for precision diagnostics and targeted clinical recruitment.
Collapse
|
48
|
Al Olaimat M, Martinez J, Saeed F, Bozdag S, Alzheimer’s Disease Neuroimaging Initiative. PPAD: a deep learning architecture to predict progression of Alzheimer's disease. Bioinformatics 2023; 39:i149-i157. [PMID: 37387135 PMCID: PMC10311312 DOI: 10.1093/bioinformatics/btad249] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/01/2023] Open
Abstract
MOTIVATION Alzheimer's disease (AD) is a neurodegenerative disease that affects millions of people worldwide. Mild cognitive impairment (MCI) is an intermediary stage between cognitively normal state and AD. Not all people who have MCI convert to AD. The diagnosis of AD is made after significant symptoms of dementia such as short-term memory loss are already present. Since AD is currently an irreversible disease, diagnosis at the onset of the disease brings a huge burden on patients, their caregivers, and the healthcare sector. Thus, there is a crucial need to develop methods for the early prediction AD for patients who have MCI. Recurrent neural networks (RNN) have been successfully used to handle electronic health records (EHR) for predicting conversion from MCI to AD. However, RNN ignores irregular time intervals between successive events which occurs common in electronic health record data. In this study, we propose two deep learning architectures based on RNN, namely Predicting Progression of Alzheimer's Disease (PPAD) and PPAD-Autoencoder. PPAD and PPAD-Autoencoder are designed for early predicting conversion from MCI to AD at the next visit and multiple visits ahead for patients, respectively. To minimize the effect of the irregular time intervals between visits, we propose using age in each visit as an indicator of time change between successive visits. RESULTS Our experimental results conducted on Alzheimer's Disease Neuroimaging Initiative and National Alzheimer's Coordinating Center datasets showed that our proposed models outperformed all baseline models for most prediction scenarios in terms of F2 and sensitivity. We also observed that the age feature was one of top features and was able to address irregular time interval problem. AVAILABILITY AND IMPLEMENTATION https://github.com/bozdaglab/PPAD.
Collapse
Affiliation(s)
- Mohammad Al Olaimat
- Department of Computer Science and Engineering, University of North Texas, Denton, TX, United States
| | - Jared Martinez
- Department of Computer Science and Engineering, University of North Texas, Denton, TX, United States
| | - Fahad Saeed
- School of Computing and Information Sciences, Florida International University, Miami, FL, United States
| | - Serdar Bozdag
- Department of Computer Science and Engineering, University of North Texas, Denton, TX, United States
- Department of Mathematics, University of North Texas, Denton, TX, United States
- BioDiscovery Institute, University of North Texas, Denton, TX, United States
| | | |
Collapse
|
49
|
Lee BN, Wang J, Nho K, Saykin AJ, Shen L. Discovering Precision AD Biomarkers with Varying Prognosis Effects in Genetics Driven Subpopulations. AMIA JOINT SUMMITS ON TRANSLATIONAL SCIENCE PROCEEDINGS. AMIA JOINT SUMMITS ON TRANSLATIONAL SCIENCE 2023; 2023:340-349. [PMID: 37350892 PMCID: PMC10283147] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 06/24/2023]
Abstract
Alzheimer's Disease (AD) is a highly heritable neurodegenerative disorder characterized by memory impairments. Understanding how genetic factors contribute to AD pathology may inform interventions to slow or prevent the progression of AD. We performed stratified genetic analyses of 1,574 Alzheimer's Disease Neuroimaging Initiative (ADNI) participants to examine associations between levels of quantitative traits (QT's) and future diagnosis. The Chow test was employed to determine if an individual's genetic profile affects identified predictive relationships between QT's and future diagnosis. Our chow test analysis discovered that cognitive and PET-based biomarkers differentially predicted future diagnosis when stratifying on allelic dosage of AD loci. Post-hoc bootstrapped and association analyses of biomarkers confirmed differential effects, emphasizing the necessity of stratified models to realize individualized AD diagnosis prediction. This novel application of the Chow test allows for the quantification and direct comparison of genetic-based differences. Our findings, as well as the identified QT-future diagnosis relationships, warrant future investigation from a biological context.
Collapse
Affiliation(s)
- Brian N Lee
- University of Pennsylvania, Philadelphia, PA, USA
| | | | | | | | - Li Shen
- University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
50
|
Hua L, Gao F, Xia X, Guo Q, Zhao Y, Huang S, Yuan Z. Individual-specific functional connectivity improves prediction of Alzheimer's disease's symptoms in elderly people regardless of APOE ε4 genotype. Commun Biol 2023; 6:581. [PMID: 37258640 PMCID: PMC10232409 DOI: 10.1038/s42003-023-04952-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Accepted: 05/18/2023] [Indexed: 06/02/2023] Open
Abstract
To date, reliable biomarkers remain unclear that could link functional connectivity to patients' symptoms for detecting and predicting the process from normal aging to Alzheimer's disease (AD) in elderly people with specific genotypes. To address this, individual-specific functional connectivity is constructed for elderly participants with/without APOE ε4 allele. Then, we utilize recursive feature selection-based machine learning to reveal individual brain-behavior relationships and to predict the symptom transition in different genotypes. Our findings reveal that compared with conventional atlas-based functional connectivity, individual-specific functional connectivity exhibits higher classification and prediction performance from normal aging to AD in both APOE ε4 groups, while no significant performance is detected when the data of two genotyping groups are combined. Furthermore, individual-specific between-network connectivity constitutes a major contributor to assessing cognitive symptoms. This study highlights the essential role of individual variation in cortical functional anatomy and the integration of brain and behavior in predicting individualized symptoms.
Collapse
Affiliation(s)
- Lin Hua
- Faculty of Health Sciences, University of Macau, Avenida da Universidade, Taipa, Macau, SAR 999078, China
- Centre for Cognitive and Brain Sciences, University of Macau, Avenida da Universidade, Taipa, Macau, SAR 999078, China
| | - Fei Gao
- Institute of Modern Languages and Linguistics, Fudan University, Shanghai, 200433, China
| | - Xiaoluan Xia
- Faculty of Health Sciences, University of Macau, Avenida da Universidade, Taipa, Macau, SAR 999078, China
- Centre for Cognitive and Brain Sciences, University of Macau, Avenida da Universidade, Taipa, Macau, SAR 999078, China
| | - Qiwei Guo
- Faculty of Health Sciences, University of Macau, Avenida da Universidade, Taipa, Macau, SAR 999078, China
- Centre for Cognitive and Brain Sciences, University of Macau, Avenida da Universidade, Taipa, Macau, SAR 999078, China
| | - Yonghua Zhao
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Avenida da Universidade, Taipa, Macau, SAR 999078, China
| | - Shaohui Huang
- Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China
| | - Zhen Yuan
- Faculty of Health Sciences, University of Macau, Avenida da Universidade, Taipa, Macau, SAR 999078, China.
- Centre for Cognitive and Brain Sciences, University of Macau, Avenida da Universidade, Taipa, Macau, SAR 999078, China.
| |
Collapse
|