101
|
Hanyu H, Koyama Y, Horita H, Watanabe S, Sato T, Kanetaka H, Shimizu S, Hirao K. Longitudinal patterns of Alzheimer's disease subtypes: A follow-up magnetic resonance imaging and single-photon emission computed tomography study. Geriatr Gerontol Int 2023; 23:919-924. [PMID: 37905589 DOI: 10.1111/ggi.14712] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2023] [Revised: 09/21/2023] [Accepted: 10/07/2023] [Indexed: 11/02/2023]
Abstract
AIM Alzheimer's disease (AD) is a biologically heterogenous disease. In a previous study, we classified 245 patients with probable AD into the typical AD (TAD), limbic-predominant (LP), hippocampal-sparing (HS) and minimal-change (MC) subtypes based on their medial temporal lobe atrophy on magnetic resonance imaging and posterior hypoperfusion on single-photon emission computed tomography, and described differences in clinical features among the patients with different AD subtypes. This study aimed to clarify the longitudinal patterns of changes in patients with the various AD subtypes by follow-up brain imaging analyses. METHODS Follow-up magnetic resonance imaging or single-photon emission computed tomography data obtained 12-48 months after the first brain imaging were investigated in 79 patients with probable AD, comprising 25 of the TAD subtype, 19 of the LP subtype, 17 of the HS subtype and 18 of the MC subtype. RESULTS All patients of the TAD subtype remained as the same subtype at follow up. Approximately 37% of patients of the LP subtype and 29% of patients of the HS subtype progressed to the TAD subtype, and 17%, 33% and 6% of the MC subtype progressed to the TAD, LP and HS subtypes, respectively. The group of patients showing subtype progression was associated only with a longer follow-up duration. CONCLUSIONS There might be different progression patterns and progression rates of changes among the atypical AD subtypes. Further longitudinal brain imaging studies might provide information regarding the pathophysiological association between the various AD subtypes, and might be helpful for determining appropriate therapies and management methods. Geriatr Gerontol Int 2023; 23: 919-924.
Collapse
Affiliation(s)
- Haruo Hanyu
- Dementia Research Center, Tokyo General Hospital, Tokyo, Japan
- Department of Geriatric Medicine, Tokyo Medical University, Tokyo, Japan
| | - Yumi Koyama
- Department of Rehabilitation, Tokyo General Hospital, Tokyo, Japan
| | - Haruka Horita
- Department of Rehabilitation, Tokyo General Hospital, Tokyo, Japan
| | | | - Tomohiko Sato
- Department of Geriatric Medicine, Tokyo Medical University, Tokyo, Japan
| | - Hidekazu Kanetaka
- Department of Geriatric Medicine, Tokyo Medical University, Tokyo, Japan
| | - Soichiro Shimizu
- Department of Geriatric Medicine, Tokyo Medical University, Tokyo, Japan
| | - Kentaro Hirao
- Department of Geriatric Medicine, Tokyo Medical University, Tokyo, Japan
| |
Collapse
|
102
|
Fu Z, Zhao M, Li Y, He Y, Wang X, Zhou Z, Han Y, Li S. Heterogeneity in subjective cognitive decline in the Sino Longitudinal Study on Cognitive Decline(SILCODE): Empirically derived subtypes, structural and functional verification. CNS Neurosci Ther 2023; 29:4032-4042. [PMID: 37475187 PMCID: PMC10651943 DOI: 10.1111/cns.14327] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2023] [Revised: 03/01/2023] [Accepted: 06/17/2023] [Indexed: 07/22/2023] Open
Abstract
AIMS We evaluated whether Subjective Cognitive Decline (SCD) subtypes could be empirically derived within the Sino Longitudinal Study on Cognitive Decline (SILCODE) SCD cohort and examined associated neuroimaging markers, biomarkers, and clinical outcomes. METHODS A cluster analysis was performed on eight neuropsychological test scores from 124 SCD SILCODE participants and 57 normal control (NC) subjects. Structural and functional neuroimaging indices were used to evaluate the SCD subgroups. RESULTS Four subtypes emerged: (1) dysexecutive/mixed SCD (n = 23), (2) neuropsychiatric SCD (n = 24), (3) amnestic SCD (n = 22), and (4) cluster-derived normal (n = 55) who exhibited normal performance in neuropsychological tests. Compared with the NC group, each subgroup showed distinct patterns in gray matter (GM) volume and the amplitude of low-frequency fluctuations (ALFF). Lower fractional anisotropy (FA) values were only found in the neuropsychiatric SCD group relative to NC. CONCLUSION The identification of empirically derived SCD subtypes demonstrates the presence of heterogeneity in SCD neuropsychological profiles. The cluster-derived normal group may represent the majority of SCD individuals who do not show progressive cognitive decline; the dysexecutive/mixed SCD and amnestic SCD might represent high-risk groups with progressing cognitive decline; and finally, the neuropsychiatric SCD may represent a new topic in SCD research.
Collapse
Affiliation(s)
- Zhenrong Fu
- Key Laboratory of Adolescent Cyberpsychology and Behavior (CCNU)Ministry of EducationWuhanChina
- School of Psychology, Key Laboratory of Human Development and Mental Health of Hubei ProvinceCentral China Normal UniversityWuhanChina
| | - Mingyan Zhao
- Department of PsychologyTangshan Gongren HospitalTangshanChina
| | - Yuxia Li
- Department of NeurologyTangshan Central HospitalTangshanChina
- Department of NeurologyXuanwu Hospital of Capital Medical UniversityBeijingChina
| | - Yirong He
- State Key Laboratory of Cognitive Neuroscience and LearningBeijing Normal UniversityBeijingChina
| | - Xuetong Wang
- State Key Laboratory of Cognitive Neuroscience and LearningBeijing Normal UniversityBeijingChina
| | - Zongkui Zhou
- Key Laboratory of Adolescent Cyberpsychology and Behavior (CCNU)Ministry of EducationWuhanChina
- School of Psychology, Key Laboratory of Human Development and Mental Health of Hubei ProvinceCentral China Normal UniversityWuhanChina
| | - Ying Han
- Department of NeurologyXuanwu Hospital of Capital Medical UniversityBeijingChina
- Key Laboratory of Biomedical Engineering of Hainan Province, School of Biomedical EngineeringHainan UniversityHaikouChina
- Center of Alzheimer's DiseaseBeijing Institute for Brain DisordersBeijingChina
- National Clinical Research Center for Geriatric DisordersBeijingChina
- Institute of Biomedical EngineeringShenzhen Bay LaboratoryShenzhenChina
| | - Shuyu Li
- State Key Laboratory of Cognitive Neuroscience and LearningBeijing Normal UniversityBeijingChina
| |
Collapse
|
103
|
Doherty T, Yao Z, Khleifat AAL, Tantiangco H, Tamburin S, Albertyn C, Thakur L, Llewellyn DJ, Oxtoby NP, Lourida I, Ranson JM, Duce JA. Artificial intelligence for dementia drug discovery and trials optimization. Alzheimers Dement 2023; 19:5922-5933. [PMID: 37587767 DOI: 10.1002/alz.13428] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2023] [Revised: 05/26/2023] [Accepted: 07/05/2023] [Indexed: 08/18/2023]
Abstract
Drug discovery and clinical trial design for dementia have historically been challenging. In part these challenges have arisen from patient heterogeneity, length of disease course, and the tractability of a target for the brain. Applying big data analytics and machine learning tools for drug discovery and utilizing them to inform successful clinical trial design has the potential to accelerate progress. Opportunities arise at multiple stages in the therapy pipeline and the growing availability of large medical data sets opens possibilities for big data analyses to answer key questions in clinical and therapeutic challenges. However, before this goal is reached, several challenges need to be overcome and only a multi-disciplinary approach can promote data-driven decision-making to its full potential. Herein we review the current state of machine learning applications to clinical trial design and drug discovery, while presenting opportunities and recommendations that can break down the barriers to implementation.
Collapse
Affiliation(s)
- Thomas Doherty
- Eisai Europe Ltd, Hatfield, UK
- University of Westminster, London, UK
| | | | - Ahmad A L Khleifat
- Institute of Psychiatry, Psychology & Neuroscience, Department of Basic and Clinical Neuroscience, King's College London, London, UK
| | | | - Stefano Tamburin
- University of Verona, Department of Neurosciences, Biomedicine & Movement Sciences, Verona, Italy
| | - Chris Albertyn
- Department of Old Age Psychiatry, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| | - Lokendra Thakur
- Division of Genetics and Genomics, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts, USA
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - David J Llewellyn
- University of Exeter Medical School, Exeter, UK
- Alan Turing Institute, London, UK
| | - Neil P Oxtoby
- UCL Centre for Medical Image Computing, Department of Computer Science, University College London, London, UK
| | | | | | - James A Duce
- The ALBORADA Drug Discovery Institute, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK
| |
Collapse
|
104
|
Congdon EE, Ji C, Tetlow AM, Jiang Y, Sigurdsson EM. Tau-targeting therapies for Alzheimer disease: current status and future directions. Nat Rev Neurol 2023; 19:715-736. [PMID: 37875627 PMCID: PMC10965012 DOI: 10.1038/s41582-023-00883-2] [Citation(s) in RCA: 84] [Impact Index Per Article: 42.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/20/2023] [Indexed: 10/26/2023]
Abstract
Alzheimer disease (AD) is the most common cause of dementia in older individuals. AD is characterized pathologically by amyloid-β (Aβ) plaques and tau neurofibrillary tangles in the brain, with associated loss of synapses and neurons, which eventually results in dementia. Many of the early attempts to develop treatments for AD focused on Aβ, but a lack of efficacy of these treatments in terms of slowing disease progression led to a change of strategy towards targeting of tau pathology. Given that tau shows a stronger correlation with symptom severity than does Aβ, targeting of tau is more likely to be efficacious once cognitive decline begins. Anti-tau therapies initially focused on post-translational modifications, inhibition of tau aggregation and stabilization of microtubules. However, trials of many potential drugs were discontinued because of toxicity and/or lack of efficacy. Currently, the majority of tau-targeting agents in clinical trials are immunotherapies. In this Review, we provide an update on the results from the initial immunotherapy trials and an overview of new therapeutic candidates that are in clinical development, as well as considering future directions for tau-targeting therapies.
Collapse
Affiliation(s)
- Erin E Congdon
- Department of Neuroscience and Physiology, New York University Grossman School of Medicine, New York, NY, USA
- Neuroscience Institute, New York University Grossman School of Medicine, New York, NY, USA
| | - Changyi Ji
- Department of Neuroscience and Physiology, New York University Grossman School of Medicine, New York, NY, USA
- Neuroscience Institute, New York University Grossman School of Medicine, New York, NY, USA
| | - Amber M Tetlow
- Department of Neuroscience and Physiology, New York University Grossman School of Medicine, New York, NY, USA
- Neuroscience Institute, New York University Grossman School of Medicine, New York, NY, USA
| | - Yixiang Jiang
- Department of Neuroscience and Physiology, New York University Grossman School of Medicine, New York, NY, USA
- Neuroscience Institute, New York University Grossman School of Medicine, New York, NY, USA
| | - Einar M Sigurdsson
- Department of Neuroscience and Physiology, New York University Grossman School of Medicine, New York, NY, USA.
- Neuroscience Institute, New York University Grossman School of Medicine, New York, NY, USA.
- Department of Psychiatry, New York University Grossman School of Medicine, New York, NY, USA.
| |
Collapse
|
105
|
Ramos AA, Galiano-Castillo N, Machado L. Cognitive Functioning of Unaffected First-degree Relatives of Individuals With Late-onset Alzheimer's Disease: A Systematic Literature Review and Meta-analysis. Neuropsychol Rev 2023; 33:659-674. [PMID: 36057684 PMCID: PMC10770217 DOI: 10.1007/s11065-022-09555-2] [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: 12/05/2021] [Accepted: 06/10/2022] [Indexed: 10/14/2022]
Abstract
First-degree relatives of individuals with late-onset Alzheimer's disease (LOAD) are at increased risk for developing dementia, yet the associations between family history of LOAD and cognitive dysfunction remain unclear. In this quantitative review, we provide the first meta-analysis on the cognitive profile of unaffected first-degree blood relatives of LOAD-affected individuals compared to controls without a family history of LOAD. A systematic literature search was conducted in PsycINFO, PubMed /MEDLINE, and Scopus. We fitted a three-level structural equation modeling meta-analysis to control for non-independent effect sizes. Heterogeneity and risk of publication bias were also investigated. Thirty-four studies enabled us to estimate 218 effect sizes across several cognitive domains. Overall, first-degree relatives (n = 4,086, mean age = 57.40, SD = 4.71) showed significantly inferior cognitive performance (Hedges' g = -0.16; 95% CI, -0.25 to -0.08; p < .001) compared to controls (n = 2,388, mean age = 58.43, SD = 5.69). Specifically, controls outperformed first-degree relatives in language, visuospatial and verbal long-term memory, executive functions, verbal short-term memory, and verbal IQ. Among the first-degree relatives, APOE ɛ4 carriership was associated with more significant dysfunction in cognition (g = -0.24; 95% CI, -0.38 to -0.11; p < .001) compared to non-carriers (g = -0.14; 95% CI, -0.28 to -0.01; p = .04). Cognitive test type was significantly associated with between-group differences, accounting for 65% (R23 = .6499) of the effect size heterogeneity in the fitted regression model. No evidence of publication bias was found. The current findings provide support for mild but robust cognitive dysfunction in first-degree relatives of LOAD-affected individuals that appears to be moderated by cognitive domain, cognitive test type, and APOE ɛ4.
Collapse
Affiliation(s)
- Ari Alex Ramos
- Department of Psychology and Brain Health Research Centre, University of Otago, Dunedin, New Zealand.
- Brain Research New Zealand, Auckland, New Zealand.
- Department of Psychology, Pontifical Catholic University of Paraná, Rua Imaculada Conceição, 1155, Curitiba, CEP 80.215-901, Brazil.
| | - Noelia Galiano-Castillo
- Department of Physical Therapy, Health Sciences Faculty, "Cuidate" from Biomedical Group (BIO277), Instituto de Investigación Biosanitaria (ibs.GRANADA), and Sport and Health Research Center (IMUDs), Granada, Spain, University of Granada, Granada, Spain
| | - Liana Machado
- Department of Psychology and Brain Health Research Centre, University of Otago, Dunedin, New Zealand
- Brain Research New Zealand, Auckland, New Zealand
| |
Collapse
|
106
|
Park HY, Shim WH, Suh CH, Heo H, Oh HW, Kim J, Sung J, Lim JS, Lee JH, Kim HS, Kim SJ. Development and validation of an automatic classification algorithm for the diagnosis of Alzheimer's disease using a high-performance interpretable deep learning network. Eur Radiol 2023; 33:7992-8001. [PMID: 37170031 DOI: 10.1007/s00330-023-09708-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Revised: 03/01/2023] [Accepted: 03/18/2023] [Indexed: 05/13/2023]
Abstract
OBJECTIVES To develop and validate an automatic classification algorithm for diagnosing Alzheimer's disease (AD) or mild cognitive impairment (MCI). METHODS AND MATERIALS This study evaluated a high-performance interpretable network algorithm (TabNet) and compared its performance with that of XGBoost, a widely used classifier. Brain segmentation was performed using a commercially approved software. TabNet and XGBoost were trained on the volumes or radiomics features of 102 segmented regions for classifying subjects into AD, MCI, or cognitively normal (CN) groups. The diagnostic performances of the two algorithms were compared using areas under the curves (AUCs). Additionally, 20 deep learning-based AD signature areas were investigated. RESULTS Between December 2014 and March 2017, 161 AD, 153 MCI, and 306 CN cases were enrolled. Another 120 AD, 90 MCI, and 141 CN cases were included for the internal validation. Public datasets were used for external validation. TabNet with volume features had an AUC of 0.951 (95% confidence interval [CI], 0.947-0.955) for AD vs CN, which was similar to that of XGBoost (0.953 [95% CI, 0.951-0.955], p = 0.41). External validation revealed the similar performances of two classifiers using volume features (0.871 vs. 0.871, p = 0.86). Likewise, two algorithms showed similar performances with one another in classifying MCI. The addition of radiomics data did not improve the performance of TabNet. TabNet and XGBoost focused on the same 13/20 regions of interest, including the hippocampus, inferior lateral ventricle, and entorhinal cortex. CONCLUSIONS TabNet shows high performance in AD classification and detailed interpretation of the selected regions. CLINICAL RELEVANCE STATEMENT Using a high-performance interpretable deep learning network, the automatic classification algorithm assisted in accurate Alzheimer's disease detection using 3D T1-weighted brain MRI and detailed interpretation of the selected regions. KEY POINTS • MR volumetry data revealed that TabNet had a high diagnostic performance in differentiating Alzheimer's disease (AD) from cognitive normal cases, which was comparable with that of XGBoost. • The addition of radiomics data to the volume data did not improve the diagnostic performance of TabNet. • Both TabNet and XGBoost selected the clinically meaningful regions of interest in AD, including the hippocampus, inferior lateral ventricle, and entorhinal cortex.
Collapse
Affiliation(s)
- Ho Young Park
- Department of Radiology and Research Institute of Radiology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Woo Hyun Shim
- Department of Convergence Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Chong Hyun Suh
- Department of Radiology and Research Institute of Radiology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea.
| | - Hwon Heo
- Department of Convergence Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | | | | | | | - Jae-Sung Lim
- Department of Neurology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Jae-Hong Lee
- Department of Neurology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Ho Sung Kim
- Department of Radiology and Research Institute of Radiology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Sang Joon Kim
- Department of Radiology and Research Institute of Radiology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| |
Collapse
|
107
|
Poulakis K, Westman E. Clustering and disease subtyping in Neuroscience, toward better methodological adaptations. Front Comput Neurosci 2023; 17:1243092. [PMID: 37927546 PMCID: PMC10620518 DOI: 10.3389/fncom.2023.1243092] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Accepted: 10/04/2023] [Indexed: 11/07/2023] Open
Affiliation(s)
- Konstantinos Poulakis
- Division of Clinical Geriatrics, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden
| | - Eric Westman
- Division of Clinical Geriatrics, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden
- Department of Neuroimaging, Centre for Neuroimaging Sciences, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, United Kingdom
| |
Collapse
|
108
|
Ding J, Shen C, Wang Z, Yang Y, El Fakhri G, Lu J, Liang D, Zheng H, Zhou Y, Sun T. Tau-PET abnormality as a biomarker for Alzheimer's disease staging and early detection: a topological perspective. Cereb Cortex 2023; 33:10649-10659. [PMID: 37653600 DOI: 10.1093/cercor/bhad312] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2023] [Revised: 08/10/2023] [Accepted: 08/10/2023] [Indexed: 09/02/2023] Open
Abstract
Alzheimer's disease can be detected early through biomarkers such as tau positron emission tomography (PET) imaging, which shows abnormal protein accumulations in the brain. The standardized uptake value ratio (SUVR) is often used to quantify tau-PET imaging, but topological information from multiple brain regions is also linked to tau pathology. Here a new method was developed to investigate the correlations between brain regions using subject-level tau networks. Participants with cognitive normal (74), early mild cognitive impairment (35), late mild cognitive impairment (32), and Alzheimer's disease (40) were included. The abnormality network from each scan was constructed to extract topological features, and 7 functional clusters were further analyzed for connectivity strengths. Results showed that the proposed method performed better than conventional SUVR measures for disease staging and prodromal sign detection. For example, when to differ healthy subjects with and without amyloid deposition, topological biomarker is significant with P < 0.01, SUVR is not with P > 0.05. Functionally significant clusters, i.e. medial temporal lobe, default mode network, and visual-related regions, were identified as critical hubs vulnerable to early disease conversion before mild cognitive impairment. These findings were replicated in an independent data cohort, demonstrating the potential to monitor the early sign and progression of Alzheimer's disease from a topological perspective for individual.
