1
|
Lei H, Lv J, Zhang F, Wei L, Shi K, Liu J, He T, Xiong R, Sun F, Zhong T, Zhao J, Ke D, Wang Q, Jiang P, Bao AM, Wang JZ, Yang Y. Improving vulnerable Calbindin1 - neurons in the ventral hippocampus rescues tau-induced impairment of episodic memory. Transl Neurodegener 2025; 14:12. [PMID: 40038800 DOI: 10.1186/s40035-025-00473-w] [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: 10/09/2024] [Accepted: 02/04/2025] [Indexed: 03/06/2025] Open
Abstract
BACKGROUND Intraneuronal accumulation of hyperphosphorylated tau is a hallmark of Alzheimer's disease (AD). Given the significant correlation between tau pathology and memory loss in AD patients, identifying vulnerable brain regions, particularly susceptible neuron types in these regions, will advance our understanding of AD onset and shed light on therapeutic strategies to manage its progression. METHODS Immunofluorescent staining was employed to identify the brain regions and neuron types vulnerable to tau pathology in AD. A combination of chemogenetics, electrophysiological recording, in vivo Ca2+ recording, and a modified temporal-order discrimination behavior test was utilized to investigate the toxicity of tau accumulation to susceptible neurons in the dorsal part of the ventral hippocampus. Proteomics, phosphoproteomics, and molecular targeting were used to explore the underlying mechanisms of neuron susceptibility to tau accumulation in AD. The beneficial effects of microtubule affinity regulating kinase 4 (MARK4) knockdown and administration of DEPhosphorylation TArgeting Chimera (DEPTAC) were evaluated in AD mice with tau pathology. RESULTS In postmortem brains of AD patients, we observed robust accumulation of hyperphosphorylated tau in the anterior hippocampal CA1 region, particularly in its Calbindin1- (Calb1-) neurons, as opposed to the posterior hippocampal CA1 region and Calb1+ neurons. The susceptibility of Calb1- neurons to phospho-tau accumulation was also observed in P301L mice, especially in the dorsal part of ventral (anterior in human) hippocampal CA1 (dvCA1). In P301L mice, dvCA1 displayed distinct protein and phosphorylated protein networks compared with dorsal CA1, accompanied by overactivation of MARK4. Overexpressing human tau in Calb1- neurons in the dvCA1 (dvCA1Calb1- neurons) specifically impairs the temporal-order discrimination of objects. Meanwhile, tau accumulation significantly inhibited the excitability and firing patterns of dvCA1Calb1- neurons associated with temporal-order discrimination. Knocking down MARK4 or reducing hyperphosporylated tau via DEPTAC in P301L mice significantly ameliorated AD-like tau pathology in dvCA1Calb1- neurons and improved temporal-order discrimination of objects. CONCLUSION These findings highlight the crucial role of dvCA1Calb1- neurons in the early stage of tau pathology and demonstrate the potential of targeting phosphorylated tau through MARK4 knockdown or DEPTAC administration to counter the vulnerability of dvCA1Calb1- neurons and, consequently, ameliorate episodic memory deficits in AD.
Collapse
Affiliation(s)
- Huiyang Lei
- Department of Pathophysiology, School of Basic Medicine, Key Laboratory of Education Ministry of China/Hubei Province for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
- Department of Clinical Laboratory, Institute of Translational Medicine, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei, China
| | - Jingru Lv
- Department of Pathophysiology, School of Basic Medicine, Key Laboratory of Education Ministry of China/Hubei Province for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Fuqiang Zhang
- Department of Pathophysiology, School of Basic Medicine, Key Laboratory of Education Ministry of China/Hubei Province for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Linyu Wei
- Department of Pathophysiology, School of Basic Medicine, Key Laboratory of Education Ministry of China/Hubei Province for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Kun Shi
- Department of Pathophysiology, School of Basic Medicine, Key Laboratory of Education Ministry of China/Hubei Province for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Jiale Liu
- Department of Pathophysiology, School of Basic Medicine, Key Laboratory of Education Ministry of China/Hubei Province for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Ting He
- Department of Pathophysiology, School of Basic Medicine, Key Laboratory of Education Ministry of China/Hubei Province for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Rui Xiong
- Department of Pathophysiology, School of Basic Medicine, Key Laboratory of Education Ministry of China/Hubei Province for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Fei Sun
- Department of Pathophysiology, School of Basic Medicine, Key Laboratory of Education Ministry of China/Hubei Province for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Tongkai Zhong
- Department of Pathophysiology, School of Basic Medicine, Key Laboratory of Education Ministry of China/Hubei Province for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Jingqi Zhao
- Department of Pathophysiology, School of Basic Medicine, Key Laboratory of Education Ministry of China/Hubei Province for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Dan Ke
- Department of Pathophysiology, School of Basic Medicine, Key Laboratory of Education Ministry of China/Hubei Province for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Qun Wang
- Department of Pathophysiology, School of Basic Medicine, Key Laboratory of Education Ministry of China/Hubei Province for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Peiran Jiang
- Department of Neurobiology and Department of Neurology of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- National Health and Disease Human Brain Tissue Resource Center, Hangzhou, China
| | - Ai-Min Bao
- Department of Neurobiology and Department of Neurology of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- National Health and Disease Human Brain Tissue Resource Center, Hangzhou, China
| | - Jian-Zhi Wang
- Department of Pathophysiology, School of Basic Medicine, Key Laboratory of Education Ministry of China/Hubei Province for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
- Co-innovation Center of Neuroregeneration, Nantong University, Nantong, 226000, China.
| | - Ying Yang
- Department of Pathophysiology, School of Basic Medicine, Key Laboratory of Education Ministry of China/Hubei Province for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
| |
Collapse
|
2
|
Wu J, Wang J, Xiao Z, Lu J, Ma X, Zhou X, Wu Y, Liang X, Zheng L, Ding D, Zhang H, Guan Y, Zuo C, Zhao Q. Clinical characteristics and biomarker profile in early- and late-onset Alzheimer's disease: the Shanghai Memory Study. Brain Commun 2025; 7:fcaf015. [PMID: 39850631 PMCID: PMC11756380 DOI: 10.1093/braincomms/fcaf015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2024] [Revised: 12/12/2024] [Accepted: 01/13/2025] [Indexed: 01/25/2025] Open
Abstract
Early-onset Alzheimer's disease constitutes ∼5-10% of Alzheimer's disease. Its clinical characteristics and biomarker profiles are not well documented. To compare the characteristics covering clinical, neuropsychological and biomarker profiles between patients with early- and late-onset Alzheimer's disease, we enrolled 203 patients (late-onset Alzheimer's disease = 99; early-onset Alzheimer's disease = 104) from a Chinese hospital-based cohort, the Shanghai Memory Study. A full panel of plasma biomarkers under the amyloid/tau/neurodegeneration framework including plasma amyloid beta 40, amyloid beta 42, total-tau, neurofilament light chain and phosphorylated tau 181 were assayed using ultra-sensitive Simoa technology. Seventy-five patients underwent an amyloid molecular positron emission tomography scan whereas 43 received comprehensive amyloid, Tau deposition and hypometabolism analysis. Clinical features, plasma and imaging biomarkers were compared cross-sectionally. Compared to those with late-onset Alzheimer's disease, patients with early-onset Alzheimer's disease presented more severe impairment in language function, lower frequency of APOE ɛ4 and lower levels of plasma neurofilament light chain (all P < 0.05). The plasma phosphorylated tau 181 concentration and phosphorylated tau 181/amyloid beta 42 ratios were higher in early-onset Alzheimer's disease than in late-onset Alzheimer's disease (all P < 0.05). More severe Tau deposition as indicated by 18F-florzolotau binding in the precuneus, posterior cingulate cortex and angular gyrus was observed in the early-onset Alzheimer's disease group. Plasma phosphorylated tau 181 was associated with earlier age at onset and domain-specific cognitive impairment, especially in patients with early-onset Alzheimer's disease. We concluded that patients with early-onset Alzheimer's disease differed from late-onset Alzheimer's disease in cognitive performance and biomarker profile. A higher burden of pathological tau was observed in early-onset Alzheimer's disease and was associated with earlier age at onset and more profound cognitive impairment.