Collapse
Affiliation(s)
- Jie Ding
- Paul C. Lauterbur Research Center for Biomedical Imaging, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 100864, People's Republic of China
| | - Chushu Shen
- Paul C. Lauterbur Research Center for Biomedical Imaging, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 100864, People's Republic of China
| | - Zhenguo Wang
- Paul C. Lauterbur Research Center for Biomedical Imaging, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 100864, People's Republic of China
| | - Yongfeng Yang
- Paul C. Lauterbur Research Center for Biomedical Imaging, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 100864, People's Republic of China
| | - Georges El Fakhri
- Gordon Center for Medical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts 02114, United States
| | - Jie Lu
- Department of Radiology and Nuclear Medicine, Xuanwu Hospital, Capital Medical University, Beijing 100053, People's Republic of China
| | - Dong Liang
- Paul C. Lauterbur Research Center for Biomedical Imaging, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 100864, People's Republic of China
| | - Hairong Zheng
- Paul C. Lauterbur Research Center for Biomedical Imaging, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 100864, People's Republic of China
| | - Yun Zhou
- Central Research Institute, United Imaging Healthcare Group Co., Ltd, Shanghai 201807, People's Republic of China
- School of Biomedical Engineering, Shanghai Tech University, Shanghai 201210, People's Republic of China
| | - Tao Sun
- Paul C. Lauterbur Research Center for Biomedical Imaging, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 100864, People's Republic of China
- United Imaging Research Institute of Innovative Medical Equipment, Shenzhen 518055, People's Republic of China
| |
Collapse
|
109
|
Boon BDC, Labuzan SA, Peng Z, Matchett BJ, Kouri N, Hinkle KM, Lachner C, Ross OA, Ertekin-Taner N, Carter RE, Ferman TJ, Duara R, Dickson DW, Graff-Radford NR, Murray ME. Retrospective Evaluation of Neuropathologic Proxies of the Minimal Atrophy Subtype Compared With Corticolimbic Alzheimer Disease Subtypes. Neurology 2023; 101:e1412-e1423. [PMID: 37580158 PMCID: PMC10573142 DOI: 10.1212/wnl.0000000000207685] [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: 01/03/2023] [Accepted: 06/07/2023] [Indexed: 08/16/2023] Open
Abstract
BACKGROUND AND OBJECTIVES Alzheimer disease (AD) is neuropathologically classified into 3 corticolimbic subtypes based on the neurofibrillary tangle distribution throughout the hippocampus and association cortices: limbic predominant, typical, and hippocampal sparing. In vivo, a fourth subtype, dubbed "minimal atrophy," was identified using structural MRI. The objective of this study was to identify a neuropathologic proxy for the neuroimaging-defined minimal atrophy subtype. METHODS We applied 2 strategies in the Florida Autopsied Multi-Ethnic (FLAME) cohort to evaluate a neuropathologic proxy for the minimal atrophy subtype. In the first strategy, we selected AD cases with a Braak tangle stage IV (Braak IV) because of the relative paucity of neocortical tangle involvement compared with Braak >IV. Braak IV cases were compared with the 3 AD subtypes. In the alternative strategy, typical AD was stratified by brain weight and cases having a relatively high brain weight (>75th percentile) were defined as minimal atrophy. RESULTS Braak IV cases (n = 37) differed from AD subtypes (limbic predominant [n = 174], typical [n = 986], and hippocampal sparing [n = 187] AD) in having the least years of education (median 12 years, group-wise p < 0.001) and the highest brain weight (median 1,140 g, p = 0.002). Braak IV cases most resembled the limbic predominant cases owing to their high proportion of APOE ε4 carriers (75%, p < 0.001), an amnestic syndrome (100%, p < 0.001), as well as older age of cognitive symptom onset and death (median 79 and 85 years, respectively, p < 0.001). Only 5% of Braak IV cases had amygdala-predominant Lewy bodies (the lowest frequency observed, p = 0.017), whereas 32% had coexisting pathology of Lewy body disease, which was greater than the other subtypes (p = 0.005). Nearly half (47%) of the Braak IV samples had coexisting limbic predominant age-related TAR DNA-binding protein 43 encephalopathy neuropathologic change. Cases with a high brain weight (n = 201) were less likely to have amygdala-predominant Lewy bodies (14%, p = 0.006) and most likely to have Lewy body disease (31%, p = 0.042) compared with those with middle (n = 455) and low (n = 203) brain weight. DISCUSSION The frequency of Lewy body disease was increased in both neuropathologic proxies of the minimal atrophy subtype. We hypothesize that Lewy body disease may underlie cognitive decline observed in minimal atrophy cases.
Collapse
Affiliation(s)
- Baayla D C Boon
- From the Department of Neuroscience (B.D.C.B., S.A.L., B.J.M., N.K., K.M.H., O.A.R., N.E.-T., D.W.D., M.E.M.), Department of Quantitative Health Sciences (Z.P., R.E.C.), Department of Neurology (C.L., N.E.-T., N.R.G.-R.), and Department of Psychiatry & Psychology (C.L., T.J.F.), Mayo Clinic, Jacksonville; and Wien Center for Alzheimer's Disease and Memory Disorders (R.D.), Mount Sinai Medical Center, Miami Beach, FL
| | - Sydney A Labuzan
- From the Department of Neuroscience (B.D.C.B., S.A.L., B.J.M., N.K., K.M.H., O.A.R., N.E.-T., D.W.D., M.E.M.), Department of Quantitative Health Sciences (Z.P., R.E.C.), Department of Neurology (C.L., N.E.-T., N.R.G.-R.), and Department of Psychiatry & Psychology (C.L., T.J.F.), Mayo Clinic, Jacksonville; and Wien Center for Alzheimer's Disease and Memory Disorders (R.D.), Mount Sinai Medical Center, Miami Beach, FL
| | - Zhongwei Peng
- From the Department of Neuroscience (B.D.C.B., S.A.L., B.J.M., N.K., K.M.H., O.A.R., N.E.-T., D.W.D., M.E.M.), Department of Quantitative Health Sciences (Z.P., R.E.C.), Department of Neurology (C.L., N.E.-T., N.R.G.-R.), and Department of Psychiatry & Psychology (C.L., T.J.F.), Mayo Clinic, Jacksonville; and Wien Center for Alzheimer's Disease and Memory Disorders (R.D.), Mount Sinai Medical Center, Miami Beach, FL
| | - Billie J Matchett
- From the Department of Neuroscience (B.D.C.B., S.A.L., B.J.M., N.K., K.M.H., O.A.R., N.E.-T., D.W.D., M.E.M.), Department of Quantitative Health Sciences (Z.P., R.E.C.), Department of Neurology (C.L., N.E.-T., N.R.G.-R.), and Department of Psychiatry & Psychology (C.L., T.J.F.), Mayo Clinic, Jacksonville; and Wien Center for Alzheimer's Disease and Memory Disorders (R.D.), Mount Sinai Medical Center, Miami Beach, FL
| | - Naomi Kouri
- From the Department of Neuroscience (B.D.C.B., S.A.L., B.J.M., N.K., K.M.H., O.A.R., N.E.-T., D.W.D., M.E.M.), Department of Quantitative Health Sciences (Z.P., R.E.C.), Department of Neurology (C.L., N.E.-T., N.R.G.-R.), and Department of Psychiatry & Psychology (C.L., T.J.F.), Mayo Clinic, Jacksonville; and Wien Center for Alzheimer's Disease and Memory Disorders (R.D.), Mount Sinai Medical Center, Miami Beach, FL
| | - Kelly M Hinkle
- From the Department of Neuroscience (B.D.C.B., S.A.L., B.J.M., N.K., K.M.H., O.A.R., N.E.-T., D.W.D., M.E.M.), Department of Quantitative Health Sciences (Z.P., R.E.C.), Department of Neurology (C.L., N.E.-T., N.R.G.-R.), and Department of Psychiatry & Psychology (C.L., T.J.F.), Mayo Clinic, Jacksonville; and Wien Center for Alzheimer's Disease and Memory Disorders (R.D.), Mount Sinai Medical Center, Miami Beach, FL
| | - Christian Lachner
- From the Department of Neuroscience (B.D.C.B., S.A.L., B.J.M., N.K., K.M.H., O.A.R., N.E.-T., D.W.D., M.E.M.), Department of Quantitative Health Sciences (Z.P., R.E.C.), Department of Neurology (C.L., N.E.-T., N.R.G.-R.), and Department of Psychiatry & Psychology (C.L., T.J.F.), Mayo Clinic, Jacksonville; and Wien Center for Alzheimer's Disease and Memory Disorders (R.D.), Mount Sinai Medical Center, Miami Beach, FL
| | - Owen A Ross
- From the Department of Neuroscience (B.D.C.B., S.A.L., B.J.M., N.K., K.M.H., O.A.R., N.E.-T., D.W.D., M.E.M.), Department of Quantitative Health Sciences (Z.P., R.E.C.), Department of Neurology (C.L., N.E.-T., N.R.G.-R.), and Department of Psychiatry & Psychology (C.L., T.J.F.), Mayo Clinic, Jacksonville; and Wien Center for Alzheimer's Disease and Memory Disorders (R.D.), Mount Sinai Medical Center, Miami Beach, FL
| | - Nilufer Ertekin-Taner
- From the Department of Neuroscience (B.D.C.B., S.A.L., B.J.M., N.K., K.M.H., O.A.R., N.E.-T., D.W.D., M.E.M.), Department of Quantitative Health Sciences (Z.P., R.E.C.), Department of Neurology (C.L., N.E.-T., N.R.G.-R.), and Department of Psychiatry & Psychology (C.L., T.J.F.), Mayo Clinic, Jacksonville; and Wien Center for Alzheimer's Disease and Memory Disorders (R.D.), Mount Sinai Medical Center, Miami Beach, FL
| | - Rickey E Carter
- From the Department of Neuroscience (B.D.C.B., S.A.L., B.J.M., N.K., K.M.H., O.A.R., N.E.-T., D.W.D., M.E.M.), Department of Quantitative Health Sciences (Z.P., R.E.C.), Department of Neurology (C.L., N.E.-T., N.R.G.-R.), and Department of Psychiatry & Psychology (C.L., T.J.F.), Mayo Clinic, Jacksonville; and Wien Center for Alzheimer's Disease and Memory Disorders (R.D.), Mount Sinai Medical Center, Miami Beach, FL
| | - Tanis J Ferman
- From the Department of Neuroscience (B.D.C.B., S.A.L., B.J.M., N.K., K.M.H., O.A.R., N.E.-T., D.W.D., M.E.M.), Department of Quantitative Health Sciences (Z.P., R.E.C.), Department of Neurology (C.L., N.E.-T., N.R.G.-R.), and Department of Psychiatry & Psychology (C.L., T.J.F.), Mayo Clinic, Jacksonville; and Wien Center for Alzheimer's Disease and Memory Disorders (R.D.), Mount Sinai Medical Center, Miami Beach, FL
| | - Ranjan Duara
- From the Department of Neuroscience (B.D.C.B., S.A.L., B.J.M., N.K., K.M.H., O.A.R., N.E.-T., D.W.D., M.E.M.), Department of Quantitative Health Sciences (Z.P., R.E.C.), Department of Neurology (C.L., N.E.-T., N.R.G.-R.), and Department of Psychiatry & Psychology (C.L., T.J.F.), Mayo Clinic, Jacksonville; and Wien Center for Alzheimer's Disease and Memory Disorders (R.D.), Mount Sinai Medical Center, Miami Beach, FL
| | - Dennis W Dickson
- From the Department of Neuroscience (B.D.C.B., S.A.L., B.J.M., N.K., K.M.H., O.A.R., N.E.-T., D.W.D., M.E.M.), Department of Quantitative Health Sciences (Z.P., R.E.C.), Department of Neurology (C.L., N.E.-T., N.R.G.-R.), and Department of Psychiatry & Psychology (C.L., T.J.F.), Mayo Clinic, Jacksonville; and Wien Center for Alzheimer's Disease and Memory Disorders (R.D.), Mount Sinai Medical Center, Miami Beach, FL
| | - Neill R Graff-Radford
- From the Department of Neuroscience (B.D.C.B., S.A.L., B.J.M., N.K., K.M.H., O.A.R., N.E.-T., D.W.D., M.E.M.), Department of Quantitative Health Sciences (Z.P., R.E.C.), Department of Neurology (C.L., N.E.-T., N.R.G.-R.), and Department of Psychiatry & Psychology (C.L., T.J.F.), Mayo Clinic, Jacksonville; and Wien Center for Alzheimer's Disease and Memory Disorders (R.D.), Mount Sinai Medical Center, Miami Beach, FL
| | - Melissa E Murray
- From the Department of Neuroscience (B.D.C.B., S.A.L., B.J.M., N.K., K.M.H., O.A.R., N.E.-T., D.W.D., M.E.M.), Department of Quantitative Health Sciences (Z.P., R.E.C.), Department of Neurology (C.L., N.E.-T., N.R.G.-R.), and Department of Psychiatry & Psychology (C.L., T.J.F.), Mayo Clinic, Jacksonville; and Wien Center for Alzheimer's Disease and Memory Disorders (R.D.), Mount Sinai Medical Center, Miami Beach, FL.
| |
Collapse
|
110
|
Crnich E, Sanchez E, Havens MA, Kissel DS. Sulfur-bridging the gap: investigating the electrochemistry of novel copper chelating agents for Alzheimer's disease applications. J Biol Inorg Chem 2023; 28:643-653. [PMID: 37594567 DOI: 10.1007/s00775-023-02013-1] [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: 01/09/2023] [Accepted: 07/15/2023] [Indexed: 08/19/2023]
Abstract
There is currently an unmet demand for multi-functional precision treatments for Alzheimer's disease (AD) after several failed attempts at designing drugs based on the amyloid hypothesis. The focus of this work is to investigate sulfur-bridged quinoline ligands that could potentially be used in chelation therapies for a subpopulation of AD patients presenting with an overload of labile copper ions, which are known to catalyze the production of reactive oxygen species (ROS) and exacerbate other markers of AD progression. The ligands 1-(2'-thiopyridyl)isoquinoline (1TPIQ) and 2-(2'-thiopyridyl)quinoline (2TPQ) were synthesized and characterized before being electrochemically investigated in the presence of different oxidizing and reducing agents in solution with a physiological pH relevant to the brain. The electrochemical response of each compound with copper was studied by employing both hydrogen peroxide (H2O2) as an oxidizing agent and ascorbic acid (AA) as an antioxidant during analysis using cyclic voltammetry (CV). The cyclic voltammograms of each quinoline were compared with similar ligands that contained aromatic N-donor groups but no sulfur groups to provide relative electrochemical properties of each complex in solution. In a dose-dependent manner, it was observed that AA exerted dual-efficacy when combined with these chelating ligands: promoting synergistic metal binding while also scavenging harmful ROS, suggesting AA is an effective adjuvant therapeutic agent. Overall, this study shows how coordination by sulfur-bridged quinoline ligands can alter copper electrochemistry in the presence of AA to limit ROS production in solution.
Collapse
Affiliation(s)
- Emma Crnich
- Department of Biology, Lewis University, One University Pkwy, Romeoville, IL, 60446, USA
| | - Erik Sanchez
- Department of Chemistry, Lewis University, One University Pkwy, Romeoville, IL, 60446, USA
| | - Mallory A Havens
- Department of Biology, Lewis University, One University Pkwy, Romeoville, IL, 60446, USA
| | - Daniel S Kissel
- Department of Chemistry, Lewis University, One University Pkwy, Romeoville, IL, 60446, USA.
| |
Collapse
|
111
|
Yue J, Shi Y. Uncovering Heterogeneity in Alzheimer's Disease from Graphical Modeling of the Tau Spatiotemporal Topography. MEDICAL IMAGE COMPUTING AND COMPUTER-ASSISTED INTERVENTION : MICCAI ... INTERNATIONAL CONFERENCE ON MEDICAL IMAGE COMPUTING AND COMPUTER-ASSISTED INTERVENTION 2023; 14224:262-271. [PMID: 38510994 PMCID: PMC10951551 DOI: 10.1007/978-3-031-43904-9_26] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/22/2024]
Abstract
Growing evidence from post-mortem and in vivo studies have demonstrated the substantial variability of tau pathology spreading patterns in Alzheimer's disease(AD). Automated tools for characterizing the heterogeneity of tau pathology will enable a more accurate understanding of the disease and help the development of targeted treatment. In this paper, we propose a Reeb graph representation of tau pathology topography on cortical surfaces using tau PET imaging data. By comparing the spatial and temporal coherence of the Reeb graph representation across subjects, we can build a directed graph to represent the distribution of tau topography over a population, which naturally facilitates the discovery of spatiotemporal subtypes of tau pathology with graph-based clustering. In our experiments, we conducted extensive comparisons with state-of-the-art event-based model on synthetic and large-scale tau PET imaging data from ADNI3 and A4 studies. We demonstrated that our proposed method can more robustly achieve the subtyping of tau pathology with clear clinical significance and demonstrated superior generalization performance than event-based model.
Collapse
Affiliation(s)
- Jiaxin Yue
- Stevens Neuroimaging and Informatics Institute, Keck School of Medicine, University of Southern California (USC), Los Angeles, CA 90033, USA
- Ming Hsieh Department of Electrical and Computer Engineering, Viterbi School of Engineering, University of Southern California (USC), Los Angeles, CA 90089, USA
| | - Yonggang Shi
- Stevens Neuroimaging and Informatics Institute, Keck School of Medicine, University of Southern California (USC), Los Angeles, CA 90033, USA
- Ming Hsieh Department of Electrical and Computer Engineering, Viterbi School of Engineering, University of Southern California (USC), Los Angeles, CA 90089, USA
| |
Collapse
|
112
|
Gouilly D, Rafiq M, Nogueira L, Salabert AS, Payoux P, Péran P, Pariente J. Beyond the amyloid cascade: An update of Alzheimer's disease pathophysiology. Rev Neurol (Paris) 2023; 179:812-830. [PMID: 36906457 DOI: 10.1016/j.neurol.2022.12.006] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Revised: 10/02/2022] [Accepted: 12/02/2022] [Indexed: 03/13/2023]
Abstract
Alzheimer's disease (AD) is a multi-etiology disease. The biological system of AD is associated with multidomain genetic, molecular, cellular, and network brain dysfunctions, interacting with central and peripheral immunity. These dysfunctions have been primarily conceptualized according to the assumption that amyloid deposition in the brain, whether from a stochastic or a genetic accident, is the upstream pathological change. However, the arborescence of AD pathological changes suggests that a single amyloid pathway might be too restrictive or inconsistent with a cascading effect. In this review, we discuss the recent human studies of late-onset AD pathophysiology in an attempt to establish a general updated view focusing on the early stages. Several factors highlight heterogenous multi-cellular pathological changes in AD, which seem to work in a self-amplifying manner with amyloid and tau pathologies. Neuroinflammation has an increasing importance as a major pathological driver, and perhaps as a convergent biological basis of aging, genetic, lifestyle and environmental risk factors.
Collapse
Affiliation(s)
- D Gouilly
- Toulouse Neuroimaging Center, Toulouse, France.
| | - M Rafiq
- Toulouse Neuroimaging Center, Toulouse, France; Department of Cognitive Neurology, Epilepsy and Movement Disorders, CHU Toulouse Purpan, France
| | - L Nogueira
- Department of Cell Biology and Cytology, CHU Toulouse Purpan, France
| | - A-S Salabert
- Toulouse Neuroimaging Center, Toulouse, France; Department of Nuclear Medicine, CHU Toulouse Purpan, France
| | - P Payoux
- Toulouse Neuroimaging Center, Toulouse, France; Department of Nuclear Medicine, CHU Toulouse Purpan, France; Center of Clinical Investigation, CHU Toulouse Purpan (CIC1436), France
| | - P Péran
- Toulouse Neuroimaging Center, Toulouse, France
| | - J Pariente
- Toulouse Neuroimaging Center, Toulouse, France; Department of Cognitive Neurology, Epilepsy and Movement Disorders, CHU Toulouse Purpan, France; Center of Clinical Investigation, CHU Toulouse Purpan (CIC1436), France
| |
Collapse
|
113
|
Badji A, Youwakim J, Cooper A, Westman E, Marseglia A. Vascular cognitive impairment - Past, present, and future challenges. Ageing Res Rev 2023; 90:102042. [PMID: 37634888 DOI: 10.1016/j.arr.2023.102042] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2023] [Revised: 08/22/2023] [Accepted: 08/23/2023] [Indexed: 08/29/2023]
Abstract
Vascular cognitive impairment (VCI) is a lifelong process encompassing a broad spectrum of cognitive disorders, ranging from subtle or mild deficits to prodromal and fully developed dementia, originating from cerebrovascular lesions such as large and small vessel disease. Genetic predisposition and environmental exposure to risk factors such as unhealthy lifestyles, hypertension, cardiovascular disease, and metabolic disorders will synergistically interact, yielding biochemical and structural brain changes, ultimately culminating in VCI. However, little is known about the pathological processes underlying VCI and the temporal dynamics between risk factors and disease mechanisms (biochemical and structural brain changes). This narrative review aims to provide an evidence-based summary of the link between individual vascular risk/disorders and cognitive dysfunction and the potential structural and biochemical pathophysiological processes. We also discuss some key challenges for future research on VCI. There is a need to shift from individual risk factors/disorders to comorbid vascular burden, identifying and integrating imaging and fluid biomarkers, implementing a life-course approach, considering possible neuroprotective influences of positive life exposures, and addressing biological sex at birth and gender differences. Finally, this review highlights the need for future researchers to leverage and integrate multidimensional data to advance our understanding of the mechanisms and pathophysiology of VCI.
Collapse
Affiliation(s)
- Atef Badji
- Division of Clinical Geriatrics, Center for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden; Theme Inflammation and Aging, Karolinska University Hospital, Stockholm, Sweden
| | - Jessica Youwakim
- Department of Pharmacology and Physiology, Université de Montréal, Montreal, QC, Canada; Centre interdisciplinaire de recherche sur le cerveau et l'apprentissage (CIRCA), Montreal, QC, Canada; Groupe de Recherche sur la Signalisation Neuronal et la Circuiterie (SNC), Montreal, QC, Canada
| | - Alexandra Cooper
- Division of Clinical Geriatrics, Center for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden; Unit of Integrative Epidemiology, Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Eric Westman
- Division of Clinical Geriatrics, Center for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden; Department of Neuroimaging, Centre for Neuroimaging Sciences, Institute of Psychiatry, Psychology, and Neuroscience, King's College London, London, UK
| | - Anna Marseglia
- Division of Clinical Geriatrics, Center for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden.
| |
Collapse
|
114
|
Devi G. A how-to guide for a precision medicine approach to the diagnosis and treatment of Alzheimer's disease. Front Aging Neurosci 2023; 15:1213968. [PMID: 37662550 PMCID: PMC10469885 DOI: 10.3389/fnagi.2023.1213968] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Accepted: 07/24/2023] [Indexed: 09/05/2023] Open
Abstract
Article purpose The clinical approach to Alzheimer's disease (AD) is challenging, particularly in high-functioning individuals. Accurate diagnosis is crucial, especially given the significant side effects, including brain hemorrhage, of newer monoclonal antibodies approved for treating earlier stages of Alzheimer's. Although early treatment is more effective, early diagnosis is also more difficult. Several clinical mimickers of AD exist either separately, or in conjunction with AD pathology, adding to the diagnostic complexity. To illustrate the clinical decision-making process, this study includes de-identified cases and reviews of the underlying etiology and pathology of Alzheimer's and available therapies to exemplify diagnostic and treatment subtleties. Problem The clinical presentation of Alzheimer's is complex and varied. Multiple other primary brain pathologies present with clinical phenotypes that can be difficult to distinguish from AD. Furthermore, Alzheimer's rarely exists in isolation, as almost all patients also show evidence of other primary brain pathologies, including Lewy body disease and argyrophilic grain disease. The phenotype and progression of AD can vary based on the brain regions affected by pathology, the coexistence and severity of other brain pathologies, the presence and severity of systemic comorbidities such as cardiac disease, the common co-occurrence with psychiatric diagnoses, and genetic risk factors. Additionally, symptoms and progression are influenced by an individual's brain reserve and cognitive reserve, as well as the timing of the diagnosis, which depends on the demographics of both the patient and the diagnosing physician, as well as the availability of biomarkers. Methods The optimal clinical and biomarker strategy for accurately diagnosing AD, common neuropathologic co-morbidities and mimickers, and available medication and non-medication-based treatments are discussed. Real-life examples of cognitive loss illustrate the diagnostic and treatment decision-making process as well as illustrative treatment responses. Implications AD is best considered a syndromic disorder, influenced by a multitude of patient and environmental characteristics. Additionally, AD existing alone is a unicorn, as there are nearly always coexisting other brain pathologies. Accurate diagnosis with biomarkers is essential. Treatment response is affected by the variables involved, and the effective treatment of Alzheimer's disease, as well as its prevention, requires an individualized, precision medicine strategy.
Collapse
Affiliation(s)
- Gayatri Devi
- Neurology and Psychiatry, Zucker School of Medicine, Hempstead, NY, United States
- Neurology and Psychiatry, Lenox Hill Hospital, New York City, NY, United States
- Park Avenue Neurology, New York City, NY, United States
| |
Collapse
|
115
|
Guebel DV. Human hippocampal astrocytes: Computational dissection of their transcriptome, sexual differences and exosomes across ageing and mild-cognitive impairment. Eur J Neurosci 2023; 58:2677-2707. [PMID: 37427765 DOI: 10.1111/ejn.16081] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2022] [Revised: 02/20/2023] [Accepted: 06/16/2023] [Indexed: 07/11/2023]
Abstract
The role of astrocytes in Alzheimer's disease is often disregarded. Hence, characterization of astrocytes along their early evolution toward Alzheimer would be greatly beneficial. However, due to their exquisite responsiveness, in vivo studies are difficult. So public microarray data of hippocampal homogenates from (healthy) young, (healthy) elder and elder with mild cognitive impairment (MCI) were subjected to re-analysis by a multi-step computational pipeline. Ontologies and pathway analyses were compared after determining the differential genes that, belonging to astrocytes, have splice forms. Likewise, the subset of molecules exportable to exosomes was also determined. The results showed that astrocyte's phenotypes changed significantly. While already 'activated' astrocytes were found in the younger group, major changes occurred during ageing (increased vascular remodelling and response to mechanical stimulus, diminished long-term potentiation and increased long-term depression). MCI's astrocytes showed some 'rejuvenated' features, but their sensitivity to shear stress was markedly lost. Importantly, most of the changes showed to be sex biassed. Men's astrocytes are enriched in a type 'endfeet-astrocytome', whereas women's astrocytes appear close to the 'scar-forming' type (prone to endothelial dysfunction, hypercholesterolemia, loss of glutamatergic synapses, Ca+2 dysregulation, hypoxia, oxidative stress and 'pro-coagulant' phenotype). In conclusion, the computational dissection of the networks based on the hippocampal gene isoforms provides a relevant proxy to in vivo astrocytes, also revealing the occurrence of sexual differences. Analyses of the astrocytic exosomes did not provide an acceptable approximation to the overall functioning of astrocytes in the hippocampus, probably due to the selective cellular mechanisms which charge the cargo molecules.
Collapse
|
116
|
Salvadó G, Horie K, Barthélemy NR, Vogel JW, Binette AP, Chen CD, Aschenbrenner AJ, Gordon BA, Benzinger TL, Holtzman DM, Morris JC, Palmqvist S, Stomrud E, Janelidze S, Ossenkoppele R, Schindler SE, Bateman RJ, Hansson O. Novel CSF tau biomarkers can be used for disease staging of sporadic Alzheimer's disease. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2023:2023.07.14.23292650. [PMID: 37503281 PMCID: PMC10370223 DOI: 10.1101/2023.07.14.23292650] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/29/2023]
Abstract
Biological staging of individuals with Alzheimer's disease (AD) may improve diagnostic and prognostic work-up of dementia in clinical practice and the design of clinical trials. Here, we created a staging model using the Subtype and Stage Inference (SuStaIn) algorithm by evaluating cerebrospinal fluid (CSF) amyloid-β (Aβ) and tau biomarkers in 426 participants from BioFINDER-2, that represent the entire spectrum of AD. The model composition and main analyses were replicated in 222 participants from the Knight ADRC cohort. SuStaIn revealed in the two cohorts that the data was best explained by a single biomarker sequence (one subtype), and that five CSF biomarkers (ordered: Aβ42/40, tau phosphorylation occupancies at the residues 217 and 205 [pT217/T217 and pT205/T205], microtubule-binding region of tau containing the residue 243 [MTBR-tau243], and total tau) were sufficient to create an accurate disease staging model. Increasing CSF stages (0-5) were associated with increased abnormality in other AD-related biomarkers, such as Aβ- and tau-PET, and aligned with different phases of longitudinal biomarker changes consistent with current models of AD progression. Higher CSF stages at baseline were associated with higher hazard ratio of clinical decline. Our findings indicate that a common pathophysiologic molecular pathway develops across all AD patients, and that a single CSF collection is sufficient to reliably indicate the presence of both AD pathologies and the degree and stage of disease progression.
Collapse
Affiliation(s)
- Gemma Salvadó
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, Lund, Sweden
| | - Kanta Horie
- The Tracy Family SILQ Center, Washington University School of Medicine, St Louis, MO, United States
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, United States
- Eisai Inc., Nutley, NJ, United States
| | - Nicolas R. Barthélemy
- The Tracy Family SILQ Center, Washington University School of Medicine, St Louis, MO, United States
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, United States
| | - Jacob W. Vogel
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, Lund, Sweden
- Department of Clinical Science, Malmö, SciLifeLab, Lund University, Lund, Sweden
| | - Alexa Pichet Binette
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, Lund, Sweden
| | - Charles D. Chen
- Department of Radiology, Washington University School of Medicine, St. Louis, MO, USA
| | - Andrew J Aschenbrenner
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, United States
- Charles F. and Joanne Knight Alzheimer Disease Research Center, Washington University School of Medicine, St. Louis, MO, USA
| | - Brian A. Gordon
- Department of Radiology, Washington University School of Medicine, St. Louis, MO, USA
| | - Tammie L.S. Benzinger
- Department of Radiology, Washington University School of Medicine, St. Louis, MO, USA
- Charles F. and Joanne Knight Alzheimer Disease Research Center, Washington University School of Medicine, St. Louis, MO, USA
| | - David M. Holtzman
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, United States
- Charles F. and Joanne Knight Alzheimer Disease Research Center, Washington University School of Medicine, St. Louis, MO, USA
| | - John C. Morris
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, United States
- Charles F. and Joanne Knight Alzheimer Disease Research Center, Washington University School of Medicine, St. Louis, MO, USA
| | - Sebastian Palmqvist
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, Lund, Sweden
| | - Erik Stomrud
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, Lund, Sweden
- Memory Clinic, Skåne University Hospital, Malmö, Sweden
| | - Shorena Janelidze
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, Lund, Sweden
| | - Rik Ossenkoppele
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, Lund, Sweden
- Alzheimer Center Amsterdam, Neurology, Vrije Universiteit Amsterdam, Amsterdam UMC location VUmc, Amsterdam, The Netherlands
- Amsterdam Neuroscience, Neurodegeneration, Amsterdam, The Netherlands
| | - Suzanne E. Schindler
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, United States
- Charles F. and Joanne Knight Alzheimer Disease Research Center, Washington University School of Medicine, St. Louis, MO, USA
| | - Randall J. Bateman
- The Tracy Family SILQ Center, Washington University School of Medicine, St Louis, MO, United States
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, United States
- Charles F. and Joanne Knight Alzheimer Disease Research Center, Washington University School of Medicine, St. Louis, MO, USA
| | - Oskar Hansson
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, Lund, Sweden
- Memory Clinic, Skåne University Hospital, Malmö, Sweden
| |
Collapse
|
117
|
Lamontagne-Kam D, Ulfat AK, Hervé V, Vu TM, Brouillette J. Implication of tau propagation on neurodegeneration in Alzheimer's disease. Front Neurosci 2023; 17:1219299. [PMID: 37483337 PMCID: PMC10360202 DOI: 10.3389/fnins.2023.1219299] [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: 05/08/2023] [Accepted: 06/07/2023] [Indexed: 07/25/2023] Open
Abstract
Propagation of tau fibrils correlate closely with neurodegeneration and memory deficits seen during the progression of Alzheimer's disease (AD). Although it is not well-established what drives or attenuates tau spreading, new studies on human brain using positron emission tomography (PET) have shed light on how tau phosphorylation, genetic factors, and the initial epicenter of tau accumulation influence tau accumulation and propagation throughout the brain. Here, we review the latest PET studies performed across the entire AD continuum looking at the impact of amyloid load on tau pathology. We also explore the effects of structural, functional, and proximity connectivity on tau spreading in a stereotypical manner in the brain of AD patients. Since tau propagation can be quite heterogenous between individuals, we then consider how the speed and pattern of propagation are influenced by the starting localization of tau accumulation in connected brain regions. We provide an overview of some genetic variants that were shown to accelerate or slow down tau spreading. Finally, we discuss how phosphorylation of certain tau epitopes affect the spreading of tau fibrils. Since tau pathology is an early event in AD pathogenesis and is one of the best predictors of neurodegeneration and memory impairments, understanding the process by which tau spread from one brain region to another could pave the way to novel therapeutic avenues that are efficient during the early stages of the disease, before neurodegeneration induces permanent brain damage and severe memory loss.
Collapse
|
118
|
Miller MI, Shih LC, Kolachalama VB. Machine Learning in Clinical Trials: A Primer with Applications to Neurology. Neurotherapeutics 2023; 20:1066-1080. [PMID: 37249836 PMCID: PMC10228463 DOI: 10.1007/s13311-023-01384-2] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/21/2023] [Indexed: 05/31/2023] Open
Abstract
We reviewed foundational concepts in artificial intelligence (AI) and machine learning (ML) and discussed ways in which these methodologies may be employed to enhance progress in clinical trials and research, with particular attention to applications in the design, conduct, and interpretation of clinical trials for neurologic diseases. We discussed ways in which ML may help to accelerate the pace of subject recruitment, provide realistic simulation of medical interventions, and enhance remote trial administration via novel digital biomarkers and therapeutics. Lastly, we provide a brief overview of the technical, administrative, and regulatory challenges that must be addressed as ML achieves greater integration into clinical trial workflows.
Collapse
Affiliation(s)
- Matthew I Miller
- Department of Medicine, Boston University Chobanian & Avedisian School of Medicine, 72 E. Concord Street, Evans 636, Boston, MA, 02118, USA
| | - Ludy C Shih
- Department of Neurology, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, 02118, USA
| | - Vijaya B Kolachalama
- Department of Medicine, Boston University Chobanian & Avedisian School of Medicine, 72 E. Concord Street, Evans 636, Boston, MA, 02118, USA.
- Department of Computer Science and Faculty of Computing & Data Sciences, Boston University, Boston, MA, 02115, USA.
| |
Collapse
|
119
|
Villemagne VL, Leuzy A, Bohorquez SS, Bullich S, Shimada H, Rowe CC, Bourgeat P, Lopresti B, Huang K, Krishnadas N, Fripp J, Takado Y, Gogola A, Minhas D, Weimer R, Higuchi M, Stephens A, Hansson O, Doré V. CenTauR: Toward a universal scale and masks for standardizing tau imaging studies. ALZHEIMER'S & DEMENTIA (AMSTERDAM, NETHERLANDS) 2023; 15:e12454. [PMID: 37424964 PMCID: PMC10326476 DOI: 10.1002/dad2.12454] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Revised: 05/26/2023] [Accepted: 05/26/2023] [Indexed: 07/11/2023]
Abstract
INTRODUCTION Recently, an increasing number of tau tracers have become available. There is a need to standardize quantitative tau measures across tracers, supporting a universal scale. We developed several cortical tau masks and applied them to generate a tau imaging universal scale. METHOD One thousand forty-five participants underwent tau scans with either 18F-flortaucipir, 18F-MK6240, 18F-PI2620, 18F-PM-PBB3, 18F-GTP1, or 18F-RO948. The universal mask was generated from cognitively unimpaired amyloid beta (Aβ)- subjects and Alzheimer's disease (AD) patients with Aβ+. Four additional regional cortical masks were defined within the constraints of the universal mask. A universal scale, the CenTauRz, was constructed. RESULTS None of the regions known to display off-target signal were included in the masks. The CenTauRz allows robust discrimination between low and high levels of tau deposits. DISCUSSION We constructed several tau-specific cortical masks for the AD continuum and a universal standard scale designed to capture the location and degree of abnormality that can be applied across tracers and across centers. The masks are freely available at https://www.gaain.org/centaur-project.
Collapse
Affiliation(s)
- Victor L. Villemagne
- Department of PsychiatryUniversity of PittsburghPittsburghPennsylvaniaUSA
- Department of Molecular Imaging & TherapyAustin HealthMelbourneVictoriaAustralia
| | - Antoine Leuzy
- Clinical Memory Research UnitDepartment of Clinical SciencesLund UniversityMalmöSweden
| | | | | | - Hitoshi Shimada
- Department of Functional Brain ImagingNational Institutes for Quantum and Radiological Science and TechnologyChibaJapan
- Brain Research InstituteNiigata UniversityNiigataJapan
| | - Christopher C. Rowe
- Department of Molecular Imaging & TherapyAustin HealthMelbourneVictoriaAustralia
- Florey Department of Neurosciences & Mental HealthThe University of MelbourneMelbourneParkvilleAustralia
- The Australian Dementia Network (ADNeT)MelbourneVictoriaAustralia
| | - Pierrick Bourgeat
- Health and Biosecurity FlagshipThe Australian eHealth Research CentreCSIROBrisbaneQueenslandAustralia
| | - Brian Lopresti
- Department of RadiologyUniversity of PittsburghPittsburghPennsylvaniaUSA
| | - Kun Huang
- Department of Molecular Imaging & TherapyAustin HealthMelbourneVictoriaAustralia
| | - Natasha Krishnadas
- Department of Molecular Imaging & TherapyAustin HealthMelbourneVictoriaAustralia
- Florey Institute of Neurosciences & Mental HealthParkvilleVictoriaAustralia
| | - Jurgen Fripp
- Health and Biosecurity FlagshipThe Australian eHealth Research CentreCSIROBrisbaneQueenslandAustralia
| | - Yuhei Takado
- Department of Functional Brain ImagingNational Institutes for Quantum and Radiological Science and TechnologyChibaJapan
| | - Alexandra Gogola
- Department of RadiologyUniversity of PittsburghPittsburghPennsylvaniaUSA
| | - Davneet Minhas
- Department of RadiologyUniversity of PittsburghPittsburghPennsylvaniaUSA
| | | | - Makoto Higuchi
- Department of Functional Brain ImagingNational Institutes for Quantum and Radiological Science and TechnologyChibaJapan
| | | | - Oskar Hansson
- Clinical Memory Research UnitDepartment of Clinical SciencesLund UniversityMalmöSweden
- Memory ClinicSkåne University HospitalMalmöSweden
| | - Vincent Doré
- Department of Molecular Imaging & TherapyAustin HealthMelbourneVictoriaAustralia
- Health and Biosecurity FlagshipThe Australian eHealth Research CentreCSIROHeidelbergVictoriaAustralia
| | | |
Collapse
|
120
|
Daghlas I, Nassan M, Gill D. Genetically proxied lean mass and risk of Alzheimer's disease: mendelian randomisation study. BMJ MEDICINE 2023; 2:e000354. [PMID: 37564828 PMCID: PMC10410880 DOI: 10.1136/bmjmed-2022-000354] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/24/2022] [Accepted: 04/05/2023] [Indexed: 08/12/2023]
Abstract
Objective To examine whether genetically proxied lean mass is associated with risk of Alzheimer's disease. Design Mendelian randomisation study. Setting The UK Biobank study and genome wide association study meta-analyses of Alzheimer's disease and cognitive performance. Participants Summary level genetic data from: 450 243 UK Biobank participants with impedance measures of lean mass and fat mass; an independent sample of 21 982 patients with Alzheimer's disease and 41 944 controls without Alzheimer's disease; a replication sample of 7329 patients with Alzheimer's disease and 252 879 controls; and 269 867 individuals taking part in a genome wide association study of cognitive performance. Main outcome measure Effect of genetically proxied lean mass on the risk of Alzheimer's disease, and the related phenotype of cognitive performance. Results An increase in genetically proxied appendicular lean mass of one standard deviation was associated with a 12% reduced risk of Alzheimer's disease (odds ratio 0.88, 95% confidence interval 0.82 to 0.95, P=0.001). This finding was replicated in an independent cohort of patients with Alzheimer's disease (0.91, 0.83 to 0.99, P=0.02) and was consistent in sensitivity analyses that are more robust to the inclusion of pleiotropic variants. Higher genetically proxied appendicular lean mass was also associated with increased cognitive performance (standard deviation increase in cognitive performance for each standard deviation increase in appendicular lean mass 0.09, 95% confidence interval 0.06 to 0.11, P=0.001), and adjusting for potential mediation through genetically proxied cognitive performance did not reduce the association between appendicular lean mass and risk of Alzheimer's disease. Similar results were found for the outcomes of Alzheimer's disease and cognitive performance when the risk factors of genetically proxied trunk lean mass and whole body lean mass were used, respectively, adjusted for genetically proxied fat mass. Conclusions These findings suggest that lean mass might be a possible modifiable protective factor for Alzheimer's disease. The mechanisms underlying this finding, as well as the clinical and public health implications, warrant further investigation.