Collapse
Affiliation(s)
- Jie Wu
- Institute and Department of Neurology, Huashan Hospital, Fudan University, Shanghai 200040, China
- National Clinical Research Center for Aging and Medicine, Huashan Hospital, Fudan University, Shanghai 200040, China
- National Center for Neurological Disorders, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Jing Wang
- National Clinical Research Center for Aging and Medicine, Huashan Hospital, Fudan University, Shanghai 200040, China
- Department of Nuclear Medicine and PET Center, Huashan Hospital, Fudan University, Shanghai 200235, China
| | - Zhenxu Xiao
- Institute and Department of Neurology, Huashan Hospital, Fudan University, Shanghai 200040, China
- National Clinical Research Center for Aging and Medicine, Huashan Hospital, Fudan University, Shanghai 200040, China
- National Center for Neurological Disorders, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Jiaying Lu
- National Clinical Research Center for Aging and Medicine, Huashan Hospital, Fudan University, Shanghai 200040, China
- Department of Nuclear Medicine and PET Center, Huashan Hospital, Fudan University, Shanghai 200235, China
| | - Xiaoxi Ma
- Institute and Department of Neurology, Huashan Hospital, Fudan University, Shanghai 200040, China
- National Clinical Research Center for Aging and Medicine, Huashan Hospital, Fudan University, Shanghai 200040, China
- National Center for Neurological Disorders, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Xiaowen Zhou
- Institute and Department of Neurology, Huashan Hospital, Fudan University, Shanghai 200040, China
- National Clinical Research Center for Aging and Medicine, Huashan Hospital, Fudan University, Shanghai 200040, China
- National Center for Neurological Disorders, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Yuhan Wu
- Institute and Department of Neurology, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Xiaoniu Liang
- Institute and Department of Neurology, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Li Zheng
- Institute and Department of Neurology, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Ding Ding
- Institute and Department of Neurology, Huashan Hospital, Fudan University, Shanghai 200040, China
- National Clinical Research Center for Aging and Medicine, Huashan Hospital, Fudan University, Shanghai 200040, China
- National Center for Neurological Disorders, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Huiwei Zhang
- National Clinical Research Center for Aging and Medicine, Huashan Hospital, Fudan University, Shanghai 200040, China
- Department of Nuclear Medicine and PET Center, Huashan Hospital, Fudan University, Shanghai 200235, China
| | - Yihui Guan
- National Clinical Research Center for Aging and Medicine, Huashan Hospital, Fudan University, Shanghai 200040, China
- Department of Nuclear Medicine and PET Center, Huashan Hospital, Fudan University, Shanghai 200235, China
| | - Chuantao Zuo
- National Clinical Research Center for Aging and Medicine, Huashan Hospital, Fudan University, Shanghai 200040, China
- Department of Nuclear Medicine and PET Center, Huashan Hospital, Fudan University, Shanghai 200235, China
| | - Qianhua Zhao
- Institute and Department of Neurology, Huashan Hospital, Fudan University, Shanghai 200040, China
- National Clinical Research Center for Aging and Medicine, Huashan Hospital, Fudan University, Shanghai 200040, China
- National Center for Neurological Disorders, Huashan Hospital, Fudan University, Shanghai 200040, China
- MOE Frontiers Center for Brain Science, Fudan University, Shanghai 200030, China
| |
Collapse
|
3
|
Ye Z, Lang H, Xie Z, Duan S, Peng B, Chen X, Fang Y, Xin J. Associations of combined accelerated biological aging and genetic susceptibility with incident dementia: a prospective study in the UK Biobank. BMC Med 2024; 22:425. [PMID: 39350213 PMCID: PMC11443929 DOI: 10.1186/s12916-024-03640-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Accepted: 09/16/2024] [Indexed: 10/04/2024] Open
Abstract
BACKGROUND Accelerated biological aging has been verified to be a critical risk factor for a number of age-related diseases, but its role in dementia remained unclear. Whether it modified the effects of genetic factors was also unknown. This study evaluated the associations between accelerated biological aging and dementia and the moderating role of accelerated biological aging in the genetic susceptibility to the disease. METHODS We included 200,731 participants in the UK biobank. Nine clinical blood biomarkers and chronological age were used to calculate Phenotypic age acceleration (PhenoAgeAccel), which is a novel indicator for accelerated biological aging. The associations of PhenoAgeAccel with dementia, both young-onset and late-onset dementia, were assessed by Cox proportional hazard models. Apolipoprotein E (APOE) alleles and polygenic risk scores (PRS) were used to evaluate the genetic risk of dementia. The interactions between genetic susceptibility and biological aging were tested on both multiplicative and additive scales. RESULTS These findings showed individuals who were in the highest quartile of PhenoAgeAccel had a higher risk with incidence of dementia compared to individuals in the lowest quartile of PhenoAgeAccel (HR: 1.145 (95% CI: 1.050, 1.249)). Individuals with biologically older had a higher risk of dementia than individuals with biologically younger (HR: 1.069 (95% CI: 1.004, 1.138)). Furthermore, compared to individuals with biologically younger and low APOE ε4-related genetic risk, individuals with biologically younger and high APOE ε4-related genetic risk (HR:3.048 (95% CI: 2.811, 3.305)) had a higher risk of dementia than individuals with biologically older and high APOE ε4-related genetic risk (HR: 2.765 (95% CI: 2.523, 3.029)). Meanwhile, referring to low dementia PRS and biologically younger, the risk of dementia increased by 72.7% (HR: 1.727 (95% CI: 1.538, 1.939) in the biologically younger and high PRS group and 58.7% (HR: 1.587 (95% CI: 1.404, 1.793) in the biologically older and high PRS group, respectively. The negative interactions between PhenoAgeAccel with APOE ε4 and PRS were also tested on the additive scale. CONCLUSIONS Accelerated biological aging could bring the extra risk of dementia but attenuate the effects of genetic risk on dementia. These findings provide insights for precise prevention and intervention of dementia.