Collapse
Affiliation(s)
- Iyas Daghlas
- Department of Neurology, University of California San Francisco, San Francisco, California, USA
| | - Malik Nassan
- Mesulam Center for Cognitive Neurology and Alzheimer’s Disease, Northwestern University, Chicago, Illinois, USA
| | - Dipender Gill
- Department of Epidemiology and Biostatistics, Imperial College London, London, UK
- Chief Scientific Advisor Office, Research and Early Development, Novo Nordisk, Copenhagen, Denmark
| |
Collapse
|
121
|
Shigemizu D, Akiyama S, Suganuma M, Furutani M, Yamakawa A, Nakano Y, Ozaki K, Niida S. Classification and deep-learning-based prediction of Alzheimer disease subtypes by using genomic data. Transl Psychiatry 2023; 13:232. [PMID: 37386009 DOI: 10.1038/s41398-023-02531-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/02/2023] [Revised: 06/16/2023] [Accepted: 06/19/2023] [Indexed: 07/01/2023] Open
Abstract
Late-onset Alzheimer's disease (LOAD) is the most common multifactorial neurodegenerative disease among elderly people. LOAD is heterogeneous, and the symptoms vary among patients. Genome-wide association studies (GWAS) have identified genetic risk factors for LOAD but not for LOAD subtypes. Here, we examined the genetic architecture of LOAD based on Japanese GWAS data from 1947 patients and 2192 cognitively normal controls in a discovery cohort and 847 patients and 2298 controls in an independent validation cohort. Two distinct groups of LOAD patients were identified. One was characterized by major risk genes for developing LOAD (APOC1 and APOC1P1) and immune-related genes (RELB and CBLC). The other was characterized by genes associated with kidney disorders (AXDND1, FBP1, and MIR2278). Subsequent analysis of albumin and hemoglobin values from routine blood test results suggested that impaired kidney function could lead to LOAD pathogenesis. We developed a prediction model for LOAD subtypes using a deep neural network, which achieved an accuracy of 0.694 (2870/4137) in the discovery cohort and 0.687 (2162/3145) in the validation cohort. These findings provide new insights into the pathogenic mechanisms of LOAD.
Collapse
Affiliation(s)
- Daichi Shigemizu
- Medical Genome Center, Research Institute, National Center for Geriatrics and Gerontology, Obu, Aichi, 474-8511, Japan.
- RIKEN Center for Integrative Medical Sciences, Yokohama, Kanagawa, 230-0045, Japan.
| | - Shintaro Akiyama
- Medical Genome Center, Research Institute, National Center for Geriatrics and Gerontology, Obu, Aichi, 474-8511, Japan
| | - Mutsumi Suganuma
- Medical Genome Center, Research Institute, National Center for Geriatrics and Gerontology, Obu, Aichi, 474-8511, Japan
| | - Motoki Furutani
- Medical Genome Center, Research Institute, National Center for Geriatrics and Gerontology, Obu, Aichi, 474-8511, Japan
- Department of Cardiovascular Medicine, Hiroshima University Graduate School of Biomedical and Health Sciences, Hiroshima, 734-8553, Japan
| | - Akiko Yamakawa
- Medical Genome Center, Research Institute, National Center for Geriatrics and Gerontology, Obu, Aichi, 474-8511, Japan
| | - Yukiko Nakano
- Department of Cardiovascular Medicine, Hiroshima University Graduate School of Biomedical and Health Sciences, Hiroshima, 734-8553, Japan
| | - Kouichi Ozaki
- Medical Genome Center, Research Institute, National Center for Geriatrics and Gerontology, Obu, Aichi, 474-8511, Japan
- RIKEN Center for Integrative Medical Sciences, Yokohama, Kanagawa, 230-0045, Japan
- Department of Cardiovascular Medicine, Hiroshima University Graduate School of Biomedical and Health Sciences, Hiroshima, 734-8553, Japan
| | - Shumpei Niida
- Core Facility Administration, Research Institute, National Center for Geriatrics and Gerontology, Obu, Aichi, 474-8511, Japan
| |
Collapse
|
122
|
Diaz-Galvan P, Lorenzon G, Mohanty R, Mårtensson G, Cavedo E, Lista S, Vergallo A, Kantarci K, Hampel H, Dubois B, Grothe MJ, Ferreira D, Westman E. Differential response to donepezil in MRI subtypes of mild cognitive impairment. Alzheimers Res Ther 2023; 15:117. [PMID: 37353809 PMCID: PMC10288762 DOI: 10.1186/s13195-023-01253-2] [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: 09/02/2022] [Accepted: 06/01/2023] [Indexed: 06/25/2023]
Abstract
BACKGROUND Donepezil is an approved therapy for the treatment of Alzheimer's disease (AD). Results across clinical trials have been inconsistent, which may be explained by design-methodological issues, the pathophysiological heterogeneity of AD, and diversity of included study participants. We investigated whether response to donepezil differs in mild cognitive impaired (MCI) individuals demonstrating different magnetic resonance imaging (MRI) subtypes. METHODS From the Hippocampus Study double-blind, randomized clinical trial, we included 173 MCI individuals (donepezil = 83; placebo = 90) with structural MRI data, at baseline and at clinical follow-up assessments (6-12-month). Efficacy outcomes were the annualized percentage change (APC) in hippocampal, ventricular, and total grey matter volumes, as well as in the AD cortical thickness signature. Participants were classified into MRI subtypes as typical AD, limbic-predominant, hippocampal-sparing, or minimal atrophy at baseline. We primarily applied a subtyping approach based on continuous scale of two subtyping dimensions. We also used the conventional categorical subtyping approach for comparison. RESULTS Donepezil-treated MCI individuals showed slower atrophy rates compared to the placebo group, but only if they belonged to the minimal atrophy or hippocampal-sparing subtypes. Importantly, only the continuous subtyping approach, but not the conventional categorical approach, captured this differential response. CONCLUSIONS Our data suggest that individuals with MCI, with hippocampal-sparing or minimal atrophy subtype, may have improved benefit from donepezil, as compared with MCI individuals with typical or limbic-predominant patterns of atrophy. The newly proposed continuous subtyping approach may have advantages compared to the conventional categorical approach. Future research is warranted to demonstrate the potential of subtype stratification for disease prognosis and response to treatment. TRIAL REGISTRATION ClinicalTrial.gov NCT00403520. Submission Date: November 21, 2006.
Collapse
Affiliation(s)
| | - Giulia Lorenzon
- Division of Clinical Geriatrics, Center for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Huddinge, Sweden
| | - Rosaleena Mohanty
- Division of Clinical Geriatrics, Center for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Huddinge, Sweden
| | - Gustav Mårtensson
- Division of Clinical Geriatrics, Center for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Huddinge, Sweden
| | - Enrica Cavedo
- Alzheimer Precision Medicine (APM), Sorbonne University, AP-HP, Pitié-Salpêtrière Hospital, Boulevard de L'hôpital, Paris, France
| | - Simone Lista
- Alzheimer Precision Medicine (APM), Sorbonne University, AP-HP, Pitié-Salpêtrière Hospital, Boulevard de L'hôpital, Paris, France
| | - Andrea Vergallo
- Alzheimer Precision Medicine (APM), Sorbonne University, AP-HP, Pitié-Salpêtrière Hospital, Boulevard de L'hôpital, Paris, France
| | - Kejal Kantarci
- Department of Radiology, Mayo Clinic, Rochester, MN, USA
| | - Harald Hampel
- Alzheimer Precision Medicine (APM), Sorbonne University, AP-HP, Pitié-Salpêtrière Hospital, Boulevard de L'hôpital, Paris, France
| | - Bruno Dubois
- Alzheimer Precision Medicine (APM), Sorbonne University, AP-HP, Pitié-Salpêtrière Hospital, Boulevard de L'hôpital, Paris, France
| | - 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, Sevilla, Spain
- Wallenberg Center for Molecular and Translational Medicine, Department of Psychiatry and Neurochemistry, University of Gothenburg, Gothenburg, Sweden
| | - Daniel Ferreira
- Department of Radiology, Mayo Clinic, Rochester, MN, USA
- Division of Clinical Geriatrics, Center for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Huddinge, Sweden
| | - Eric Westman
- Division of Clinical Geriatrics, Center for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Huddinge, Sweden.
- Department of Neuroimaging, Centre for Neuroimaging Sciences, Institute of Psychiatry, Psychology, and Neuroscience, King's College London, London, UK.
| |
Collapse
|
123
|
Chiari-Correia RD, Tumas V, Santos AC, Salmon CEG. Structural and functional differences in the brains of patients with MCI with and without depressive symptoms and their relations with Alzheimer's disease: an MRI study. PSYCHORADIOLOGY 2023; 3:kkad008. [PMID: 38666129 PMCID: PMC10917365 DOI: 10.1093/psyrad/kkad008] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/17/2023] [Revised: 05/19/2023] [Accepted: 06/12/2023] [Indexed: 04/28/2024]
Abstract
Background The mild cognitive impairment (MCI) stage among elderly individuals is very complex, and the level of diagnostic accuracy is far from ideal. Some studies have tried to improve the 'MCI due to Alzheimer's disease (AD)' classification by further stratifying these patients into subgroups. Depression-related symptoms may play an important role in helping to better define the MCI stage in elderly individuals. Objective In this work, we explored functional and structural differences in the brains of patients with nondepressed MCI (nDMCI) and patients with MCI with depressive symptoms (DMCI), and we examined how these groups relate to AD atrophy patterns and cognitive functioning. Methods Sixty-five participants underwent MRI exams and were divided into four groups: cognitively normal, nDMCI, DMCI, and AD. We compared the regional brain volumes, cortical thickness, and white matter microstructure measures using diffusion tensor imaging among groups. Additionally, we evaluated changes in functional connectivity using fMRI data. Results In comparison to the nDMCI group, the DMCI patients had more pronounced atrophy in the hippocampus and amygdala. Additionally, DMCI patients had asymmetric damage in the limbic-frontal white matter connection. Furthermore, two medial posterior regions, the isthmus of cingulate gyrus and especially the lingual gyrus, had high importance in the structural and functional differentiation between the two groups. Conclusion It is possible to differentiate nDMCI from DMCI patients using MRI techniques, which may contribute to a better characterization of subtypes of the MCI stage.
Collapse
Affiliation(s)
- Rodolfo Dias Chiari-Correia
- Department of Neurosciences and Behavioral Sciences, Ribeirao Preto Medical School, University of Sao Paulo, 3900 Bandeirantes Avenue, Ribeirao Preto SP, 14040-900, Brazil
| | - Vitor Tumas
- Department of Neurosciences and Behavioral Sciences, Ribeirao Preto Medical School, University of Sao Paulo, 3900 Bandeirantes Avenue, Ribeirao Preto SP, 14040-900, Brazil
| | - Antônio Carlos Santos
- Department of Medical Imaging, Hematology and Clinical Oncology, Ribeirao Preto Medical School, University of Sao Paulo, 3900 Bandeirantes Avenue, Ribeirao Preto SP, 14040-900, Brazil
| | - Carlos Ernesto G Salmon
- Department of Physics, Faculty of Philosophy, Sciences and Letters, University of Sao Paulo, 3900 Bandeirantes Avenue, Ribeirao Preto SP, 14040-900, Brazil
| |
Collapse
|
124
|
He XY, Wu BS, Kuo K, Zhang W, Ma Q, Xiang ST, Li YZ, Wang ZY, Dong Q, Feng JF, Cheng W, Yu JT. Association between polygenic risk for Alzheimer's disease and brain structure in children and adults. Alzheimers Res Ther 2023; 15:109. [PMID: 37312172 DOI: 10.1186/s13195-023-01256-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Accepted: 06/01/2023] [Indexed: 06/15/2023]
Abstract
BACKGROUND The correlations between genetic risk for Alzheimer's disease (AD) with comprehensive brain regions at a regional scale are still not well understood. We aim to explore whether these associations vary across different age stages. METHODS This study used large existing genome-wide association datasets to calculate polygenic risk score (PRS) for AD in two populations from the UK Biobank (N ~ 23 000) and Adolescent Brain Cognitive Development Study (N ~ 4660) who had multimodal macrostructural and microstructural magnetic resonance imaging (MRI) metrics. We used linear mixed-effect models to assess the strength of the association between AD PRS and multiple MRI metrics of regional brain structures at different stages of life. RESULTS Compared to those with lower PRSs, adolescents with higher PRSs had thinner cortex in the caudal anterior cingulate and supramarginal. In the middle-aged and elderly population, AD PRS had correlations with regional structure shrink primarily located in the cingulate, prefrontal cortex, hippocampus, thalamus, amygdala, and striatum, whereas the brain expansion was concentrated near the occipital lobe. Furthermore, both adults and adolescents with higher PRSs exhibited widespread white matter microstructural changes, indicated by decreased fractional anisotropy (FA) or increased mean diffusivity (MD). CONCLUSIONS In conclusion, our results suggest genetic loading for AD may influence brain structures in a highly dynamic manner, with dramatically different patterns at different ages. This age-specific change is consistent with the classical pattern of brain impairment observed in AD patients.
Collapse
Affiliation(s)
- Xiao-Yu He
- Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, National Center for Neurological Disorders, Fudan University, Shanghai, China
| | - Bang-Sheng Wu
- Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, National Center for Neurological Disorders, Fudan University, Shanghai, China
| | - Kevin Kuo
- Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, National Center for Neurological Disorders, Fudan University, Shanghai, China
| | - Wei Zhang
- Institute of Science and Technology for Brain-Inspired Intelligence, Fudan University, Shanghai, China
| | - Qing Ma
- Institute of Science and Technology for Brain-Inspired Intelligence, Fudan University, Shanghai, China
- MOE Frontiers Center for Brain Science, Fudan University, Shanghai, China
| | - Shi-Tong Xiang
- Institute of Science and Technology for Brain-Inspired Intelligence, Fudan University, Shanghai, China
| | - Yu-Zhu Li
- Institute of Science and Technology for Brain-Inspired Intelligence, Fudan University, Shanghai, China
| | - Zi-Yi Wang
- Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, National Center for Neurological Disorders, Fudan University, Shanghai, China
| | - Qiang Dong
- Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, National Center for Neurological Disorders, Fudan University, Shanghai, China
| | - Jian-Feng Feng
- Institute of Science and Technology for Brain-Inspired Intelligence, Fudan University, Shanghai, China
- MOE Frontiers Center for Brain Science, Fudan University, Shanghai, China
- Key Laboratory of Computational Neuroscience and Brain-Inspired Intelligence, Ministry of Education, Fudan University, Shanghai, China
- ISTBI-ZJNU Algorithm Centre for Brain-Inspired Intelligence, Zhejiang Normal University, Jinhua, China
- Zhangjiang Fudan International Innovation Center, Shanghai, China
| | - Wei Cheng
- Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, National Center for Neurological Disorders, Fudan University, Shanghai, China.
- Institute of Science and Technology for Brain-Inspired Intelligence, Fudan University, Shanghai, China.
- Key Laboratory of Computational Neuroscience and Brain-Inspired Intelligence, Ministry of Education, Fudan University, Shanghai, China.
- ISTBI-ZJNU Algorithm Centre for Brain-Inspired Intelligence, Zhejiang Normal University, Jinhua, China.
| | - Jin-Tai Yu
- Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, National Center for Neurological Disorders, Fudan University, Shanghai, China.
| |
Collapse
|
125
|
St-Onge F, Chapleau M, Breitner JCS, Villeneuve S, Binette AP. Tau accumulation and its spatial progression across the Alzheimer's disease spectrum. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2023:2023.06.02.23290880. [PMID: 37333413 PMCID: PMC10274981 DOI: 10.1101/2023.06.02.23290880] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/20/2023]
Abstract
The spread of tau abnormality in sporadic Alzheimer's disease is believed typically to follow neuropathologically defined Braak staging. Recent in-vivo positron emission tomography (PET) evidence challenges this belief, however, as spreading patterns for tau appear heterogenous among individuals with varying clinical expression of Alzheimer's disease. We therefore sought better understanding of the spatial distribution of tau in the preclinical and clinical phases of sporadic Alzheimer's disease and its association with cognitive decline. Longitudinal tau-PET data (1,370 scans) from 832 participants (463 cognitively unimpaired, 277 with mild cognitive impairment (MCI) and 92 with Alzheimer's disease dementia) were obtained from the Alzheimer's Disease Neuroimaging Initiative. Among these, we defined thresholds of abnormal tau deposition in 70 brain regions from the Desikan atlas, and for each group of regions characteristic of Braak staging. We summed each scan's number of regions with abnormal tau deposition to form a spatial extent index. We then examined patterns of tau pathology cross-sectionally and longitudinally and assessed their heterogeneity. Finally, we compared our spatial extent index of tau uptake with a temporal meta region of interest-a commonly used proxy of tau burden-assessing their association with cognitive scores and clinical progression. More than 80% of amyloid-beta positive participants across diagnostic groups followed typical Braak staging, both cross-sectionally and longitudinally. Within each Braak stage, however, the pattern of abnormality demonstrated significant heterogeneity such that overlap of abnormal regions across participants averaged less than 50%. The annual rate of change in number of abnormal tau-PET regions was similar among individuals without cognitive impairment and those with Alzheimer's disease dementia. Spread of disease progressed more rapidly, however, among participants with MCI. The latter's change on our spatial extent measure amounted to 2.5 newly abnormal regions per year, as contrasted with 1 region/year among the other groups. Comparing the association of tau pathology and cognitive performance in MCI and Alzheimer's disease dementia, our spatial extent index was superior to the temporal meta-ROI for measures of executive function. Thus, while participants broadly followed Braak stages, significant individual regional heterogeneity of tau binding was observed at each clinical stage. Progression of spatial extent of tau pathology appears to be fastest in persons with MCI. Exploring the spatial distribution of tau deposits throughout the entire brain may uncover further pathological variations and their correlation with impairments in cognitive functions beyond memory.