Collapse
Affiliation(s)
- Zirong Ye
- Key Laboratory of Health Technology Assessment of Fujian Province, School of Public Health, Xiamen University, Xiang'an Nan Road, Xiang'an District, Xiamen, Fujian, 361102, China
- National Institute for Data Science in Health and Medicine, Xiamen University, Xiang'an Nan Road, Xiang'an District, Xiamen, Fujian, 361102, China
| | - Haoxiang Lang
- Key Laboratory of Health Technology Assessment of Fujian Province, School of Public Health, Xiamen University, Xiang'an Nan Road, Xiang'an District, Xiamen, Fujian, 361102, China
- National Institute for Data Science in Health and Medicine, Xiamen University, Xiang'an Nan Road, Xiang'an District, Xiamen, Fujian, 361102, China
| | - Zishan Xie
- College of Mechanical and Electrical Engineering, Fujian Agriculture and Forestry University, No. 63, Xiyuan Gong Road, Minhou County, Fuzhou, Fujian, 350000, China
| | - Siyu Duan
- Key Laboratory of Health Technology Assessment of Fujian Province, School of Public Health, Xiamen University, Xiang'an Nan Road, Xiang'an District, Xiamen, Fujian, 361102, China
- National Institute for Data Science in Health and Medicine, Xiamen University, Xiang'an Nan Road, Xiang'an District, Xiamen, Fujian, 361102, China
| | - Bihao Peng
- Department of Neurology, Fujian Medical University Union Hospital, Fujian Key Laboratory of Molecular Neurology and Institute of Neuroscience, Fujian Medical University, No.29, Xinquan Road, Gulou District, Fuzhou, Fujian Province, 350000, China
- Institute of Clinical Neurology, Fujian Medical University, No.29, Xinquan Road, Gulou District, Fuzhou, Fujian Province, 350000, China
| | - Xiaochun Chen
- Department of Neurology, Fujian Medical University Union Hospital, Fujian Key Laboratory of Molecular Neurology and Institute of Neuroscience, Fujian Medical University, No.29, Xinquan Road, Gulou District, Fuzhou, Fujian Province, 350000, China
- Institute of Clinical Neurology, Fujian Medical University, No.29, Xinquan Road, Gulou District, Fuzhou, Fujian Province, 350000, China
| | - Ya Fang
- Key Laboratory of Health Technology Assessment of Fujian Province, School of Public Health, Xiamen University, Xiang'an Nan Road, Xiang'an District, Xiamen, Fujian, 361102, China.
- National Institute for Data Science in Health and Medicine, Xiamen University, Xiang'an Nan Road, Xiang'an District, Xiamen, Fujian, 361102, China.
| | - Jiawei Xin
- Department of Neurology, Fujian Medical University Union Hospital, Fujian Key Laboratory of Molecular Neurology and Institute of Neuroscience, Fujian Medical University, No.29, Xinquan Road, Gulou District, Fuzhou, Fujian Province, 350000, China.
- Institute of Clinical Neurology, Fujian Medical University, No.29, Xinquan Road, Gulou District, Fuzhou, Fujian Province, 350000, China.
| |
Collapse
|
4
|
Oh S, Kim S, Lee JE, Park BY, Hye Won J, Park H. Multimodal analysis of disease onset in Alzheimer's disease using Connectome, Molecular, and genetics data. Neuroimage Clin 2024; 43:103660. [PMID: 39197213 PMCID: PMC11393605 DOI: 10.1016/j.nicl.2024.103660] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2024] [Revised: 08/23/2024] [Accepted: 08/23/2024] [Indexed: 09/01/2024]
Abstract
Alzheimer's disease (AD) and its related age at onset (AAO) are highly heterogeneous, due to the inherent complexity of the disease. They are affected by multiple factors, such as neuroimaging and genetic predisposition. Multimodal integration of various data types is necessary; however, it has been nontrivial due to the high dimensionality of each modality. We aimed to identify multimodal biomarkers of AAO in AD using an extended version of sparse canonical correlation analysis, in which we integrated two imaging modalities, functional magnetic resonance imaging (fMRI) and positron emission tomography (PET), and genetic data in the form of single-nucleotide polymorphisms (SNPs) obtained from the Alzheimer's disease neuroimaging initiative database. These three modalities cover low-to-high-level complementary information and offer multiscale insights into the AAO. We identified multivariate markers of AAO in AD using fMRI, PET, and SNP. Furthermore, the markers identified were largely consistent with those reported in the existing literature. In particular, our serial mediation analysis suggests that genetic variants influence the AAO in AD by indirectly affecting brain connectivity by mediation of amyloid-beta protein accumulation, supporting a plausible path in existing research. Our approach provides comprehensive biomarkers related to AAO in AD and offers novel multimodal insights into AD.
Collapse
Affiliation(s)
- Sewook Oh
- Department of Electrical and Computer Engineering, Sungkyunkwan University, Suwon, Republic of Korea
| | - Sunghun Kim
- Department of Electrical and Computer Engineering, Sungkyunkwan University, Suwon, Republic of Korea; Department of Artificial Intelligence, Sungkyunkwan University, Suwon, Republic of Korea; Center for Neuroscience Imaging Research, Institute for Basic Science, Suwon, Republic of Korea
| | - Jong-Eun Lee
- Department of Electrical and Computer Engineering, Sungkyunkwan University, Suwon, Republic of Korea; Center for Neuroscience Imaging Research, Institute for Basic Science, Suwon, Republic of Korea
| | - Bo-Yong Park
- Center for Neuroscience Imaging Research, Institute for Basic Science, Suwon, Republic of Korea; Department of Brain and Cognitive Engineering, Korea University, Seoul, Republic of Korea
| | - Ji Hye Won
- Department of Computer Engineering, Pukyong National University, Busan, Republic of Korea
| | - Hyunjin Park
- Department of Electrical and Computer Engineering, Sungkyunkwan University, Suwon, Republic of Korea; Department of Artificial Intelligence, Sungkyunkwan University, Suwon, Republic of Korea; Center for Neuroscience Imaging Research, Institute for Basic Science, Suwon, Republic of Korea.
| |
Collapse
|
5
|
Zhou X, Liu Y, Wu Z, Zhang X, Tao H. Alzheimer's disease and epilepsy: Research hotspots for comorbidity in the era of global aging. Epilepsy Behav 2024; 157:109849. [PMID: 38820684 DOI: 10.1016/j.yebeh.2024.109849] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Revised: 05/18/2024] [Accepted: 05/19/2024] [Indexed: 06/02/2024]
Abstract
Neurological conditions such as Alzheimer's disease (AD) and epilepsy share a significant clinical overlap, particularly in the elderly, with each disorder potentiating the risk of the other. This interplay is significant amidst an aging global demographic. The review explores the classical pathologies of AD, including amyloid-beta plaques and hyperphosphorylated tau, and their potential role in the genesis of epilepsy. It also delves into the imbalance of glutamate and gamma-amino butyric acid activities, a key mechanism in epilepsy that may be influenced by AD pathology. The impact of age of onset on comorbidity is examined, with early-onset AD and Down syndrome presenting higher risks of epilepsy. The review suggests that epilepsy might precede cognitive symptoms in AD, indicating a complex interaction. Sleep modulation is highlighted as a factor, with sleep disturbances potentially contributing to AD progression. The necessity for cautious medication management is emphasized due to the cognitive effects of certain antiepileptic drugs. Animal models are recognized for their importance in understanding the relationship between AD and epilepsy, though creating fully representative models presents a challenge. The review concludes by noting the efficacy of medications such as lamotrigine, levetiracetam, and memantine in managing both conditions and suggests the ketogenic diet and cannabidiol as emerging treatment options, warranting further investigation for comprehensive patient care strategies.
Collapse
Affiliation(s)
- Xu Zhou
- Clinical Research and Experimental Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong 524001, China
| | - Yang Liu
- Department of Neurology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong 524001, China
| | - Zhengjuan Wu
- Department of Neurology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong 524001, China
| | - Xiaolu Zhang
- Department of Neurology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong 524001, China
| | - Hua Tao
- Department of Neurology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong 524001, China; Guangdong Key Laboratory of Age-related Cardiac and Cerebral Diseases, Guangdong Medical University, Zhanjiang, Guangdong 524001, China.
| |
Collapse
|
6
|
McInvale JJ, Canoll P, Hargus G. Induced pluripotent stem cell models as a tool to investigate and test fluid biomarkers in Alzheimer's disease and frontotemporal dementia. Brain Pathol 2024; 34:e13231. [PMID: 38246596 PMCID: PMC11189780 DOI: 10.1111/bpa.13231] [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: 10/03/2023] [Accepted: 11/29/2023] [Indexed: 01/23/2024] Open
Abstract
Neurodegenerative diseases are increasing in prevalence and comprise a large socioeconomic burden on patients and their caretakers. The need for effective therapies and avenues for disease prevention and monitoring is of paramount importance. Fluid biomarkers for neurodegenerative diseases have gained a variety of uses, including informing participant selection for clinical trials, lending confidence to clinical diagnosis and disease staging, determining prognosis, and monitoring therapeutic response. Their role is expected to grow as disease-modifying therapies start to be available to a broader range of patients and as prevention strategies become established. Many of the underlying molecular mechanisms of currently used biomarkers are incompletely understood. Animal models and in vitro systems using cell lines have been extensively employed but face important translatability limitations. Induced pluripotent stem cell (iPSC) technology, where a theoretically unlimited range of cell types can be reprogrammed from peripheral cells sampled from patients or healthy individuals, has gained prominence over the last decade. It is a promising avenue to study physiological and pathological biomarker function and response to experimental therapeutics. Such systems are amenable to high-throughput drug screening or multiomics readouts such as transcriptomics, lipidomics, and proteomics for biomarker discovery, investigation, and validation. The present review describes the current state of biomarkers in the clinical context of neurodegenerative diseases, with a focus on Alzheimer's disease and frontotemporal dementia. We include a discussion of how iPSC models have been used to investigate and test biomarkers such as amyloid-β, phosphorylated tau, neurofilament light chain or complement proteins, and even nominate novel biomarkers. We discuss the limitations of current iPSC methods, mentioning alternatives such as coculture systems and three-dimensional organoids which address some of these concerns. Finally, we propose exciting prospects for stem cell transplantation paradigms using animal models as a preclinical tool to study biomarkers in the in vivo context.
Collapse
Affiliation(s)
- Julie J. McInvale
- Department of Pathology and Cell BiologyColumbia UniversityNew YorkNew YorkUSA
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia UniversityNew YorkNew YorkUSA
- Medical Scientist Training Program, Columbia UniversityNew YorkNew YorkUSA
| | - Peter Canoll
- Department of Pathology and Cell BiologyColumbia UniversityNew YorkNew YorkUSA
| | - Gunnar Hargus
- Department of Pathology and Cell BiologyColumbia UniversityNew YorkNew YorkUSA
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia UniversityNew YorkNew YorkUSA
| |
Collapse
|
7
|
Barrera-Ocampo A. Monoclonal antibodies and aptamers: The future therapeutics for Alzheimer's disease. Acta Pharm Sin B 2024; 14:2795-2814. [PMID: 39027235 PMCID: PMC11252463 DOI: 10.1016/j.apsb.2024.03.034] [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: 02/06/2024] [Revised: 03/06/2024] [Accepted: 03/14/2024] [Indexed: 07/20/2024] Open
Abstract
Alzheimer's disease (AD) is considered the most common and prevalent form of dementia of adult-onset with characteristic progressive impairment in cognition and memory. The cure for AD has not been found yet and the treatments available until recently were only symptomatic. Regardless of multidisciplinary approaches and efforts made by pharmaceutical companies, it was only in the past two years that new drugs were approved for the treatment of the disease. Amyloid beta (Aβ) immunotherapy is at the core of this therapy, which is one of the most innovative approaches looking to change the course of AD. This technology is based on synthetic peptides or monoclonal antibodies (mAb) to reduce Aβ levels in the brain and slow down the advance of neurodegeneration. Hence, this article reviews the state of the art about AD neuropathogenesis, the traditional pharmacologic treatment, as well as the modern active and passive immunization describing approved drugs, and drug prototypes currently under investigation in different clinical trials. In addition, future perspectives on immunotherapeutic strategies for AD and the rise of the aptamer technology as a non-immunogenic alternative to curb the disease progression are discussed.
Collapse
Affiliation(s)
- Alvaro Barrera-Ocampo
- Facultad de Ingeniería, Diseño y Ciencias Aplicadas, Departamento de Ciencias Farmacéuticas y Químicas, Grupo Natura, Universidad Icesi, Cali 760031, Colombia
| |
Collapse
|
8
|
Valdez-Gaxiola CA, Rosales-Leycegui F, Gaxiola-Rubio A, Moreno-Ortiz JM, Figuera LE. Early- and Late-Onset Alzheimer's Disease: Two Sides of the Same Coin? Diseases 2024; 12:110. [PMID: 38920542 PMCID: PMC11202866 DOI: 10.3390/diseases12060110] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2024] [Revised: 05/04/2024] [Accepted: 05/18/2024] [Indexed: 06/27/2024] Open
Abstract
Early-onset Alzheimer's disease (EOAD), defined as Alzheimer's disease onset before 65 years of age, has been significantly less studied than the "classic" late-onset form (LOAD), although EOAD often presents with a more aggressive disease course, caused by variants in the APP, PSEN1, and PSEN2 genes. EOAD has significant differences from LOAD, including encompassing diverse phenotypic manifestations, increased genetic predisposition, and variations in neuropathological burden and distribution. Phenotypically, EOAD can be manifested with non-amnestic variants, sparing the hippocampi with increased tau burden. The aim of this article is to review the different genetic bases, risk factors, pathological mechanisms, and diagnostic approaches between EOAD and LOAD and to suggest steps to further our understanding. The comprehension of the monogenic form of the disease can provide valuable insights that may serve as a roadmap for understanding the common form of the disease.
Collapse
Affiliation(s)
- César A. Valdez-Gaxiola
- División de Genética, Centro de Investigación Biomédica de Occidente, IMSS, Guadalajara 44340, Jalisco, Mexico; (C.A.V.-G.); (F.R.-L.)
- Doctorado en Genética Humana, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Guadalajara 44340, Jalisco, Mexico
| | - Frida Rosales-Leycegui
- División de Genética, Centro de Investigación Biomédica de Occidente, IMSS, Guadalajara 44340, Jalisco, Mexico; (C.A.V.-G.); (F.R.-L.)
- Maestría en Ciencias del Comportamiento, Instituto de Neurociencias, Centro Universitario de Ciencias Biológicas y Agropecuarias, Universidad de Guadalajara, Guadalajara 44340, Jalisco, Mexico
| | - Abigail Gaxiola-Rubio
- Instituto de Investigación en Ciencias Biomédicas, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Guadalajara 44340, Jalisco, Mexico;
- Facultad de Medicina, Universidad Autónoma de Guadalajara, Zapopan 45129, Jalisco, Mexico
| | - José Miguel Moreno-Ortiz
- Doctorado en Genética Humana, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Guadalajara 44340, Jalisco, Mexico
- Instituto de Genética Humana “Dr. Enrique Corona Rivera”, Departamento de Biología Molecular y Genómica, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Guadalajara 44340, Jalisco, Mexico
| | - Luis E. Figuera
- División de Genética, Centro de Investigación Biomédica de Occidente, IMSS, Guadalajara 44340, Jalisco, Mexico; (C.A.V.-G.); (F.R.-L.)
- Doctorado en Genética Humana, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Guadalajara 44340, Jalisco, Mexico
| |
Collapse
|
9
|
Korczyn AD, Grinberg LT. Is Alzheimer disease a disease? Nat Rev Neurol 2024; 20:245-251. [PMID: 38424454 DOI: 10.1038/s41582-024-00940-4] [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] [Accepted: 02/01/2024] [Indexed: 03/02/2024]
Abstract
Dementia, a prevalent condition among older individuals, has profound societal implications. Extensive research has resulted in no cure for what is perceived as the most common dementing illness: Alzheimer disease (AD). AD is defined by specific brain abnormalities - amyloid-β plaques and tau protein neurofibrillary tangles - that are proposed to actively influence the neurodegenerative process. However, conclusive evidence of amyloid-β toxicity is lacking, the mechanisms leading to the accumulation of plaques and tangles are unknown, and removing amyloid-β has not halted neurodegeneration. So, the question remains, are we making progress towards a solution? The complexity of AD is underscored by numerous genetic and environmental risk factors, and diverse clinical presentations, suggesting that AD is more akin to a syndrome than to a traditional disease, with its pathological manifestation representing a convergence of pathogenic pathways. Therefore, a solution requires a multifaceted approach over a single 'silver bullet'. Improved recognition and classification of conditions that converge in plaques and tangle accumulation and their treatment requires the use of multiple strategies simultaneously.