Collapse
Affiliation(s)
- Frédéric St-Onge
- Integrated Program in Neuroscience, Faculty of medicine, McGill University, Montreal, Qc, H3A 2B4, Canada
- Research Center of the Douglas Mental Health University Institute, Montreal, Qc, H4H 1R3, Canada
| | - Marianne Chapleau
- Faculty of medicine, University of California San Francisco, San Francisco, CA, 94143, United-States
| | - John CS Breitner
- Research Center of the Douglas Mental Health University Institute, Montreal, Qc, H4H 1R3, Canada
- Department of psychiatry, Faculty of medicine, McGill University, Montreal, QC, H3A 1Y2, Canada
| | - Sylvia Villeneuve
- Research Center of the Douglas Mental Health University Institute, Montreal, Qc, H4H 1R3, Canada
- Department of psychiatry, Faculty of medicine, McGill University, Montreal, QC, H3A 1Y2, Canada
- McConnell Brain Imaging Centre, Montreal Neurological Institute, Montreal, QC, H3A 2B4, Canada
| | - Alexa Pichet Binette
- Clinical Memory Research Unit, Faculty of Medicine, Lund University, Malmö, 205 02, Sweden
| |
Collapse
|
126
|
Avelar-Pereira B, Belloy ME, O'Hara R, Hosseini SMH. Decoding the heterogeneity of Alzheimer's disease diagnosis and progression using multilayer networks. Mol Psychiatry 2023; 28:2423-2432. [PMID: 36539525 PMCID: PMC10279806 DOI: 10.1038/s41380-022-01886-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Revised: 09/19/2022] [Accepted: 11/10/2022] [Indexed: 12/24/2022]
Abstract
Alzheimer's disease (AD) is a multifactorial and heterogeneous disorder, which makes early detection a challenge. Studies have attempted to combine biomarkers to improve AD detection and predict progression. However, most of the existing work reports results in parallel or compares normalized findings but does not analyze data simultaneously. We tested a multi-dimensional network framework, applied to 490 subjects (cognitively normal [CN] = 147; mild cognitive impairment [MCI] = 287; AD = 56) from ADNI, to create a single model capable of capturing the heterogeneity and progression of AD. First, we constructed subject similarity networks for structural magnetic resonance imaging, amyloid-β positron emission tomography, cerebrospinal fluid, cognition, and genetics data and then applied multilayer community detection to find groups with shared similarities across modalities. Individuals were also followed-up longitudinally, with AD subjects having, on average, 4.5 years of follow-up. Our findings show that multilayer community detection allows for accurate identification of present and future AD (≈90%) and is also able to identify cases that were misdiagnosed clinically. From all MCI participants who developed AD or reverted to CN, the multilayer model correctly identified 90.8% and 88.5% of cases respectively. We observed similar subtypes across the full sample and when examining multimodal data from subjects with no AD pathology (i.e., amyloid negative). Finally, these results were also validated using an independent testing set. In summary, the multilayer framework is successful in detecting AD and provides unique insight into the heterogeneity of the disease by identifying subtypes that share similar multidisciplinary profiles of neurological, cognitive, pathological, and genetics information.
Collapse
Affiliation(s)
- Bárbara Avelar-Pereira
- Department of Psychiatry and Behavioral Sciences, School of Medicine, Stanford University, Stanford, CA, 94304, USA.
| | - Michael E Belloy
- Department of Neurology and Neurological Sciences, School of Medicine, Stanford University, Stanford, CA, 94304, USA
| | - Ruth O'Hara
- Department of Psychiatry and Behavioral Sciences, School of Medicine, Stanford University, Stanford, CA, 94304, USA
| | - S M Hadi Hosseini
- Department of Psychiatry and Behavioral Sciences, School of Medicine, Stanford University, Stanford, CA, 94304, USA.
| |
Collapse
|
127
|
Corriveau-Lecavalier N, Barnard LR, Lee J, Dicks E, Botha H, Graff-Radford J, Machulda MM, Boeve BF, Knopman DS, Lowe VJ, Petersen RC, Jack, Jr CR, Jones DT. Deciphering the clinico-radiological heterogeneity of dysexecutive Alzheimer's disease. Cereb Cortex 2023; 33:7026-7043. [PMID: 36721911 PMCID: PMC10233237 DOI: 10.1093/cercor/bhad017] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2022] [Revised: 12/24/2022] [Accepted: 01/13/2023] [Indexed: 02/02/2023] Open
Abstract
Dysexecutive Alzheimer's disease (dAD) manifests as a progressive dysexecutive syndrome without prominent behavioral features, and previous studies suggest clinico-radiological heterogeneity within this syndrome. We uncovered this heterogeneity using unsupervised machine learning in 52 dAD patients with multimodal imaging and cognitive data. A spectral decomposition of covariance between FDG-PET images yielded six latent factors ("eigenbrains") accounting for 48% of variance in patterns of hypometabolism. These eigenbrains differentially related to age at onset, clinical severity, and cognitive performance. A hierarchical clustering on the eigenvalues of these eigenbrains yielded four dAD subtypes, i.e. "left-dominant," "right-dominant," "bi-parietal-dominant," and "heteromodal-diffuse." Patterns of FDG-PET hypometabolism overlapped with those of tau-PET distribution and MRI neurodegeneration for each subtype, whereas patterns of amyloid deposition were similar across subtypes. Subtypes differed in age at onset and clinical severity where the heteromodal-diffuse exhibited a worse clinical picture, and the bi-parietal had a milder clinical presentation. We propose a conceptual framework of executive components based on the clinico-radiological associations observed in dAD. We demonstrate that patients with dAD, despite sharing core clinical features, are diagnosed with variability in their clinical and neuroimaging profiles. Our findings support the use of data-driven approaches to delineate brain-behavior relationships relevant to clinical practice and disease physiology.
Collapse
Affiliation(s)
| | | | - Jeyeon Lee
- Department of Radiology, Mayo Clinic, Rochester, MN 55905, USA
| | - Ellen Dicks
- Department of Neurology, Mayo Clinic, Rochester, MN 55905, USA
| | - Hugo Botha
- Department of Neurology, Mayo Clinic, Rochester, MN 55905, USA
| | | | - Mary M Machulda
- Department of Psychiatry and Psychology, Mayo Clinic, Rochester, MN 55905, USA
| | - Bradley F Boeve
- Department of Neurology, Mayo Clinic, Rochester, MN 55905, USA
| | - David S Knopman
- Department of Neurology, Mayo Clinic, Rochester, MN 55905, USA
| | - Val J Lowe
- Department of Radiology, Mayo Clinic, Rochester, MN 55905, USA
| | | | | | - David T Jones
- Department of Neurology, Mayo Clinic, Rochester, MN 55905, USA
- Department of Radiology, Mayo Clinic, Rochester, MN 55905, USA
| |
Collapse
|
128
|
Dronse J, Ohndorf A, Richter N, Bischof GN, Fassbender R, Behfar Q, Gramespacher H, Dillen K, Jacobs HIL, Kukolja J, Fink GR, Onur OA. Serum cortisol is negatively related to hippocampal volume, brain structure, and memory performance in healthy aging and Alzheimer's disease. Front Aging Neurosci 2023; 15:1154112. [PMID: 37251803 PMCID: PMC10213232 DOI: 10.3389/fnagi.2023.1154112] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Accepted: 04/19/2023] [Indexed: 05/31/2023] Open
Abstract
Objective Elevated cortisol levels have been frequently reported in Alzheimer's disease (AD) and linked to brain atrophy, especially of the hippocampus. Besides, high cortisol levels have been shown to impair memory performance and increase the risk of developing AD in healthy individuals. We investigated the associations between serum cortisol levels, hippocampal volume, gray matter volume and memory performance in healthy aging and AD. Methods In our cross-sectional study, we analyzed the relationships between morning serum cortisol levels, verbal memory performance, hippocampal volume, and whole-brain voxel-wise gray matter volume in an independent sample of 29 healthy seniors (HS) and 29 patients along the spectrum of biomarker-based AD. Results Cortisol levels were significantly elevated in patients with AD as compared to HS, and higher cortisol levels were correlated with worse memory performance in AD. Furthermore, higher cortisol levels were significantly associated with smaller left hippocampal volumes in HS and indirectly negatively correlated to memory function through hippocampal volume. Higher cortisol levels were further related to lower gray matter volume in the hippocampus and temporal and parietal areas in the left hemisphere in both groups. The strength of this association was similar in HS and AD. Conclusion In AD, cortisol levels are elevated and associated with worse memory performance. Furthermore, in healthy seniors, higher cortisol levels show a detrimental relationship with brain regions typically affected by AD. Thus, increased cortisol levels seem to be indirectly linked to worse memory function even in otherwise healthy individuals. Cortisol may therefore not only serve as a biomarker of increased risk for AD, but maybe even more importantly, as an early target for preventive and therapeutic interventions.
Collapse
Affiliation(s)
- Julian Dronse
- Cognitive Neuroscience, Institute of Neuroscience and Medicine (INM-3), Jülich Research Centre, Jülich, Germany
- Department of Neurology, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Anna Ohndorf
- Department of Neurology, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Nils Richter
- Cognitive Neuroscience, Institute of Neuroscience and Medicine (INM-3), Jülich Research Centre, Jülich, Germany
- Department of Neurology, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Gérard N. Bischof
- Department of Nuclear Medicine, Multimodal Neuroimaging Group, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Ronja Fassbender
- Cognitive Neuroscience, Institute of Neuroscience and Medicine (INM-3), Jülich Research Centre, Jülich, Germany
- Department of Neurology, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Qumars Behfar
- Cognitive Neuroscience, Institute of Neuroscience and Medicine (INM-3), Jülich Research Centre, Jülich, Germany
- Department of Neurology, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Hannes Gramespacher
- Department of Neurology, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Kim Dillen
- Cognitive Neuroscience, Institute of Neuroscience and Medicine (INM-3), Jülich Research Centre, Jülich, Germany
- Department of Palliative Medicine, Multimodal Neuroimaging Group, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Heidi I. L. Jacobs
- Department of Radiology, Gordon Center for Medical Imaging, Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States
- School for Mental Health and Neuroscience, Alzheimer Centre Limburg, Maastricht University, Maastricht, Netherlands
| | - Juraj Kukolja
- Department of Neurology and Clinical Neurophysiology, Helios University Hospital Wuppertal, Wuppertal, Germany
- Faculty of Health Witten/Herdecke University, Witten, Germany
| | - Gereon R. Fink
- Cognitive Neuroscience, Institute of Neuroscience and Medicine (INM-3), Jülich Research Centre, Jülich, Germany
- Department of Neurology, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Oezguer A. Onur
- Cognitive Neuroscience, Institute of Neuroscience and Medicine (INM-3), Jülich Research Centre, Jülich, Germany
- Department of Neurology, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| |
Collapse
|
129
|
Abbate C. The Adult Neurogenesis Theory of Alzheimer's Disease. J Alzheimers Dis 2023:JAD221279. [PMID: 37182879 DOI: 10.3233/jad-221279] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/16/2023]
Abstract
Alzheimer's disease starts in neural stem cells (NSCs) in the niches of adult neurogenesis. All primary factors responsible for pathological tau hyperphosphorylation are inherent to adult neurogenesis and migration. However, when amyloid pathology is present, it strongly amplifies tau pathogenesis. Indeed, the progressive accumulation of extracellular amyloid-β deposits in the brain triggers a state of chronic inflammation by microglia. Microglial activation has a significant pro-neurogenic effect that fosters the process of adult neurogenesis and supports neuronal migration. Unfortunately, this "reactive" pro-neurogenic activity ultimately perturbs homeostatic equilibrium in the niches of adult neurogenesis by amplifying tau pathogenesis in AD. This scenario involves NSCs in the subgranular zone of the hippocampal dentate gyrus in late-onset AD (LOAD) and NSCs in the ventricular-subventricular zone along the lateral ventricles in early-onset AD (EOAD), including familial AD (FAD). Neuroblasts carrying the initial seed of tau pathology travel throughout the brain via neuronal migration driven by complex signals and convey the disease from the niches of adult neurogenesis to near (LOAD) or distant (EOAD) brain regions. In these locations, or in close proximity, a focus of degeneration begins to develop. Then, tau pathology spreads from the initial foci to large neuronal networks along neural connections through neuron-to-neuron transmission.
Collapse
Affiliation(s)
- Carlo Abbate
- IRCCS Fondazione Don Carlo Gnocchi ONLUS, Milan, Italy
| |
Collapse
|
130
|
Almkvist O, Nordberg A. A biomarker-validated time scale in years of disease progression has identified early- and late-onset subgroups in sporadic Alzheimer's disease. Alzheimers Res Ther 2023; 15:89. [PMID: 37131241 PMCID: PMC10152764 DOI: 10.1186/s13195-023-01231-8] [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: 11/29/2022] [Accepted: 04/14/2023] [Indexed: 05/04/2023]
Abstract
BACKGROUND It is possible to calculate the number of years to the expected clinical onset (YECO) of autosomal-dominant Alzheimer's disease (adAD). A similar time scale is lacking for sporadic Alzheimer's disease (sAD). The purpose was to design and validate a time scale in YECO for patients with sAD in relation to CSF and PET biomarkers. METHODS Patients diagnosed with Alzheimer's disease (AD, n = 48) or mild cognitive impairment (MCI, n = 46) participated in the study. They underwent a standardized clinical examination at the Memory clinic, Karolinska University Hospital, Stockholm, Sweden, which included present and previous medical history, laboratory screening, cognitive assessment, CSF biomarkers (Aβ42, total-tau, and p-tau), and an MRI of the brain. They were also assessed with two PET tracers, 11C-Pittsburgh compound B and 18F-fluorodeoxyglucose. Assuming concordance of cognitive decline in sAD and adAD, YECO for these patients was calculated using equations for the relationship between cognitive performance, YECO, and years of education in adAD (Almkvist et al. J Int Neuropsychol Soc 23:195-203, 2017). RESULTS The mean current point of disease progression was 3.2 years after the estimated clinical onset in patients with sAD and 3.4 years prior to the estimated clinical onset in patients with MCI, as indicated by the median YECO from five cognitive tests. The associations between YECO and biomarkers were significant, while those between chronological age and biomarkers were nonsignificant. The estimated disease onset (chronological age minus YECO) followed a bimodal distribution with frequency maxima before (early-onset) and after (late-onset) 65 years of age. The early- and late-onset subgroups differed significantly in biomarkers and cognition, but after control for YECO, this difference disappeared for all except the APOE e4 gene (more frequent in early- than in late-onset). CONCLUSIONS A novel time scale in years of disease progression based on cognition was designed and validated in patients with AD using CSF and PET biomarkers. Two early- and late-disease onset subgroups were identified differing with respect to APOE e4.
Collapse
Affiliation(s)
- Ove Almkvist
- Division of Clinical Geriatrics, Department of Neurobiology Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden.
- Theme Inflammation and Aging, Karolinska University Hospital, Stockholm, Sweden.
- Department of Psychology, Stockholm University, Stockholm, Sweden.
| | - Agneta Nordberg
- Division of Clinical Geriatrics, Department of Neurobiology Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden
- Theme Inflammation and Aging, Karolinska University Hospital, Stockholm, Sweden
| |
Collapse
|
131
|
Chen P, Yao H, Tijms BM, Wang P, Wang D, Song C, Yang H, Zhang Z, Zhao K, Qu Y, Kang X, Du K, Fan L, Han T, Yu C, Zhang X, Jiang T, Zhou Y, Lu J, Han Y, Liu B, Zhou B, Liu Y. Four Distinct Subtypes of Alzheimer's Disease Based on Resting-State Connectivity Biomarkers. Biol Psychiatry 2023; 93:759-769. [PMID: 36137824 DOI: 10.1016/j.biopsych.2022.06.019] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/13/2022] [Revised: 05/19/2022] [Accepted: 06/13/2022] [Indexed: 11/29/2022]
Abstract
BACKGROUND Alzheimer's disease (AD) is a neurodegenerative disorder with significant heterogeneity. Different AD phenotypes may be associated with specific brain network changes. Uncovering disease heterogeneity by using functional networks could provide insights into precise diagnoses. METHODS We investigated the subtypes of AD using nonnegative matrix factorization clustering on the previously identified 216 resting-state functional connectivities that differed between AD and normal control subjects. We conducted the analysis using a discovery dataset (n = 809) and a validated dataset (n = 291). Next, we grouped individuals with mild cognitive impairment according to the model obtained in the AD groups. Finally, the clinical measures and brain structural characteristics were compared among the subtypes to assess their relationship with differences in the functional network. RESULTS Individuals with AD were clustered into 4 subtypes reproducibly, which included those with 1) diffuse and mild functional connectivity disruption (subtype 1), 2) predominantly decreased connectivity in the default mode network accompanied by an increase in the prefrontal circuit (subtype 2), 3) predominantly decreased connectivity in the anterior cingulate cortex accompanied by an increase in prefrontal cortex connectivity (subtype 3), and 4) predominantly decreased connectivity in the basal ganglia accompanied by an increase in prefrontal cortex connectivity (subtype 4). In addition to these differences in functional connectivity, differences between the AD subtypes were found in cognition, structural measures, and cognitive decline patterns. CONCLUSIONS These comprehensive results offer new insights that may advance precision medicine for AD and facilitate strategies for future clinical trials.
Collapse
Affiliation(s)
- Pindong Chen
- Brainnetome Center & National Laboratory of Pattern Recognition, Institute of Automation, Chinese Academy of Sciences, Beijing, China; School of Artificial Intelligence, University of Chinese Academy of Sciences, Beijing, China
| | - Hongxiang Yao
- Department of Radiology, the Second Medical Centre, National Clinical Research Centre for Geriatric Diseases, Chinese PLA General Hospital, Beijing, China
| | - Betty M Tijms
- Alzheimer Center Amsterdam, Department of Neurology, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam UMC - Location VUmc, The Netherlands
| | - Pan Wang
- Department of Neurology, Tianjin Huanhu Hospital, Tianjin University, Tianjin, China
| | - Dawei Wang
- Department of Radiology, Qilu Hospital of Shandong University, Ji'nan, China
| | - Chengyuan Song
- Department of Neurology, Qilu Hospital of Shandong University, Ji'nan, China
| | - Hongwei Yang
- Department of Radiology, Xuanwu Hospital of Capital Medical University, Beijing, China
| | | | - Kun Zhao
- Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science & Medical Engineering, Beihang University, Beijing, China
| | - Yida Qu
- Brainnetome Center & National Laboratory of Pattern Recognition, Institute of Automation, Chinese Academy of Sciences, Beijing, China; School of Artificial Intelligence, University of Chinese Academy of Sciences, Beijing, China
| | - Xiaopeng Kang
- Brainnetome Center & National Laboratory of Pattern Recognition, Institute of Automation, Chinese Academy of Sciences, Beijing, China; School of Artificial Intelligence, University of Chinese Academy of Sciences, Beijing, China
| | - Kai Du
- Brainnetome Center & National Laboratory of Pattern Recognition, Institute of Automation, Chinese Academy of Sciences, Beijing, China; School of Artificial Intelligence, University of Chinese Academy of Sciences, Beijing, China
| | - Lingzhong Fan
- Brainnetome Center & National Laboratory of Pattern Recognition, Institute of Automation, Chinese Academy of Sciences, Beijing, China
| | - Tong Han
- Department of Radiology, Tianjin Huanhu Hospital, Tianjin University, Tianjin, China
| | - Chunshui Yu
- Department of Radiology, Tianjin Medical University General Hospital, Tianjin, China
| | - Xi Zhang
- Department of Neurology, the Second Medical Centre, National Clinical Research Centre for Geriatric Diseases, Chinese PLA General Hospital, Beijing, China
| | - Tianzi Jiang
- Brainnetome Center & National Laboratory of Pattern Recognition, Institute of Automation, Chinese Academy of Sciences, Beijing, China; School of Artificial Intelligence, University of Chinese Academy of Sciences, Beijing, China
| | - Yuying Zhou
- Department of Neurology, Tianjin Huanhu Hospital, Tianjin University, Tianjin, China
| | - Jie Lu
- Department of Radiology, Xuanwu Hospital of Capital Medical University, Beijing, China
| | - Ying Han
- Department of Neurology, Xuanwu Hospital of Capital Medical University, Beijing, China; Beijing Institute of Geriatrics, Beijing, China; National Clinical Research Center for Geriatric Disorders, Beijing, China
| | - Bing Liu
- State Key Laboratory of Cognition Neuroscience & Learning, Beijing Normal University, Beijing, China
| | - Bo Zhou
- Department of Neurology, the Second Medical Centre, National Clinical Research Centre for Geriatric Diseases, Chinese PLA General Hospital, Beijing, China.
| | - Yong Liu
- Brainnetome Center & National Laboratory of Pattern Recognition, Institute of Automation, Chinese Academy of Sciences, Beijing, China; School of Artificial Intelligence, University of Chinese Academy of Sciences, Beijing, China; School of Artificial Intelligence, Beijing University of Posts and Telecommunications, Beijing, China.
| |
Collapse
|
132
|
Homolak J. Targeting the microbiota-mitochondria crosstalk in neurodegeneration with senotherapeutics. ADVANCES IN PROTEIN CHEMISTRY AND STRUCTURAL BIOLOGY 2023; 136:339-383. [PMID: 37437983 DOI: 10.1016/bs.apcsb.2023.02.018] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/14/2023]
Abstract
Neurodegenerative diseases are a group of age-related disorders characterized by a chronic and progressive loss of function and/or structure of synapses, neurons, and glial cells. The etiopathogenesis of neurodegenerative diseases is characterized by a complex network of intricately intertwined pathophysiological processes that are still not fully understood. Safe and effective disease-modifying treatments are urgently needed, but still not available. Accumulating evidence suggests that gastrointestinal dyshomeostasis and microbial dysbiosis might play an important role in neurodegeneration by acting as either primary or secondary pathophysiological factors. The research on the role of microbiota in neurodegeneration is in its early phase; however, accumulating evidence suggests that dysbiosis might promote neurodegenerative diseases by disrupting mitochondrial function and inducing mitochondrial dysfunction-associated senescence (MiDAS), possibly due to bidirectional crosstalk based on the common evolutionary origin of mitochondria and bacteria. Cellular senescence is an onco-supressive homeostatic mechanism that results in an irreversible cell cycle arrest upon exposure to noxious stimuli. Senescent cells resist apoptosis via senescent cell anti-apoptotic pathways (SCAPs) and transition into a state known as senescence-associated secretory phenotype (SASP) that generates a cytotoxic proinflammatory microenvironment. Cellular senescence results in the adoption of a detrimental vicious cycle driven by dysbiosis, mitochondrial dysfunction, inflammation, and oxidative stress - a pathophysiological positive feedback loop that results in neuroinflammation and neurodegeneration. Detrimental effects of MiDAS might be prevented and abolished by mitochondria-targeted senotherapeutics, a group of drugs specifically designed to alleviate senescence by inhibiting SCAPs (senolytics), or inhibiting SASP (senomorphics).