Collapse
Affiliation(s)
- Amos D Korczyn
- Departments of Neurology, Physiology and Pharmacology, Tel Aviv University, Tel Aviv, Israel.
| | - Lea T Grinberg
- Departments of Neurology and Pathology, UCSF, San Francisco, CA, USA
- Global Brain Health Institute, UCSF, San Francisco, CA, USA
- Department of Pathology, University of Sao Paulo Medical School, Sao Paulo, Brazil
| |
Collapse
|
10
|
Mekkes NJ, Groot M, Hoekstra E, de Boer A, Dagkesamanskaia E, Bouwman S, Wehrens SMT, Herbert MK, Wever DD, Rozemuller A, Eggen BJL, Huitinga I, Holtman IR. Identification of clinical disease trajectories in neurodegenerative disorders with natural language processing. Nat Med 2024; 30:1143-1153. [PMID: 38472295 PMCID: PMC11031398 DOI: 10.1038/s41591-024-02843-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2023] [Accepted: 01/31/2024] [Indexed: 03/14/2024]
Abstract
Neurodegenerative disorders exhibit considerable clinical heterogeneity and are frequently misdiagnosed. This heterogeneity is often neglected and difficult to study. Therefore, innovative data-driven approaches utilizing substantial autopsy cohorts are needed to address this complexity and improve diagnosis, prognosis and fundamental research. We present clinical disease trajectories from 3,042 Netherlands Brain Bank donors, encompassing 84 neuropsychiatric signs and symptoms identified through natural language processing. This unique resource provides valuable new insights into neurodegenerative disorder symptomatology. To illustrate, we identified signs and symptoms that differed between frequently misdiagnosed disorders. In addition, we performed predictive modeling and identified clinical subtypes of various brain disorders, indicative of neural substructures being differently affected. Finally, integrating clinical diagnosis information revealed a substantial proportion of inaccurately diagnosed donors that masquerade as another disorder. The unique datasets allow researchers to study the clinical manifestation of signs and symptoms across neurodegenerative disorders, and identify associated molecular and cellular features.
Collapse
Affiliation(s)
- Nienke J Mekkes
- Department of Biomedical Sciences, Section Molecular Neurobiology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands.
- Machine Learning Lab, Data Science Center in Health, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands.
| | - Minke Groot
- The Netherlands Brain Bank, Netherlands Institute for Neuroscience, Amsterdam, The Netherlands
| | - Eric Hoekstra
- Department of Biomedical Sciences, Section Molecular Neurobiology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Alyse de Boer
- Department of Biomedical Sciences, Section Molecular Neurobiology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Ekaterina Dagkesamanskaia
- Department of Biomedical Sciences, Section Molecular Neurobiology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
- Machine Learning Lab, Data Science Center in Health, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Sander Bouwman
- Department of Biomedical Sciences, Section Molecular Neurobiology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Sophie M T Wehrens
- The Netherlands Brain Bank, Netherlands Institute for Neuroscience, Amsterdam, The Netherlands
| | - Megan K Herbert
- The Netherlands Brain Bank, Netherlands Institute for Neuroscience, Amsterdam, The Netherlands
| | - Dennis D Wever
- The Netherlands Brain Bank, Netherlands Institute for Neuroscience, Amsterdam, The Netherlands
| | | | - Bart J L Eggen
- Department of Biomedical Sciences, Section Molecular Neurobiology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Inge Huitinga
- The Netherlands Brain Bank, Netherlands Institute for Neuroscience, Amsterdam, The Netherlands
- Swammerdam Institute for Life Sciences, University of Amsterdam, Amsterdam, The Netherlands
| | - Inge R Holtman
- Department of Biomedical Sciences, Section Molecular Neurobiology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands.
- Machine Learning Lab, Data Science Center in Health, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands.
- The Netherlands Brain Bank, Netherlands Institute for Neuroscience, Amsterdam, The Netherlands.
| |
Collapse
|
11
|
Puris E, Saveleva L, Auriola S, Gynther M, Kanninen KM, Fricker G. Sex-specific changes in protein expression of membrane transporters in the brain cortex of 5xFAD mouse model of Alzheimer's disease. Front Pharmacol 2024; 15:1365051. [PMID: 38572427 PMCID: PMC10989684 DOI: 10.3389/fphar.2024.1365051] [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: 01/03/2024] [Accepted: 02/27/2024] [Indexed: 04/05/2024] Open
Abstract
Membrane transporters playing an important role in the passage of drugs, metabolites and nutrients across the membranes of the brain cells have been shown to be involved in pathogenesis of Alzheimer's disease (AD). However, little is known about sex-specific changes in transporter protein expression at the brain in AD. Here, we investigated sex-specific alterations in protein expression of three ATP-binding cassette (ABC) and five solute carriers (SLC) transporters in the prefrontal cortex of a commonly used model of familial AD (FAD), 5xFAD mice. Sensitive liquid chromatography tandem mass spectrometry-based quantitative targeted absolute proteomic analysis was applied for absolute quantification of transporter protein expression. We compared the changes in transporter protein expressions in 7-month-old male and female 5xFAD mice versus sex-matched wild-type mice. The study revealed a significant sex-specific increase in protein expression of ABCC1 (p = 0.007) only in male 5xFAD mice as compared to sex-matched wild-type animals. In addition, the increased protein expression of glucose transporter 1 (p = 0.01), 4F2 cell-surface antigen heavy chain (p = 0.01) and long-chain fatty acid transport protein 1 (p = 0.02) were found only in female 5xFAD mice as compared to sex-matched wild-type animals. Finally, protein expression of alanine/serine/cysteine/threonine transporter 1 was upregulated in both male (p = 0.02) and female (p = 0.002) 5xFAD mice. The study provides important information about sex-specific changes in brain cortical transporter expression in 5xFAD mice, which will facilitate drug development of therapeutic strategies for AD targeting these transporters and drug delivery research.
Collapse
Affiliation(s)
- Elena Puris
- Institute of Pharmacy and Molecular Biotechnology, Ruprecht-Karls-University, Heidelberg, Germany
| | - Liudmila Saveleva
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - Seppo Auriola
- School of Pharmacy, University of Eastern Finland, Kuopio, Finland
| | - Mikko Gynther
- School of Pharmacy, University of Eastern Finland, Kuopio, Finland
| | - Katja M. Kanninen
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - Gert Fricker
- Institute of Pharmacy and Molecular Biotechnology, Ruprecht-Karls-University, Heidelberg, Germany
| |
Collapse
|
12
|
Zhang Y, Xie X, Chen B, Pan L, Li J, Wang W, Wang J, Tang R, Huang Q, Chen X, Ren R, Zhang Z, Fu W, Wang G. E674Q (Shanghai APP mutant), a novel amyloid precursor protein mutation, in familial late-onset Alzheimer's disease. Genes Dis 2024; 11:1022-1034. [PMID: 37692508 PMCID: PMC10491941 DOI: 10.1016/j.gendis.2023.02.051] [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: 10/26/2022] [Revised: 02/08/2023] [Accepted: 02/21/2023] [Indexed: 09/12/2023] Open
Abstract
Identified as the pathogenic genes of Alzheimer's disease (AD), APP, PSEN1, and PSEN2 mainly lead to early-onset AD, whose course is more aggressive, and atypical symptoms are more common than sporadic AD. Here, a novel missense mutation, APP E674Q (also named "Shanghai APP"), was detected in a Chinese index patient with typical late-onset AD (LOAD) who developed memory decline in his mid-70s. The results from neuroimaging were consistent with AD, where widespread amyloid β deposition was demonstrated in 18F-florbetapir Positron Emission Tomography (PET). APP E674Q is close to the β-secretase cleavage site and the well-studied Swedish APP mutation (KM670/671NL), which was predicted to be pathogenic in silico. Molecular dynamics simulation indicated that the E674Q mutation resulted in a rearrangement of the interaction mode between APP and BACE1 and that the E674Q mutation was more prone to cleavage by BACE1. The in vitro results suggested that the E674Q mutation was pathogenic by facilitating the BACE1-mediated processing of APP and the production of Aβ. Furthermore, we applied an adeno-associated virus (AAV)-mediated transfer of the human E674Q mutant APP gene to the hippocampi of two-month-old C57Bl/6 J mice. AAV-E674Q-injected mice exhibited impaired learning behavior and increased pathological burden in the brain, implying that the E674Q mutation had a pathogenicity that bore a comparison with the classical Swedish mutation. Collectively, we report a strong amyloidogenic effect of the E674Q substitution in AD. To our knowledge, E674Q is the only pathogenic mutation within the amyloid processing sequence causing LOAD.
Collapse
Affiliation(s)
- Yongfang Zhang
- Department of Neurology, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
- Department of Pharmacology and Chemical Biology, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Xinyi Xie
- Department of Neurology, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Boyu Chen
- Department of Medicinal Chemistry, School of Pharmacy, Fudan University, Shanghai 201203, China
| | - Lina Pan
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, China
| | - Jianping Li
- Department of Neurology, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Wanbing Wang
- Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, Institute of Neuroscience, School of Medicine, Xiamen University, Xiamen, Fujian 361102, China
- Shenzhen Research Institute of Xiamen University, Shenzhen, Guangdong 518063, China
| | - Jintao Wang
- Department of Neurology, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Ran Tang
- Department of Neurology, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Qiang Huang
- Department of Neurology, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Xiaofen Chen
- Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, Institute of Neuroscience, School of Medicine, Xiamen University, Xiamen, Fujian 361102, China
- Shenzhen Research Institute of Xiamen University, Shenzhen, Guangdong 518063, China
| | - Rujing Ren
- Department of Neurology, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Zhentao Zhang
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, China
| | - Wei Fu
- Department of Medicinal Chemistry, School of Pharmacy, Fudan University, Shanghai 201203, China
| | - Gang Wang
- Department of Neurology, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| |
Collapse
|
13
|
Wang Y, Li Q, Yao L, He N, Tang Y, Chen L, Long F, Chen Y, Kemp GJ, Lui S, Li F. Shared and differing functional connectivity abnormalities of the default mode network in mild cognitive impairment and Alzheimer's disease. Cereb Cortex 2024; 34:bhae094. [PMID: 38521993 DOI: 10.1093/cercor/bhae094] [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: 10/02/2023] [Revised: 02/19/2024] [Accepted: 02/21/2024] [Indexed: 03/25/2024] Open
Abstract
Alzheimer's disease (AD) and mild cognitive impairment (MCI) both show abnormal resting-state functional connectivity (rsFC) of default mode network (DMN), but it is unclear to what extent these abnormalities are shared. Therefore, we performed a comprehensive meta-analysis, including 31 MCI studies and 20 AD studies. MCI patients, compared to controls, showed decreased within-DMN rsFC in bilateral medial prefrontal cortex/anterior cingulate cortex (mPFC/ACC), precuneus/posterior cingulate cortex (PCC), right temporal lobes, and left angular gyrus and increased rsFC between DMN and left inferior temporal gyrus. AD patients, compared to controls, showed decreased rsFC within DMN in bilateral mPFC/ACC and precuneus/PCC and between DMN and left inferior occipital gyrus and increased rsFC between DMN and right dorsolateral prefrontal cortex. Conjunction analysis showed shared decreased rsFC in mPFC/ACC and precuneus/PCC. Compared to MCI, AD had decreased rsFC in left precuneus/PCC and between DMN and left inferior occipital gyrus and increased rsFC in right temporal lobes. MCI and AD share a decreased within-DMN rsFC likely underpinning episodic memory deficits and neuropsychiatric symptoms, but differ in DMN rsFC alterations likely related to impairments in other cognitive domains such as language, vision, and execution. This may throw light on neuropathological mechanisms in these two stages of dementia.
Collapse
Affiliation(s)
- Yaxuan Wang
- Department of Radiology and Huaxi MR Research Center (HMRRC), Functional and Molecular lmaging Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, No. 37 Guo Xue Alley, Wuhou District, Chengdu 610041, Sichuan Province, P.R. China
| | - Qian Li
- Department of Radiology and Huaxi MR Research Center (HMRRC), Functional and Molecular lmaging Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, No. 37 Guo Xue Alley, Wuhou District, Chengdu 610041, Sichuan Province, P.R. China
| | - Li Yao
- Department of Radiology and Huaxi MR Research Center (HMRRC), Functional and Molecular lmaging Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, No. 37 Guo Xue Alley, Wuhou District, Chengdu 610041, Sichuan Province, P.R. China
| | - Ning He
- Department of Psychiatry, West China Hospital of Sichuan University, No. 37 Guo Xue Alley, Wuhou District, Chengdu 610041, Sichuan, P.R. China
| | - Yingying Tang
- Department of Neurology, West China Hospital of Sichuan University, No. 37 Guo Xue Alley, Wuhou District, Chengdu 610041, Sichuan, P.R. China
| | - Lizhou Chen
- Department of Radiology and Huaxi MR Research Center (HMRRC), Functional and Molecular lmaging Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, No. 37 Guo Xue Alley, Wuhou District, Chengdu 610041, Sichuan Province, P.R. China
| | - Fenghua Long
- Department of Radiology and Huaxi MR Research Center (HMRRC), Functional and Molecular lmaging Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, No. 37 Guo Xue Alley, Wuhou District, Chengdu 610041, Sichuan Province, P.R. China
| | - Yufei Chen
- Department of Radiology and Huaxi MR Research Center (HMRRC), Functional and Molecular lmaging Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, No. 37 Guo Xue Alley, Wuhou District, Chengdu 610041, Sichuan Province, P.R. China