Collapse
Affiliation(s)
- Jan Homolak
- Department of Pharmacology, University of Zagreb School of Medicine, Zagreb, Croatia; Croatian Institute for Brain Research, University of Zagreb School of Medicine, Zagreb, Croatia.
| |
Collapse
|
133
|
Llamas-Rodríguez J, Oltmer J, Marshall M, Champion S, Frosch MP, Augustinack JC. TDP-43 and tau concurrence in the entorhinal subfields in primary age-related tauopathy and preclinical Alzheimer's disease. Brain Pathol 2023:e13159. [PMID: 37037195 DOI: 10.1111/bpa.13159] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2023] [Accepted: 03/24/2023] [Indexed: 04/12/2023] Open
Abstract
Phosphorylated tau (p-tau) pathology correlates strongly with cognitive decline and is a pathological hallmark of Alzheimer's Disease (AD). In recent years, phosphorylated transactive response DNA-binding protein (pTDP-43) has emerged as a common comorbidity, found in up to 70% of all AD cases (Josephs et al., Acta Neuropathol, 131(4), 571-585; Josephs, Whitwell, et al., Acta Neuropathol, 127(6), 811-824). Current staging schemes for pTDP-43 in AD and primary age-related tauopathy (PART) track its progression throughout the brain, but the distribution of pTDP-43 within the entorhinal cortex (EC) at the earliest stages has not been studied. Moreover, the exact nature of p-tau and pTDP-43 co-localization is debated. We investigated the selective vulnerability of the entorhinal subfields to phosphorylated pTDP-43 pathology in preclinical AD and PART postmortem tissue. Within the EC, posterior-lateral subfields showed the highest semi-quantitative pTDP-43 density scores, while the anterior-medial subfields had the lowest. On the rostrocaudal axis, pTDP-43 scores were higher posteriorly than anteriorly (p < 0.010), peaking at the posterior-most level (p < 0.050). Further, we showed the relationship between pTDP-43 and p-tau in these regions at pathology-positive but clinically silent stages. P-tau and pTDP-43 presented a similar pattern of affected subregions (p < 0.0001) but differed in density magnitude (p < 0.0001). P-tau burden was consistently higher than pTDP-43 at every anterior-posterior level and in most EC subfields. These findings highlight pTDP-43 burden heterogeneity within the EC and the posterior-lateral subfields as the most vulnerable regions within stage II of the current pTDP-43 staging schemes for AD and PART. The EC is a point of convergence for p-tau and pTDP-43 and identifying its most vulnerable neuronal populations will prove key for early diagnosis and disease intervention.
Collapse
Affiliation(s)
- Josué Llamas-Rodríguez
- Department of Radiology, Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Charlestown, Massachusetts, USA
| | - Jan Oltmer
- Department of Radiology, Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Charlestown, Massachusetts, USA
| | - Michael Marshall
- Department of Neuropathology, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Samantha Champion
- Department of Neuropathology, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Matthew P Frosch
- Department of Neuropathology, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Jean C Augustinack
- Department of Radiology, Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Charlestown, Massachusetts, USA
| |
Collapse
|
134
|
Yang Y, Schilling K, Shashikumar N, Jasodanand V, Moore EE, Pechman KR, Bilgel M, Beason‐Held LL, An Y, Shafer A, Risacher SL, Landman BA, Jefferson AL, Saykin AJ, Resnick SM, Hohman TJ, Archer DB. White matter microstructural metrics are sensitively associated with clinical staging in Alzheimer's disease. ALZHEIMER'S & DEMENTIA (AMSTERDAM, NETHERLANDS) 2023; 15:e12425. [PMID: 37213219 PMCID: PMC10192723 DOI: 10.1002/dad2.12425] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/18/2022] [Revised: 03/06/2023] [Accepted: 03/12/2023] [Indexed: 05/23/2023]
Abstract
Introduction White matter microstructure may be abnormal along the Alzheimer's disease (AD) continuum. Methods Diffusion magnetic resonance imaging (dMRI) data from the Alzheimer's Disease Neuroimaging Initiative (ADNI, n = 627), Baltimore Longitudinal Study of Aging (BLSA, n = 684), and Vanderbilt Memory & Aging Project (VMAP, n = 296) cohorts were free-water (FW) corrected and conventional, and FW-corrected microstructural metrics were quantified within 48 white matter tracts. Microstructural values were subsequently harmonized using the Longitudinal ComBat technique and inputted as independent variables to predict diagnosis (cognitively unimpaired [CU], mild cognitive impairment [MCI], AD). Models were adjusted for age, sex, race/ethnicity, education, apolipoprotein E (APOE) ε4 carrier status, and APOE ε2 carrier status. Results Conventional dMRI metrics were associated globally with diagnostic status; following FW correction, the FW metric itself exhibited global associations with diagnostic status, but intracellular metric associations were diminished. Discussion White matter microstructure is altered along the AD continuum. FW correction may provide further understanding of the white matter neurodegenerative process in AD. Highlights Longitudinal ComBat successfully harmonized large-scale diffusion magnetic resonance imaging (dMRI) metrics.Conventional dMRI metrics were globally sensitive to diagnostic status.Free-water (FW) correction mitigated intracellular associations with diagnostic status.The FW metric itself was globally sensitive to diagnostic status. Multivariate conventional and FW-corrected models may provide complementary information.
Collapse
Affiliation(s)
- Yisu Yang
- Vanderbilt Memory and Alzheimer's CenterVanderbilt University School of MedicineNashvilleTennesseeUSA
| | - Kurt Schilling
- Vanderbilt University Institute of Imaging ScienceVanderbilt University Medical CenterNashvilleTennesseeUSA
- Department of Radiology & Radiological SciencesVanderbilt University Medical CenterNashvilleTennesseeUSA
| | - Niranjana Shashikumar
- Vanderbilt Memory and Alzheimer's CenterVanderbilt University School of MedicineNashvilleTennesseeUSA
| | - Varuna Jasodanand
- Vanderbilt Memory and Alzheimer's CenterVanderbilt University School of MedicineNashvilleTennesseeUSA
| | - Elizabeth E. Moore
- Vanderbilt Memory and Alzheimer's CenterVanderbilt University School of MedicineNashvilleTennesseeUSA
| | - Kimberly R. Pechman
- Vanderbilt Memory and Alzheimer's CenterVanderbilt University School of MedicineNashvilleTennesseeUSA
| | - Murat Bilgel
- Laboratory of Behavioral NeuroscienceNational Institute on AgingNational Institutes of HealthBaltimoreMarylandUSA
| | - Lori L. Beason‐Held
- Laboratory of Behavioral NeuroscienceNational Institute on AgingNational Institutes of HealthBaltimoreMarylandUSA
| | - Yang An
- Laboratory of Behavioral NeuroscienceNational Institute on AgingNational Institutes of HealthBaltimoreMarylandUSA
| | - Andrea Shafer
- Laboratory of Behavioral NeuroscienceNational Institute on AgingNational Institutes of HealthBaltimoreMarylandUSA
| | - Shannon L. Risacher
- Indiana University School of MedicineIndianapolisIndianaUSA
- Indiana Alzheimer's Disease Research CenterIndianapolisIndianaUSA
| | - Bennett A. Landman
- Vanderbilt Memory and Alzheimer's CenterVanderbilt University School of MedicineNashvilleTennesseeUSA
- Vanderbilt University Institute of Imaging ScienceVanderbilt University Medical CenterNashvilleTennesseeUSA
- Department of Radiology & Radiological SciencesVanderbilt University Medical CenterNashvilleTennesseeUSA
- Department of Biomedical EngineeringVanderbilt UniversityNashvilleTennesseeUSA
- Department of Electrical and Computer EngineeringVanderbilt UniversityNashvilleTennesseeUSA
| | - Angela L. Jefferson
- Vanderbilt Memory and Alzheimer's CenterVanderbilt University School of MedicineNashvilleTennesseeUSA
- Vanderbilt Genetics InstituteVanderbilt University Medical CenterNashvilleTennesseeUSA
- Department of MedicineVanderbilt University Medical CenterNashvilleTennesseeUSA
| | - Andrew J. Saykin
- Indiana University School of MedicineIndianapolisIndianaUSA
- Indiana Alzheimer's Disease Research CenterIndianapolisIndianaUSA
| | - Susan M. Resnick
- Laboratory of Behavioral NeuroscienceNational Institute on AgingNational Institutes of HealthBaltimoreMarylandUSA
| | - Timothy J. Hohman
- Vanderbilt Memory and Alzheimer's CenterVanderbilt University School of MedicineNashvilleTennesseeUSA
- Vanderbilt Genetics InstituteVanderbilt University Medical CenterNashvilleTennesseeUSA
| | - Derek B. Archer
- Vanderbilt Memory and Alzheimer's CenterVanderbilt University School of MedicineNashvilleTennesseeUSA
- Vanderbilt Genetics InstituteVanderbilt University Medical CenterNashvilleTennesseeUSA
| | | |
Collapse
|
135
|
Langer Horvat L, Španić Popovački E, Babić Leko M, Zubčić K, Horvat L, Mustapić M, Hof PR, Šimić G. Anterograde and Retrograde Propagation of Inoculated Human Tau Fibrils and Tau Oligomers in a Non-Transgenic Rat Tauopathy Model. Biomedicines 2023; 11:1004. [PMID: 37189622 PMCID: PMC10135744 DOI: 10.3390/biomedicines11041004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2023] [Revised: 02/27/2023] [Accepted: 03/07/2023] [Indexed: 05/17/2023] Open
Abstract
The tauopathy of Alzheimer's disease (AD) is first observed in the brainstem and entorhinal cortex, spreading trans-synaptically along specific pathways to other brain regions with recognizable patterns. Tau propagation occurs retrogradely and anterogradely (trans-synaptically) along a given pathway and through exosomes and microglial cells. Some aspects of in vivo tau spreading have been replicated in transgenic mice models expressing a mutated human MAPT (tau) gene and in wild-type mice. In this study, we aimed to characterize the propagation of different forms of tau species in non-transgenic 3-4 months old wild-type rats after a single unilateral injection of human tau oligomers and tau fibrils into the medial entorhinal cortex (mEC). We determined whether different variants of the inoculated human tau protein, tau fibrils, and tau oligomers, would induce similar neurofibrillary changes and propagate in an AD-related pattern, and how tau-related pathological changes would correlate with presumed cognitive impairment. We injected human tau fibrils and tau oligomers stereotaxically into the mEC and examined the distribution of tau-related changes at 3 days and 4, 8, and 11 months post-injection using antibodies AT8 and MC1, which reveal early phosphorylation and aberrant conformation of tau, respectively, HT7, anti-synaptophysin, and the Gallyas silver staining method. Human tau oligomers and tau fibrils exhibited some similarities and some differences in their ability to seed and propagate tau-related changes. Both human tau fibrils and tau oligomers rapidly propagated from the mEC anterogradely into the hippocampus and various parts of the neocortex. However, using a human tau-specific HT7 antibody, 3 days post-injection we found inoculated human tau oligomers in the red nucleus, primary motor, and primary somatosensory cortex, a finding not seen in animals inoculated with human tau fibrils. In animals inoculated with human tau fibrils, 3 days post-injection the HT7 antibody showed fibrils in the pontine reticular nucleus, a finding explained only by uptake of human tau fibrils by incoming presynaptic fibers to the mEC and retrograde transport of inoculated human tau fibrils to the brainstem. Rats inoculated with human tau fibrils showed as early as 4 months after inoculation a spread of phosphorylated tau protein at the AT8 epitopes throughout the brain, dramatically faster propagation of neurofibrillary changes than with human tau oligomers. The overall severity of tau protein changes 4, 8, and 11 months after inoculation of human tau oligomers and tau fibrils correlated well with spatial working memory and cognition impairments, as measured by the T-maze spontaneous alternation, novel object recognition, and object location tests. We concluded that this non-trangenic rat model of tauopathy, especially when using human tau fibrils, demonstrates rapidly developing pathologic alterations in neurons, synapses, and identifiable pathways together with cognitive and behavioral changes, through the anterograde and retrograde spreading of neurofibrillary degeneration. Therefore, it represents a promising model for future experimental studies of primary and secondary tauopathies, especially AD.
Collapse
Affiliation(s)
- Lea Langer Horvat
- Department of Neuroscience, Croatian Institute for Brain Research, University of Zagreb School of Medicine, 10000 Zagreb, Croatia
| | - Ena Španić Popovački
- Department of Neuroscience, Croatian Institute for Brain Research, University of Zagreb School of Medicine, 10000 Zagreb, Croatia
| | - Mirjana Babić Leko
- Department of Neuroscience, Croatian Institute for Brain Research, University of Zagreb School of Medicine, 10000 Zagreb, Croatia
| | - Klara Zubčić
- Department of Neuroscience, Croatian Institute for Brain Research, University of Zagreb School of Medicine, 10000 Zagreb, Croatia
| | - Luka Horvat
- Department of Molecular Biology, Faculty of Science, University of Zagreb, 10000 Zagreb, Croatia
| | - Maja Mustapić
- Laboratory of Clinical Investigation, National Institute on Aging Intramural Research Program, National Institutes of Health, Baltimore, MD 21224, USA
| | - Patrick R. Hof
- Nash Family Department of Neuroscience, Friedman Brain Institute, and Ronald M. Loeb Center for Alzheimer’s Disease, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Goran Šimić
- Department of Neuroscience, Croatian Institute for Brain Research, University of Zagreb School of Medicine, 10000 Zagreb, Croatia
| |
Collapse
|
136
|
Bao J, Chang C, Zhang Q, Saykin AJ, Shen L, Long Q. Integrative analysis of multi-omics and imaging data with incorporation of biological information via structural Bayesian factor analysis. Brief Bioinform 2023; 24:bbad073. [PMID: 36882008 PMCID: PMC10387302 DOI: 10.1093/bib/bbad073] [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: 10/31/2022] [Revised: 01/14/2023] [Accepted: 02/10/2023] [Indexed: 03/09/2023] Open
Abstract
MOTIVATION With the rapid development of modern technologies, massive data are available for the systematic study of Alzheimer's disease (AD). Though many existing AD studies mainly focus on single-modality omics data, multi-omics datasets can provide a more comprehensive understanding of AD. To bridge this gap, we proposed a novel structural Bayesian factor analysis framework (SBFA) to extract the information shared by multi-omics data through the aggregation of genotyping data, gene expression data, neuroimaging phenotypes and prior biological network knowledge. Our approach can extract common information shared by different modalities and encourage biologically related features to be selected, guiding future AD research in a biologically meaningful way. METHOD Our SBFA model decomposes the mean parameters of the data into a sparse factor loading matrix and a factor matrix, where the factor matrix represents the common information extracted from multi-omics and imaging data. Our framework is designed to incorporate prior biological network information. Our simulation study demonstrated that our proposed SBFA framework could achieve the best performance compared with the other state-of-the-art factor-analysis-based integrative analysis methods. RESULTS We apply our proposed SBFA model together with several state-of-the-art factor analysis models to extract the latent common information from genotyping, gene expression and brain imaging data simultaneously from the ADNI biobank database. The latent information is then used to predict the functional activities questionnaire score, an important measurement for diagnosis of AD quantifying subjects' abilities in daily life. Our SBFA model shows the best prediction performance compared with the other factor analysis models. AVAILABILITY Code are publicly available at https://github.com/JingxuanBao/SBFA. CONTACT qlong@upenn.edu.
Collapse
Affiliation(s)
- Jingxuan Bao
- Department of Biostatistics, Epidemiology and Informatics, University of Pennsylvania Perelman School of Medicine, Philadelphia, 19104, PA, USA
| | - Changgee Chang
- Department of Biostatistics, Epidemiology and Informatics, University of Pennsylvania Perelman School of Medicine, Philadelphia, 19104, PA, USA
| | - Qiyiwen Zhang
- Department of Biostatistics, Epidemiology and Informatics, University of Pennsylvania Perelman School of Medicine, Philadelphia, 19104, PA, USA
| | - Andrew J Saykin
- Department of Radiology and Imaging Sciences, Indiana University, Indianapolis, 46202, IN, USA
| | - Li Shen
- Department of Biostatistics, Epidemiology and Informatics, University of Pennsylvania Perelman School of Medicine, Philadelphia, 19104, PA, USA
| | - Qi Long
- Department of Biostatistics, Epidemiology and Informatics, University of Pennsylvania Perelman School of Medicine, Philadelphia, 19104, PA, USA
| | | |
Collapse
|
137
|
Schork NJ, Elman JA. Pathway-specific polygenic risk scores correlate with clinical status and Alzheimer's-related biomarkers. RESEARCH SQUARE 2023:rs.3.rs-2583037. [PMID: 36909609 PMCID: PMC10002839 DOI: 10.21203/rs.3.rs-2583037/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/05/2023]
Abstract
Background: APOE is the largest genetic risk factor for sporadic Alzheimer's disease (AD), but there is a substantial polygenic component as well. Polygenic risk scores (PRS) can summarize small effects across the genome but may obscure differential risk associated with different molecular processes and pathways. Variability at the genetic level may contribute to the extensive phenotypic heterogeneity of Alzheimer's disease (AD). Here, we examine polygenic risk impacting specific pathways associated with AD and examined its relationship with clinical status and AD biomarkers of amyloid, tau, and neurodegeneration (A/T/N). Methods: A total of 1,411 participants from the Alzheimer's Disease Neuroimaging Initiative (ADNI) with genotyping data were included. Sets of variants identified from a pathway analysis of AD GWAS summary statistics were combined into clusters based on their assigned pathway. We constructed pathway-specific PRSs for each participant and tested their associations with diagnostic status (AD vs cognitively normal), abnormal levels of amyloid and ptau (positive vs negative), and hippocampal volume. The APOE region was excluded from all PRSs, and analyses controlled for APOE -ε4 carrier status. Results: Thirteen pathway clusters were identified relating to categories such as immune response, amyloid precursor processing, protein localization, lipid transport and binding, tyrosine kinase, and endocytosis. Eight pathway-specific PRSs were significantly associated with AD dementia diagnosis. Amyloid-positivity was associated with endocytosis and fibril formation, response misfolded protein, and regulation protein tyrosine PRSs. Ptau positivity and hippocampal volume were both related to protein localization and mitophagy PRS, and ptau positivity was additionally associated with an immune signaling PRS. A global AD PRS showed stronger associations with diagnosis and all biomarkers compared to pathway PRSs, suggesting a strong synergistic effect of all loci contributing to the global AD PRS. Conclusions: Pathway PRS may contribute to understanding separable disease processes, but do not appear to add significant power for predictive purposes. These findings demonstrate that, although genetic risk for AD is widely distributed, AD-phenotypes may be preferentially associated with risk in specific pathways. Defining genetic risk along multiple dimensions at the individual level may help clarify the etiological heterogeneity in AD.
Collapse
|
138
|
Mohanty R, Ferreira D, Nordberg A, Westman E. Associations between different tau-PET patterns and longitudinal atrophy in the Alzheimer's disease continuum: biological and methodological perspectives from disease heterogeneity. Alzheimers Res Ther 2023; 15:37. [PMID: 36814346 PMCID: PMC9945609 DOI: 10.1186/s13195-023-01173-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Accepted: 01/18/2023] [Indexed: 02/24/2023]
Abstract
BACKGROUND Subtypes and patterns are defined using tau-PET (tau pathology) and structural MRI (atrophy) in Alzheimer's disease (AD). However, the relationship between tau pathology and atrophy across these subtypes/patterns remains unclear. Therefore, we investigated the biological association between baseline tau-PET patterns and longitudinal atrophy in the AD continuum; and the methodological characterization of heterogeneity as a continuous phenomenon over the conventional discrete subgrouping. METHODS In 366 individuals (amyloid-beta-positive cognitively normal, prodromal AD, AD dementia; amyloid-beta-negative cognitively normal), we examined the association between tau-PET patterns and longitudinal MRI. We modeled tau-PET patterns as a (a) continuous phenomenon with key dimensions: typicality and severity; and (b) discrete phenomenon by categorization into patterns: typical, limbic predominant, cortical predominant and minimal tau. Tau-PET patterns and associated longitudinal atrophy were contextualized within the Amyloid/Tau/Neurodegeneration (A/T/N) biomarker scheme. RESULTS Localization and longitudinal atrophy change vary differentially across different tau-PET patterns in the AD continuum. Atrophy, a downstream event, did not always follow a topography akin to the corresponding tau-PET pattern. Further, heterogeneity as a continuous phenomenon offered an alternative and useful characterization, sharing correspondence with the conventional subgrouping. Tau-PET patterns also show differential A/T/N profiles. CONCLUSIONS The site and rate of atrophy are different across the tau-PET patterns. Heterogeneity should be treated as a continuous, not discrete, phenomenon for greater sensitivity. Pattern-specific A/T/N profiles highlight differential multimodal interactions underlying heterogeneity. Therefore, tracking multimodal interactions among biomarkers longitudinally, modeling disease heterogeneity as a continuous phenomenon, and examining heterogeneity across the AD continuum could offer avenues for precision medicine.
Collapse
Affiliation(s)
- Rosaleena Mohanty
- Division of Clinical Geriatrics, Center for Alzheimer Research. Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Blickagången 16, 14152, Huddinge, Sweden.
| | - Daniel Ferreira
- Division of Clinical Geriatrics, Center for Alzheimer Research. Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Blickagången 16, 14152, Huddinge, Sweden
- Department of Radiology, Mayo Clinic, Rochester, MN, USA
| | - Agneta Nordberg
- Division of Clinical Geriatrics, Center for Alzheimer Research. Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Blickagången 16, 14152, Huddinge, Sweden
- Theme Aging, Karolinska University Hospital, Stockholm, Sweden
| | - Eric Westman
- Division of Clinical Geriatrics, Center for Alzheimer Research. Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Blickagången 16, 14152, Huddinge, Sweden
- Department of Neuroimaging, Centre for Neuroimaging Sciences, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| |
Collapse
|
139
|
Sensi SL, Russo M, Tiraboschi P. Biomarkers of diagnosis, prognosis, pathogenesis, response to therapy: Convergence or divergence? Lessons from Alzheimer's disease and synucleinopathies. HANDBOOK OF CLINICAL NEUROLOGY 2023; 192:187-218. [PMID: 36796942 DOI: 10.1016/b978-0-323-85538-9.00015-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/16/2023]
Abstract
Alzheimer's disease (AD) is the most common disorder associated with cognitive impairment. Recent observations emphasize the pathogenic role of multiple factors inside and outside the central nervous system, supporting the notion that AD is a syndrome of many etiologies rather than a "heterogeneous" but ultimately unifying disease entity. Moreover, the defining pathology of amyloid and tau coexists with many others, such as α-synuclein, TDP-43, and others, as a rule, not an exception. Thus, an effort to shift our AD paradigm as an amyloidopathy must be reconsidered. Along with amyloid accumulation in its insoluble state, β-amyloid is becoming depleted in its soluble, normal states, as a result of biological, toxic, and infectious triggers, requiring a shift from convergence to divergence in our approach to neurodegeneration. These aspects are reflected-in vivo-by biomarkers, which have become increasingly strategic in dementia. Similarly, synucleinopathies are primarily characterized by abnormal deposition of misfolded α-synuclein in neurons and glial cells and, in the process, depleting the levels of the normal, soluble α-synuclein that the brain needs for many physiological functions. The soluble to insoluble conversion also affects other normal brain proteins, such as TDP-43 and tau, accumulating in their insoluble states in both AD and dementia with Lewy bodies (DLB). The two diseases have been distinguished by the differential burden and distribution of insoluble proteins, with neocortical phosphorylated tau deposition more typical of AD and neocortical α-synuclein deposition peculiar to DLB. We propose a reappraisal of the diagnostic approach to cognitive impairment from convergence (based on clinicopathologic criteria) to divergence (based on what differs across individuals affected) as a necessary step for the launch of precision medicine.
Collapse
Affiliation(s)
- Stefano L Sensi
- Department of Neuroscience, Imaging, and Clinical Sciences, "G. d'Annunzio" University of Chieti-Pescara, Chieti, Italy; Molecular Neurology Unit, Center for Advanced Studies and Technology-CAST and ITAB Institute for Advanced Biotechnology, "G. d'Annunzio" University of Chieti-Pescara, Chieti, Italy.
| | - Mirella Russo
- Department of Neuroscience, Imaging, and Clinical Sciences, "G. d'Annunzio" University of Chieti-Pescara, Chieti, Italy; Molecular Neurology Unit, Center for Advanced Studies and Technology-CAST and ITAB Institute for Advanced Biotechnology, "G. d'Annunzio" University of Chieti-Pescara, Chieti, Italy
| | - Pietro Tiraboschi
- Division of Neurology V-Neuropathology, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| |
Collapse
|
140
|
Simões JL, Sobierai LD, Leal IF, Dos Santos MV, Coiado JV, Bagatini MD. Action of the Purinergic and Cholinergic Anti-inflammatory Pathways on Oxidative Stress in Patients with Alzheimer's Disease in the Context of the COVID-19 Pandemic. Neuroscience 2023; 512:110-132. [PMID: 36526078 PMCID: PMC9746135 DOI: 10.1016/j.neuroscience.2022.12.007] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Revised: 12/02/2022] [Accepted: 12/05/2022] [Indexed: 12/15/2022]
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), the etiologic agent of the 2019 coronavirus disease (COVID-19), has affected more than 20 million people in Brazil and caused a global health emergency. This virus has the potential to affect various parts of the body and compromise metabolic functions. The virus-mediated neural inflammation of the nervous system is due to a storm of cytokines and oxidative stress, which are the clinical features of Alzheimer's disease (AD). This neurodegenerative disease is aggravated in cases involving SARS-CoV-2 and its inflammatory biomarkers, accelerating accumulation of β-amyloid peptide, hyperphosphorylation of tau protein, and production of reactive oxygen species, which lead to homeostasis imbalance. The cholinergic system, through neurons and the neurotransmitter acetylcholine (ACh), modulates various physiological pathways, such as the response to stress, sleep and wakefulness, sensory information, and the cognitive system. Patients with AD have low concentrations of ACh; hence, therapeutic methods are aimed at adjusting the ACh titers available to the body for maintaining functionality. Herein, we focused on acetylcholinesterase inhibitors, responsible for the degradation of ACh in the synaptic cleft, and muscarinic and nicotinic receptor agonists of the cholinergic system owing to the therapeutic potential of the cholinergic anti-inflammatory pathway in AD associated with SARS-CoV-2 infection.
Collapse
Affiliation(s)
- Júlia L.B. Simões
- Medical School, Federal University of Fronteira Sul, Chapecó, SC, Brazil
| | | | - Inayá F. Leal
- Medical School, Federal University of Fronteira Sul, Chapecó, SC, Brazil
| | | | - João Victor Coiado
- Medical School, Federal University of Fronteira Sul, Chapecó, SC, Brazil
| | - Margarete D. Bagatini
- Graduate Program in Biomedical Sciences, Federal University of Fronteira Sul, Chapecó, SC, Brazil,Corresponding author
| |
Collapse
|
141
|
Shand C, Markiewicz PJ, Cash DM, Alexander DC, Donohue MC, Barkhof F, Oxtoby NP. Heterogeneity in Preclinical Alzheimer's Disease Trial Cohort Identified by Image-based Data-Driven Disease Progression Modelling. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2023:2023.02.07.23285572. [PMID: 36798314 PMCID: PMC9934776 DOI: 10.1101/2023.02.07.23285572] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/12/2023]
Abstract
Importance Undetected biological heterogeneity adversely impacts trials in Alzheimer's disease because rate of cognitive decline - and perhaps response to treatment - differs in subgroups. Recent results show that data-driven approaches can unravel the heterogeneity of Alzheimer's disease progression. The resulting stratification is yet to be leveraged in clinical trials. Objective Investigate whether image-based data-driven disease progression modelling could identify baseline biological heterogeneity in a clinical trial, and whether these subgroups have prognostic or predictive value. Design Screening data from the Anti-Amyloid Treatment in Asymptomatic Alzheimer Disease (A4) Study collected between April 2014 and December 2017, and longitudinal data from the Alzheimer's Disease Neuroimaging Initiative (ADNI) observational study downloaded in February 2022 were used. Setting The A4 Study is an interventional trial involving 67 sites in the US, Canada, Australia, and Japan. ADNI is a multi-center observational study in North America. Participants Cognitively unimpaired amyloid-positive participants with a 3-Tesla T1-weighted MRI scan. Amyloid positivity was determined using florbetapir PET imaging (in A4) and CSF Aβ(1-42) (in ADNI). Main Outcomes and Measures Regional volumes estimated from MRI scans were used as input to the Subtype and Stage Inference (SuStaIn) algorithm. Outcomes included cognitive test scores and SUVr values from florbetapir and flortaucipir PET. Results We included 1,240 Aβ+ participants (and 407 Aβ- controls) from the A4 Study, and 731 A4-eligible ADNI participants. SuStaIn identified three neurodegeneration subtypes - Typical, Cortical, Subcortical - comprising 523 (42%) individuals. The remainder are designated subtype zero (insufficient atrophy). Baseline PACC scores (A4 primary outcome) were significantly worse in the Cortical subtype (median = -1.27, IQR=[-3.34,0.83]) relative to both subtype zero (median=-0.013, IQR=[-1.85,1.67], P<.0001) and the Subcortical subtype (median=0.03, IQR=[-1.78,1.61], P=.0006). In ADNI, over a four-year period (comparable to A4), greater cognitive decline in the mPACC was observed in both the Typical (-0.23/yr; 95% CI, [-0.41,-0.05]; P=.01) and Cortical (-0.24/yr; [-0.42,-0.06]; P=.009) subtypes, as well as the CDR-SB (Typical: +0.09/yr, [0.06,0.12], P<.0001; and Cortical: +0.07/yr, [0.04,0.10], P<.0001). Conclusions and Relevance In a large secondary prevention trial, our image-based model detected neurodegenerative heterogeneity predictive of cognitive heterogeneity. We argue that such a model is a valuable tool to be considered in future trial design to control for previously undetected variance.
Collapse
Affiliation(s)
- Cameron Shand
- Centre for Medical Image Computing, Department of Computer Science, University College London, London, UK
| | - Pawel J Markiewicz
- Centre for Medical Image Computing, Department of Medical Physics and Biomedical Engineering, University College London, London, UK
| | - David M Cash
- Dementia Research Centre, UCL Queen Square Institute of Neurology, University College London, London, UK
| | - Daniel C Alexander
- Centre for Medical Image Computing, Department of Computer Science, University College London, London, UK
| | - Michael C Donohue
- Alzheimer's Therapeutic Research Institute, Keck School of Medicine, University of Southern California, San Diego, USA
| | - Frederik Barkhof
- Centre for Medical Image Computing, Department of Medical Physics and Biomedical Engineering, University College London, London, UK
- Department of Radiology and Nuclear Medicine, VU University Medical Center, Amsterdam, Netherlands
| | - Neil P Oxtoby
- Centre for Medical Image Computing, Department of Computer Science, University College London, London, UK
| |
Collapse
|
142
|
Pan D, Zeng A, Yang B, Lai G, Hu B, Song X, Jiang T. Deep Learning for Brain MRI Confirms Patterned Pathological Progression in Alzheimer's Disease. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2204717. [PMID: 36575159 PMCID: PMC9951348 DOI: 10.1002/advs.202204717] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/17/2022] [Revised: 11/29/2022] [Indexed: 06/17/2023]
Abstract
Deep learning (DL) on brain magnetic resonance imaging (MRI) data has shown excellent performance in differentiating individuals with Alzheimer's disease (AD). However, the value of DL in detecting progressive structural MRI (sMRI) abnormalities linked to AD pathology has yet to be established. In this study, an interpretable DL algorithm named the Ensemble of 3-dimensional convolutional neural network (Ensemble 3DCNN) with enhanced parsing techniques is proposed to investigate the longitudinal trajectories of whole-brain sMRI changes denoting AD onset and progression. A set of 2369 T1-weighted images from the multi-centre Alzheimer's Disease Neuroimaging Initiative and Open Access Series of Imaging Studies cohorts are applied to model derivation, validation, testing, and pattern analysis. An Ensemble-3DCNN-based P-score is generated, based on which multiple brain regions, including amygdala, insular, parahippocampal, and temporal gyrus, exhibit early and connected progressive neurodegeneration. Complex individual variability in the sMRI is also observed. This study combining non-invasive sMRI and interpretable DL in detecting patterned sMRI changes confirmed AD pathological progression, shedding new light on predicting AD progression using whole-brain sMRI.
Collapse
Affiliation(s)
- Dan Pan
- School of Electronics and InformationGuangdong Polytechnic Normal UniversityGuangzhou510665China
| | - An Zeng
- Faculty of Computers, Guangdong University of TechnologyGuangzhou510006China
| | - Baoyao Yang
- Faculty of Computers, Guangdong University of TechnologyGuangzhou510006China
| | - Gangyong Lai
- Faculty of Computers, Guangdong University of TechnologyGuangzhou510006China
| | - Bing Hu
- Department of RadiologyThe Third Affiliated Hospital of SUN Yat‐sen UniversityGuangzhou510630China
| | - Xiaowei Song
- Clinical Research CentreSurrey Memorial HospitalFraser HealthSurreyBritish ColumbiaV3V 1Z2Canada
| | - Tianzi Jiang
- Brainnetome Center & National Laboratory of Pattern RecognitionInstitute of AutomationChinese Academy of SciencesBeijing100190China
| | | |
Collapse
|
143
|
Perovnik M, Rus T, Schindlbeck KA, Eidelberg D. Functional brain networks in the evaluation of patients with neurodegenerative disorders. Nat Rev Neurol 2023; 19:73-90. [PMID: 36539533 DOI: 10.1038/s41582-022-00753-3] [Citation(s) in RCA: 39] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/21/2022] [Indexed: 12/24/2022]
Abstract
Network analytical tools are increasingly being applied to brain imaging maps of resting metabolic activity (PET) or blood oxygenation-dependent signals (functional MRI) to characterize the abnormal neural circuitry that underlies brain diseases. This approach is particularly valuable for the study of neurodegenerative disorders, which are characterized by stereotyped spread of pathology along discrete neural pathways. Identification and validation of disease-specific brain networks facilitate the quantitative assessment of pathway changes over time and during the course of treatment. Network abnormalities can often be identified before symptom onset and can be used to track disease progression even in the preclinical period. Likewise, network activity can be modulated by treatment and might therefore be used as a marker of efficacy in clinical trials. Finally, early differential diagnosis can be achieved by simultaneously measuring the activity levels of multiple disease networks in an individual patient's scans. Although these techniques were originally developed for PET, over the past several years analogous methods have been introduced for functional MRI, a more accessible non-invasive imaging modality. This advance is expected to broaden the application of network tools to large and diverse patient populations.
Collapse
Affiliation(s)
- Matej Perovnik
- Department of Neurology, University Medical Center Ljubljana, Ljubljana, Slovenia.,Medical Faculty, University of Ljubljana, Ljubljana, Slovenia
| | - Tomaž Rus
- Department of Neurology, University Medical Center Ljubljana, Ljubljana, Slovenia.,Medical Faculty, University of Ljubljana, Ljubljana, Slovenia
| | | | - David Eidelberg
- Center for Neurosciences, The Feinstein Institutes for Medical Research, Manhasset, NY, USA.
| |
Collapse
|
144
|
Kuang G, Salowe R, O’Brien J. Genetic Factors Implicated in the Investigation of Possible Connections between Alzheimer's Disease and Primary Open Angle Glaucoma. Genes (Basel) 2023; 14:338. [PMID: 36833265 PMCID: PMC9957421 DOI: 10.3390/genes14020338] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2022] [Revised: 01/19/2023] [Accepted: 01/26/2023] [Indexed: 01/31/2023] Open
Abstract
Both Alzheimer's disease (AD) and primary open angle glaucoma (POAG) are diseases of primary global neurodegeneration with complex pathophysiologies. Throughout the published literature, researchers have highlighted similarities associated with various aspects of both diseases. In light of the increasing number of findings reporting resemblance between the two neurodegenerative processes, scientists have grown interested in possible underlying connections between AD and POAG. In the search for explanations to fundamental mechanisms, a multitude of genes have been studied in each condition, with overlap in the genes of interest between AD and POAG. Greater understanding of genetic factors can drive the research process of identifying relationships and elucidating common pathways of disease. These connections can then be utilized to advance research as well as to generate new clinical applications. Notably, AD and glaucoma are currently diseases with irreversible consequences that often lack effective therapies. An established genetic connection between AD and POAG would serve as the basis for development of gene or pathway targeted strategies relevant to both diseases. Such a clinical application could be of immense benefit to researchers, clinicians, and patients alike. This paper aims to summarize the genetic associations between AD and POAG, describe common underlying mechanisms, discuss potential areas of application, and organize the findings in a review.
Collapse
Affiliation(s)
| | | | - Joan O’Brien
- Scheie Eye Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| |
Collapse
|
145
|
Inguanzo A, Poulakis K, Mohanty R, Schwarz CG, Przybelski SA, Diaz-Galvan P, Lowe VJ, Boeve BF, Lemstra AW, van de Beek M, van der Flier W, Barkhof F, Blanc F, Loureiro de Sousa P, Philippi N, Cretin B, Demuynck C, Nedelska Z, Hort J, Segura B, Junque C, Oppedal K, Aarsland D, Westman E, Kantarci K, Ferreira D. MRI data-driven clustering reveals different subtypes of Dementia with Lewy bodies. NPJ Parkinsons Dis 2023; 9:5. [PMID: 36670121 PMCID: PMC9859778 DOI: 10.1038/s41531-023-00448-6] [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: 08/30/2022] [Accepted: 01/05/2023] [Indexed: 01/21/2023] Open
Abstract
Dementia with Lewy bodies (DLB) is a neurodegenerative disorder with a wide heterogeneity of symptoms, which suggests the existence of different subtypes. We used data-driven analysis of magnetic resonance imaging (MRI) data to investigate DLB subtypes. We included 165 DLB from the Mayo Clinic and 3 centers from the European DLB consortium and performed a hierarchical cluster analysis to identify subtypes based on gray matter (GM) volumes. To characterize the subtypes, we used demographic and clinical data, as well as β-amyloid, tau, and cerebrovascular biomarkers at baseline, and cognitive decline over three years. We identified 3 subtypes: an older subtype with reduced cortical GM volumes, worse cognition, and faster cognitive decline (n = 49, 30%); a subtype with low GM volumes in fronto-occipital regions (n = 76, 46%); and a subtype of younger patients with the highest cortical GM volumes, proportionally lower GM volumes in basal ganglia and the highest frequency of cognitive fluctuations (n = 40, 24%). This study shows the existence of MRI subtypes in DLB, which may have implications for clinical workout, research, and therapeutic decisions.