| | - Graham J Kemp
- Institute of Life Course and Medical Sciences, University of Liverpool, 6 West Derby Street, Liverpool L7 8TX, United Kingdom
| | - Su Lui
- Department of Radiology and Huaxi MR Research Center (HMRRC), Functional and Molecular lmaging Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, No. 37 Guo Xue Alley, Wuhou District, Chengdu 610041, Sichuan Province, P.R. China
| | - Fei Li
- Department of Radiology and Huaxi MR Research Center (HMRRC), Functional and Molecular lmaging Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, No. 37 Guo Xue Alley, Wuhou District, Chengdu 610041, Sichuan Province, P.R. China
| |
Collapse
|
14
|
Oh M, Oh SJ, Lee SJ, Oh JS, Seo SY, Ryu S, Roh JH, Lee JH, Kim JS. One-Year Longitudinal Changes in Tau Accumulation on [ 18F]PI-2620 PET in the Alzheimer Spectrum. J Nucl Med 2024; 65:453-461. [PMID: 38302152 DOI: 10.2967/jnumed.123.265893] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2023] [Revised: 12/08/2023] [Accepted: 12/08/2023] [Indexed: 02/03/2024] Open
Abstract
We investigated the longitudinal changes in cortical tau accumulation and their association with cognitive decline in patients in the Alzheimer disease (AD) continuum using 2-(2-([18F]fluoro)pyridin-4-yl)-9H-pyrrolo[2,3-b:4,5c']dipyridine ([18F]PI-2620) PET. Methods: We prospectively enrolled 52 participants (age, 69.7 ± 8.4 y; 18 men and 34 women): 7 with normal cognition, 28 with mild cognitive impairment, and 17 with AD. They all completed the [18F]PI-2620 and [18F]florbetaben PET, MRI, and neuropsychologic tests at baseline and, excepting the [18F]florbetaben PET, at the 1-y follow-up. Amyloid-β (Aβ) PET images were visually scored as positive (+) or negative (-). Patients on the AD continuum, including Aβ+ mild cognitive impairment and AD, were classified into early-onset (EO+) (<65 y old) or late-onset (LO+) (≥65 y old) groups. [18F]PI-2620 PET SUV ratios (SUVRs) were determined by calculating the cerebral-to-inferior cerebellar ratio. Cortical volumes were calculated using 3-dimensional T1-weighted MRI. The correlation between tau accumulation progression and cognitive decline was also investigated. Results: The global [18F]PI-2620 PET SUVRs were 1.04 ± 0.07 in 15 Aβ- patients, 1.18 ± 0.21 in 20 LO+ patients (age, 76.7 ± 3.8 y), and 1.54 ± 0.38 in 17 EO+ patients (age, 63.4 ± 5.4 y; P < 0.001) at baseline. The global SUVR increased over 1 y by 0.05 ± 0.07 (3.90%) and 0.13 ± 0.22 (8.41%) in the LO+ and EO+ groups, respectively, whereas in the Aβ- groups, it remained unchanged. The EO+ group showed higher global and regional tau deposition than did the Aβ- and LO+ groups (P < 0.05 for each) and rapid accumulation in Braak stage V (0.15 ± 0.25; 9.10% ± 12.27%; P = 0.016 and 0.008), Braak stage VI (0.08 ± 0.12; 7.16% ± 10.06%; P < 0.006 and 0.005), and global SUVR (P = 0.013) compared with the Aβ- group. In the EO+ group, the changes in SUVR in Braak stages II-VI were strongly correlated with the baseline and changes in verbal memory (P < 0.03). The LO+ group showed higher tau accumulation in Braak stage I-IV areas than did the Aβ- group (P < 0.001 for each). In the LO+ group, the change in SUVR in Braak stages III and IV moderately correlated with the change in attention (P < 0.05), and the change in SUVR in Braak stages V and VI moderately correlated with the change in visuospatial function (P < 0.005). Conclusion: These findings suggest that [18F]PI-2620 PET can be a biomarker to provide regional and chronologic information about tau pathology in the AD continuum.
Collapse
Affiliation(s)
- Minyoung Oh
- Department of Nuclear Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Seung Jun Oh
- Department of Nuclear Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Sang Ju Lee
- Department of Nuclear Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Jungsu S Oh
- Department of Nuclear Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Seung Yeon Seo
- Department of Nuclear Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
- Department of Electrical and Electronic Engineering, Yonsei University, Seoul, Korea
| | - Soorack Ryu
- Biostatistical Consulting and Research Laboratory, Medical Research Collaborating Center, Hanyang University, Seoul, Korea
| | - Jee Hoon Roh
- Department of Neurology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
- Departments of Biomedical Sciences and Physiology, Korea University College of Medicine, Seoul, Korea; and
- Department of Neurology, Anam Hospital, Korea University College of Medicine, Seoul, Korea
| | - Jae-Hong Lee
- Department of Neurology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Jae Seung Kim
- Department of Nuclear Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea;
| |
Collapse
|
15
|
Hendriks S, Ranson JM, Peetoom K, Lourida I, Tai XY, de Vugt M, Llewellyn DJ, Köhler S. Risk Factors for Young-Onset Dementia in the UK Biobank. JAMA Neurol 2024; 81:134-142. [PMID: 38147328 PMCID: PMC10751655 DOI: 10.1001/jamaneurol.2023.4929] [Citation(s) in RCA: 31] [Impact Index Per Article: 31.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Accepted: 10/30/2023] [Indexed: 12/27/2023]
Abstract
Importance There is limited information on modifiable risk factors for young-onset dementia (YOD). Objective To examine factors that are associated with the incidence of YOD. Design, Setting, and Participants This prospective cohort study used data from the UK Biobank, with baseline assessment between 2006 and 2010 and follow-up until March 31, 2021, for England and Scotland, and February 28, 2018, for Wales. Participants younger than 65 years and without a dementia diagnosis at baseline assessment were included in this study. Participants who were 65 years and older and those with dementia at baseline were excluded. Data were analyzed from May 2022 to April 2023. Exposures A total of 39 potential risk factors were identified from systematic reviews of late-onset dementia and YOD risk factors and grouped into domains of sociodemographic factors (education, socioeconomic status, and sex), genetic factors (apolipoprotein E), lifestyle factors (physical activity, alcohol use, alcohol use disorder, smoking, diet, cognitive activity, social isolation, and marriage), environmental factors (nitrogen oxide, particulate matter, pesticide, and diesel), blood marker factors (vitamin D, C-reactive protein, estimated glomerular filtration rate function, and albumin), cardiometabolic factors (stroke, hypertension, diabetes, hypoglycemia, heart disease, atrial fibrillation, and aspirin use), psychiatric factors (depression, anxiety, benzodiazepine use, delirium, and sleep problems), and other factors (traumatic brain injury, rheumatoid arthritis, thyroid dysfunction, hearing impairment, and handgrip strength). Main Outcome and Measures Multivariable Cox proportional hazards regression was used to study the association between the risk factors and incidence of YOD. Factors were tested stepwise first within domains and then across domains. Results Of 356 052 included participants, 197 036 (55.3%) were women, and the mean (SD) age at baseline was 54.6 (7.0) years. During 2 891 409 person-years of follow-up, 485 incident YOD cases (251 of 485 men [51.8%]) were observed, yielding an incidence rate of 16.8 per 100 000 person-years (95% CI, 15.4-18.3). In the final model, 15 factors were significantly associated with a higher YOD risk, namely lower formal education, lower socioeconomic status, carrying 2 apolipoprotein ε4 allele, no alcohol use, alcohol use disorder, social isolation, vitamin D deficiency, high C-reactive protein levels, lower handgrip strength, hearing impairment, orthostatic hypotension, stroke, diabetes, heart disease, and depression. Conclusions and Relevance In this study, several factors, mostly modifiable, were associated with a higher risk of YOD. These modifiable risk factors should be incorporated in future dementia prevention initiatives and raise new therapeutic possibilities for YOD.