Collapse
Grants
- R01 AG041851 NIA NIH HHS
- C06 RR018898 NCRR NIH HHS
- P50 AG016574 NIA NIH HHS
- R01 AG040042 NIA NIH HHS
- R01 NS080820 NINDS NIH HHS
- R37 AG011378 NIA NIH HHS
- U01 NS100620 NINDS NIH HHS
- U01 AG006786 NIA NIH HHS
- Alzheimerfonden
- Center for Innovative Medicine (CIMED) Swedish Brain funding (Hjärnfonden) ALF Medicine Swedish Dementia funding (Demensförbundet) Foundation for Geriatric Diseases at Karolinska Institutet Karolinska Institutet travel grants
- Little Family Foundation
- National Institutes of Health (U01-NS100620, P50-AG016574, U01-AG006786, R37-AG011378, R01-AG041851, R01-AG040042, C06-RR018898 and R01-NS080820), Foundation Dr. Corinne Schuler, the Mangurian Foundation for Lewy Body Research, the Elsie and Marvin Dekelboum Family Foundation, the Robert H. and Clarice Smith and Abigail Van Buren Alzheimer’s Disease Research Program
- Projet Hospitalier de Recherche Clinique (PHRC, IDCRB 2012-A00992-41) and Fondation Université de Strasbourg
- The Grant Agency of Charles University (grant PRIMUS 22/MED/011).
- Western Norway Regional Health Authority, the Swedish Foundation for Strategic Research (SSF), the Swedish Research Council (VR)Center for Innovative Medicine (CIMED), the Swedish Brain funding (Hjärnfonden), ALF Medicine.
Collapse
Affiliation(s)
- Anna Inguanzo
- Division of Clinical Geriatrics, Center for Alzheimer Research, Department of Neurobiology, Care Sciences, and Society, Karolinska Institutet, Stockholm, Sweden
- Medical Psychology Unit, Institute of Neurosciences, University of Barcelona, Barcelona, Spain
- Institute of Biomedical Research August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Konstantinos Poulakis
- Division of Clinical Geriatrics, Center for Alzheimer Research, Department of Neurobiology, Care Sciences, and Society, Karolinska Institutet, Stockholm, Sweden
| | - Rosaleena Mohanty
- Division of Clinical Geriatrics, Center for Alzheimer Research, Department of Neurobiology, Care Sciences, and Society, Karolinska Institutet, Stockholm, Sweden
| | | | | | - Patricia Diaz-Galvan
- Division of Clinical Geriatrics, Center for Alzheimer Research, Department of Neurobiology, Care Sciences, and Society, Karolinska Institutet, Stockholm, Sweden
- Department of Radiology, Mayo Clinic, Rochester, MN, US
| | - Val J Lowe
- Department of Radiology, Mayo Clinic, Rochester, MN, US
| | | | - Afina W Lemstra
- Department of Neurology and Alzheimer Center, VU University Medical Center, Amsterdam, Netherlands
| | - Marleen van de Beek
- Department of Neurology and Alzheimer Center, VU University Medical Center, Amsterdam, Netherlands
| | - Wiesje van der Flier
- Department of Neurology and Alzheimer Center, VU University Medical Center, Amsterdam, Netherlands
| | - Frederik Barkhof
- Department of Neurology and Alzheimer Center, VU University Medical Center, Amsterdam, Netherlands
- UCL institutes of neurology and center for medical image computing, London, UK
| | - Frederic Blanc
- Day Hospital of Geriatrics, Memory Resource and Research Center (CM2R) of Strasbourg, Department of Geriatrics, Hopitaux Universitaires de Strasbourg, Strasbourg, France
- University of Strasbourg and French National Center for Scientific Research (CNRS), ICube Laboratory and Federation de Medecine Translationnelle de Strasbourg (FMTS), Team Imagerie Multimodale Integrative en Sante (IMIS)/ICONE, Strasbourg, France
| | - Paulo Loureiro de Sousa
- Day Hospital of Geriatrics, Memory Resource and Research Center (CM2R) of Strasbourg, Department of Geriatrics, Hopitaux Universitaires de Strasbourg, Strasbourg, France
- University of Strasbourg and French National Center for Scientific Research (CNRS), ICube Laboratory and Federation de Medecine Translationnelle de Strasbourg (FMTS), Team Imagerie Multimodale Integrative en Sante (IMIS)/ICONE, Strasbourg, France
| | - Nathalie Philippi
- Day Hospital of Geriatrics, Memory Resource and Research Center (CM2R) of Strasbourg, Department of Geriatrics, Hopitaux Universitaires de Strasbourg, Strasbourg, France
- University of Strasbourg and French National Center for Scientific Research (CNRS), ICube Laboratory and Federation de Medecine Translationnelle de Strasbourg (FMTS), Team Imagerie Multimodale Integrative en Sante (IMIS)/ICONE, Strasbourg, France
| | - Benjamin Cretin
- Day Hospital of Geriatrics, Memory Resource and Research Center (CM2R) of Strasbourg, Department of Geriatrics, Hopitaux Universitaires de Strasbourg, Strasbourg, France
- University of Strasbourg and French National Center for Scientific Research (CNRS), ICube Laboratory and Federation de Medecine Translationnelle de Strasbourg (FMTS), Team Imagerie Multimodale Integrative en Sante (IMIS)/ICONE, Strasbourg, France
| | - Catherine Demuynck
- Day Hospital of Geriatrics, Memory Resource and Research Center (CM2R) of Strasbourg, Department of Geriatrics, Hopitaux Universitaires de Strasbourg, Strasbourg, France
- University of Strasbourg and French National Center for Scientific Research (CNRS), ICube Laboratory and Federation de Medecine Translationnelle de Strasbourg (FMTS), Team Imagerie Multimodale Integrative en Sante (IMIS)/ICONE, Strasbourg, France
| | - Zuzana Nedelska
- Department of Radiology, Mayo Clinic, Rochester, MN, US
- Department of Neurology, Charles University, 2nd Faculty of Medicine, Motol University Hospital, Prague, Czech Republic
- International Clinical Research Center, St. Annes University Hospital Brno, Brno, Czech Republic
| | - Jakub Hort
- Department of Neurology, Charles University, 2nd Faculty of Medicine, Motol University Hospital, Prague, Czech Republic
- International Clinical Research Center, St. Annes University Hospital Brno, Brno, Czech Republic
| | - Barbara Segura
- Medical Psychology Unit, Institute of Neurosciences, University of Barcelona, Barcelona, Spain
- Institute of Biomedical Research August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Carme Junque
- Medical Psychology Unit, Institute of Neurosciences, University of Barcelona, Barcelona, Spain
- Institute of Biomedical Research August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Ketil Oppedal
- Center for Age-Related Medicine, Stavanger University Hospital, Stavanger, Norway
- Stavanger Medical Imaging Laboratory (SMIL), Department of Radiology, Stavanger University Hospital, Stavanger, Norway
- Department of Electrical Engineering and Computer Science, University of Stavanger, Stavanger, Norway
| | - Dag Aarsland
- Department of Neuroimaging, Center for Neuroimaging Sciences, Institute of Psychiatry, Psychology and Neuroscience, Kings College London, London, UK
| | - Eric Westman
- Division of Clinical Geriatrics, Center for Alzheimer Research, Department of Neurobiology, Care Sciences, and Society, Karolinska Institutet, Stockholm, Sweden
- Department of Neuroimaging, Center for Neuroimaging Sciences, Institute of Psychiatry, Psychology and Neuroscience, Kings College London, London, UK
| | | | - Daniel Ferreira
- Division of Clinical Geriatrics, Center for Alzheimer Research, Department of Neurobiology, Care Sciences, and Society, Karolinska Institutet, Stockholm, Sweden.
- Department of Radiology, Mayo Clinic, Rochester, MN, US.
| |
Collapse
|
146
|
Huang XY, Xue LL, Chen TB, Huangfu LR, Wang TH, Xiong LL, Yu CY. Miracle fruit seed as a potential supplement for the treatment of learning and memory disorders in Alzheimer's disease. Front Pharmacol 2023; 13:1080753. [PMID: 36712676 PMCID: PMC9873977 DOI: 10.3389/fphar.2022.1080753] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Accepted: 12/21/2022] [Indexed: 01/12/2023] Open
Abstract
Currently, the treatment of Alzheimer's disease (AD) is still at the stage of symptomatic treatment due to lack of effective drugs. The research on miracle fruit seeds (MFSs) has focused on lipid-lowering and antidiabetic effects, but no therapeutic effects have been reported in AD. The purpose of this study was to provide data resources and a potential drug for treatment of AD. An AD mouse model was established and treated with MFSs for 1 month. The Morris water maze test was used to assess learning memory function in mice. Nissl staining was used to demonstrate histopathological changes. MFSs were found to have therapeutic implications in the AD mouse model, as evidenced by improved learning memory function and an increase in surviving neurons. To explore the mechanism of MFSs in treating AD, network pharmacological approaches, Gene Ontology (GO), Kyoto Encyclopedia of Genes and Genomes (KEGG), and molecular docking studies were carried out. Based on the network pharmacology strategy, 74 components from MFS corresponded to 293 targets related to the AD pathology. Among these targets, AKT1, MAPK3, ESR1, PPARG, PTGS2, EGFR, PPARA, CNR1, ABCB1, and MAPT were identified as the core targets. According to the relevant number of core targets, cis-8-octadecenoic acid, cis-10-octadecenoic acid, 2-dodecenal, and tetradecane are likely to be highly correlated with MFS for AD. Enrichment analysis indicated the common targets mainly enriched in AD and the neurodegeneration-multiple disease signaling pathway. The molecular docking predictions showed that MFSs were stably bound to core targets, specifically AKT1, EGFR, ESR1, PPARA, and PPARG. MFSs may play a therapeutic role in AD by affecting the insulin signaling pathway and the Wnt pathway. The findings of this study provide potential possibilities and drug candidates for the treatment of AD.
Collapse
Affiliation(s)
- Xue-Yan Huang
- Department of Neurology, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, China
| | - Lu-Lu Xue
- State Key Laboratory of Biotherapy, Sichuan University, Chengdu, Sichuan, China
| | - Ting-Bao Chen
- Laboratory Animal Department, Kunming Medical University, Kunming, Yunnan, China
| | - Li-Ren Huangfu
- Laboratory Animal Department, Kunming Medical University, Kunming, Yunnan, China
| | - Ting-Hua Wang
- Department of Neurology, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, China
- State Key Laboratory of Biotherapy, Sichuan University, Chengdu, Sichuan, China
- Laboratory Animal Department, Kunming Medical University, Kunming, Yunnan, China
| | - Liu-Lin Xiong
- Department of Anesthesiology, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, China
| | - Chang-Yin Yu
- Department of Neurology, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, China
| |
Collapse
|
147
|
Gorla A, Sankararaman S, Burchard E, Flint J, Zaitlen N, Rahmani E. Phenotypic subtyping via contrastive learning. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.01.05.522921. [PMID: 36711575 PMCID: PMC9881932 DOI: 10.1101/2023.01.05.522921] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Defining and accounting for subphenotypic structure has the potential to increase statistical power and provide a deeper understanding of the heterogeneity in the molecular basis of complex disease. Existing phenotype subtyping methods primarily rely on clinically observed heterogeneity or metadata clustering. However, they generally tend to capture the dominant sources of variation in the data, which often originate from variation that is not descriptive of the mechanistic heterogeneity of the phenotype of interest; in fact, such dominant sources of variation, such as population structure or technical variation, are, in general, expected to be independent of subphenotypic structure. We instead aim to find a subspace with signal that is unique to a group of samples for which we believe that subphenotypic variation exists (e.g., cases of a disease). To that end, we introduce Phenotype Aware Components Analysis (PACA), a contrastive learning approach leveraging canonical correlation analysis to robustly capture weak sources of subphenotypic variation. In the context of disease, PACA learns a gradient of variation unique to cases in a given dataset, while leveraging control samples for accounting for variation and imbalances of biological and technical confounders between cases and controls. We evaluated PACA using an extensive simulation study, as well as on various subtyping tasks using genotypes, transcriptomics, and DNA methylation data. Our results provide multiple strong evidence that PACA allows us to robustly capture weak unknown variation of interest while being calibrated and well-powered, far superseding the performance of alternative methods. This renders PACA as a state-of-the-art tool for defining de novo subtypes that are more likely to reflect molecular heterogeneity, especially in challenging cases where the phenotypic heterogeneity may be masked by a myriad of strong unrelated effects in the data.
Collapse
Affiliation(s)
- Aditya Gorla
- Bioinformatics Interdepartmental Program, University of California, Los Angeles, Los Angeles, CA, USA
| | - Sriram Sankararaman
- Department of Computer Science, University of California, Los Angeles, Los Angeles, CA, USA
- Department of Computational Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
- Department of Human Genetics, University of California, Los Angeles, Los Angeles, CA, USA
| | - Esteban Burchard
- Department of Medicine, University of California, San Francisco, San Francisco, CA, USA
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, San Francisco, CA, USA
| | - Jonathan Flint
- Department of Psychiatry and Behavioral Sciences, Brain Research Institute, University of California, Los Angeles, Los Angeles, CA, USA
| | - Noah Zaitlen
- Department of Computational Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
- Department of Human Genetics, University of California, Los Angeles, Los Angeles, CA, USA
- Department of Neurology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
| | - Elior Rahmani
- Department of Computational Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
| |
Collapse
|
148
|
Schork NJ, Elman JA. Pathway-Specific Polygenic Risk Scores Correlate with Clinical Status and Alzheimer's Disease-Related Biomarkers. J Alzheimers Dis 2023; 95:915-929. [PMID: 37661888 PMCID: PMC10697039 DOI: 10.3233/jad-230548] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/05/2023]
Abstract
BACKGROUND APOE is the largest genetic risk factor for Alzheimer's disease (AD), but there is a substantial polygenic component. Polygenic risk scores (PRS) can summarize small effects across the genome but may obscure differential risk across molecular processes and pathways that contribute to heterogeneity of disease presentation. OBJECTIVE We examined polygenic risk impacting specific AD-associated pathways and its relationship with clinical status and biomarkers of amyloid, tau, and neurodegeneration (A/T/N). METHODS We analyzed data from 1,411 participants from the Alzheimer's Disease Neuroimaging Initiative (ADNI). We applied pathway analysis and clustering to identify AD-associated "pathway clusters" and construct pathway-specific PRSs (excluding the APOE region). We tested associations with diagnostic status, abnormal levels of amyloid and ptau, and hippocampal volume. RESULTS Thirteen pathway clusters were identified, and eight pathway-specific PRSs were significantly associated with AD diagnosis. Amyloid-positivity was associated with endocytosis and fibril formation, response misfolded protein, and regulation protein tyrosine PRSs. Ptau positivity and hippocampal volume were both related to protein localization and mitophagy PRS, and ptau-positivity was also associated with an immune signaling PRS. A global AD PRS showed stronger associations with diagnosis and all biomarkers compared to pathway PRSs. CONCLUSIONS Pathway PRS may contribute to understanding separable disease processes, but do not add significant power for predictive purposes. These findings demonstrate that AD-phenotypes may be preferentially associated with risk in specific pathways, and defining genetic risk along multiple dimensions may clarify etiological heterogeneity in AD. This approach to delineate pathway-specific PRS can be used to study other complex diseases.
Collapse
Affiliation(s)
- Nicholas J. Schork
- The Translational Genomics Research Institute, Quantitative Medicine and Systems Biology, Phoenix, AZ, USA
- Department of Psychiatry University of California, San Diego, La Jolla, CA, USA
| | - Jeremy A. Elman
- Department of Psychiatry University of California, San Diego, La Jolla, CA, USA
- Center for Behavior Genetics of Aging, University of California, San Diego, La Jolla, CA, USA
| | | |
Collapse
|
149
|
Akushevich I, Kravchenko J, Yashkin A, Doraiswamy PM, Hill CV. Expanding the scope of health disparities research in Alzheimer's disease and related dementias: Recommendations from the "Leveraging Existing Data and Analytic Methods for Health Disparities Research Related to Aging and Alzheimer's Disease and Related Dementias" Workshop Series. ALZHEIMER'S & DEMENTIA (AMSTERDAM, NETHERLANDS) 2023; 15:e12415. [PMID: 36935764 PMCID: PMC10020680 DOI: 10.1002/dad2.12415] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Revised: 02/06/2023] [Accepted: 02/15/2023] [Indexed: 03/18/2023]
Abstract
Topics discussed at the "Leveraging Existing Data and Analytic Methods for Health Disparities Research Related to Aging and Alzheimer's Disease and Related Dementias" workshop, held by Duke University and the Alzheimer's Association with support from the National Institute on Aging, are summarized. Ways in which existing data resources paired with innovative applications of both novel and well-known methodologies can be used to identify the effects of multi-level societal, community, and individual determinants of race/ethnicity, sex, and geography-related health disparities in Alzheimer's disease and related dementia are proposed. Current literature on the population analyses of these health disparities is summarized with a focus on identifying existing gaps in knowledge, and ways to mitigate these gaps using data/method combinations are discussed at the workshop. Substantive and methodological directions of future research capable of advancing health disparities research related to aging are formulated.
Collapse
Affiliation(s)
- Igor Akushevich
- Social Science Research InstituteBiodemography of Aging Research UnitDuke UniversityDurhamNorth CarolinaUSA
| | - Julia Kravchenko
- Duke University School of MedicineDepartment of SurgeryDurhamNorth CarolinaUSA
| | - Arseniy Yashkin
- Social Science Research InstituteBiodemography of Aging Research UnitDuke UniversityDurhamNorth CarolinaUSA
| | - P. Murali Doraiswamy
- Departments of Psychiatry and MedicineDuke University School of MedicineDurhamNorth CarolinaUSA
| | | | | |
Collapse
|
150
|
Abstract
Tauopathies are a clinically and neuropathologically heterogeneous group of neurodegenerative disorders, characterized by abnormal tau aggregates. Tau, a microtubule-associated protein, is important for cytoskeletal structure and intracellular transport. Aberrant posttranslational modification of tau results in abnormal tau aggregates causing neurodegeneration. Tauopathies may be primary, or secondary, where a second protein, such as Aß, is necessary for pathology, for example, in Alzheimer's disease, the most common tauopathy. Primary tauopathies are classified based on tau isoform and cell types where pathology predominates. Primary tauopathies include Pick disease, corticobasal degeneration, progressive supranuclear palsy, and argyrophilic grain disease. Environmental tauopathies include chronic traumatic encephalopathy and geographically isolated tauopathies such as the Guam-Parkinsonian-dementia complex. The clinical presentation of tauopathies varies based on the brain areas affected, generally presenting with a combination of cognitive and motor symptoms either earlier or later in the disease course. As symptoms overlap and tauopathies such as Alzheimer's disease and argyrophilic grain disease often coexist, accurate clinical diagnosis is challenging when biomarkers are unavailable. Available treatments target cognitive, motor, and behavioral symptoms. Disease-modifying therapies have been the focus of drug development, particularly agents targeting Aß and tau pathology in Alzheimer's disease, although most of these trials have failed.
Collapse
Affiliation(s)
- Gayatri Devi
- Department of Neurology and Psychiatry, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY, United States.
| |
Collapse
|