Collapse
Affiliation(s)
- Stevie Hendriks
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience, Alzheimer Center Limburg, Maastricht University, Maastricht, the Netherlands
| | | | - Kirsten Peetoom
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience, Alzheimer Center Limburg, Maastricht University, Maastricht, the Netherlands
| | | | - Xin You Tai
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, United Kingdom
- Division of Clinical Neurology, John Radcliffe Hospital, Oxford University Hospitals Trust, Oxford, United Kingdom
| | - Marjolein de Vugt
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience, Alzheimer Center Limburg, Maastricht University, Maastricht, the Netherlands
| | - David J. Llewellyn
- University of Exeter Medical School, Exeter, United Kingdom
- Alan Turing Institute, London, United Kingdom
| | - Sebastian Köhler
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience, Alzheimer Center Limburg, Maastricht University, Maastricht, the Netherlands
| |
Collapse
|
16
|
Lin S, Wu S, Zhao W, Fang Z, Kang H, Liu X, Pan S, Yu F, Bao Y, Jia P. TargetGene: a comprehensive database of cell-type-specific target genes for genetic variants. Nucleic Acids Res 2024; 52:D1072-D1081. [PMID: 37870478 PMCID: PMC10767789 DOI: 10.1093/nar/gkad901] [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: 08/15/2023] [Revised: 09/27/2023] [Accepted: 10/09/2023] [Indexed: 10/24/2023] Open
Abstract
Annotating genetic variants to their target genes is of great importance in unraveling the causal variants and genetic mechanisms that underlie complex diseases. However, disease-associated genetic variants are often located in non-coding regions and manifest context-specific effects, making it challenging to accurately identify the target genes and regulatory mechanisms. Here, we present TargetGene (https://ngdc.cncb.ac.cn/targetgene/), a comprehensive database reporting target genes for human genetic variants from various aspects. Specifically, we collected a comprehensive catalog of multi-omics data at the single-cell and bulk levels and from various human tissues, cell types and developmental stages. To facilitate the identification of Single Nucleotide Polymorphism (SNP)-to-gene connections, we have implemented multiple analytical tools based on chromatin co-accessibility, 3D interaction, enhancer activities and quantitative trait loci, among others. We applied the pipeline to evaluate variants from nearly 1300 Genome-wide association studies (GWAS) and assembled a comprehensive atlas of multiscale regulation of genetic variants. TargetGene is equipped with user-friendly web interfaces that enable intuitive searching, navigation and browsing through the results. Overall, TargetGene provides a unique resource to empower researchers to study the regulatory mechanisms of genetic variants in complex human traits.
Collapse
Affiliation(s)
- Shiqi Lin
- CAS Key Laboratory of Genomic and Precision Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences and China National Center for Bioinformation, Beijing 100101, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Song Wu
- University of Chinese Academy of Sciences, Beijing 100049, China
- National Genomics Data Center & CAS Key Laboratory of Genome Sciences and Information, Beijing Institute of Genomics, Chinese Academy of Sciences and China National Center for Bioinformation, Beijing 100101, China
| | - Wei Zhao
- University of Chinese Academy of Sciences, Beijing 100049, China
- National Genomics Data Center & CAS Key Laboratory of Genome Sciences and Information, Beijing Institute of Genomics, Chinese Academy of Sciences and China National Center for Bioinformation, Beijing 100101, China
| | - Zhanjie Fang
- CAS Key Laboratory of Genomic and Precision Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences and China National Center for Bioinformation, Beijing 100101, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Hongen Kang
- CAS Key Laboratory of Genomic and Precision Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences and China National Center for Bioinformation, Beijing 100101, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Xinxuan Liu
- CAS Key Laboratory of Genomic and Precision Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences and China National Center for Bioinformation, Beijing 100101, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Siyu Pan
- CAS Key Laboratory of Genomic and Precision Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences and China National Center for Bioinformation, Beijing 100101, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Fudong Yu
- Shanghai-MOST Key Laboratory of Health and Disease Genomics, NHC Key Lab of Reproduction Regulation, Shanghai Institute for Biomedical and Pharmaceutical Technologies, Shanghai 200237, China
| | - Yiming Bao
- University of Chinese Academy of Sciences, Beijing 100049, China
- National Genomics Data Center & CAS Key Laboratory of Genome Sciences and Information, Beijing Institute of Genomics, Chinese Academy of Sciences and China National Center for Bioinformation, Beijing 100101, China
| | - Peilin Jia
- CAS Key Laboratory of Genomic and Precision Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences and China National Center for Bioinformation, Beijing 100101, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| |
Collapse
|
17
|
Ramos-Cejudo J, Scott MR, Tanner JA, Pase MP, McGrath ER, Ghosh S, Osorio RS, Thibault E, El Fakhri G, Johnson KA, Beiser A, Seshadri S. Associations of Plasma Tau with Amyloid and Tau PET: Results from the Community-Based Framingham Heart Study. J Alzheimers Dis 2024; 100:487-494. [PMID: 38875034 DOI: 10.3233/jad-231320] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/16/2024]
Abstract
Background Associations of plasma total tau levels with future risk of AD have been described. Objective To examine the extent to which plasma tau reflects underlying AD brain pathology in cognitively healthy individuals. Methods We examined cross-sectional associations of plasma total tau with 11C-Pittsburgh Compound-B (PiB)-PET and 18F-Flortaucipir (FTP)-PET in middle-aged participants at the community-based Framingham Heart Study. Results Our final sample included 425 participants (mean age 57.6± 9.9, 50% F). Plasma total tau levels were positively associated with amyloid-β deposition in the precuneus region (β±SE, 0.11±0.05; p = 0.025). A positive association between plasma total tau and tau PET in the rhinal cortex was suggested in participants with higher amyloid-PET burden and in APOEɛ4 carriers. Conclusions Our study highlights that plasma total tau is a marker of amyloid deposition as early as in middle-age.
Collapse
Affiliation(s)
- Jaime Ramos-Cejudo
- Department of Psychiatry, New York University (NYU) Grossman School of Medicine, New York, NY, USA
| | - Matthew R Scott
- Department of Biostatistics, Boston University School of Public Health, Boston, MA, USA
| | - Jeremy A Tanner
- Glenn Biggs Institute for Alzheimer's and Neurodegenerative Diseases, University of Texas Health Sciences Center, San Antonio, TX, USA
| | - Matthew P Pase
- Turner Institute for Brain and Mental Health, School of Psychological Sciences, Monash University, Clayton, VIC, Australia
- Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Emer R McGrath
- HRB Clinical Research Facility, University of Galway, Galway, Ireland
- The Framingham Study, Boston, MA, USA
- School of Medicine, University of Galway, Galway, Ireland
| | | | - Ricardo S Osorio
- Department of Psychiatry, New York University (NYU) Grossman School of Medicine, New York, NY, USA
| | - Emma Thibault
- Department of Radiology, Harvard Medical School, Boston, MA, USA
| | | | - Keith A Johnson
- Department of Radiology, Harvard Medical School, Boston, MA, USA
- Department of Neurology, Harvard Medical School, Boston, MA, USA
| | - Alexa Beiser
- Department of Biostatistics, Boston University School of Public Health, Boston, MA, USA
- The Framingham Study, Boston, MA, USA
- Department of Neurology, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, USA
| | - Sudha Seshadri
- Glenn Biggs Institute for Alzheimer's and Neurodegenerative Diseases, University of Texas Health Sciences Center, San Antonio, TX, USA
- The Framingham Study, Boston, MA, USA
- Department of Neurology, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, USA
| |
Collapse
|
18
|
Ding H, Wang B, Hamel AP, Karjadi C, Ang TFA, Au R, Lin H. Exploring cognitive progression subtypes in the Framingham Heart Study. ALZHEIMER'S & DEMENTIA (AMSTERDAM, NETHERLANDS) 2024; 16:e12574. [PMID: 38515438 PMCID: PMC10955221 DOI: 10.1002/dad2.12574] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/21/2023] [Revised: 02/23/2024] [Accepted: 02/27/2024] [Indexed: 03/23/2024]
Abstract
INTRODUCTION Alzheimer's disease (AD) is a heterogeneous disorder characterized by complex underlying neuropathology that is not fully understood. This study aimed to identify cognitive progression subtypes and examine their correlation with clinical outcomes. METHODS Participants of this study were recruited from the Framingham Heart Study. The Subtype and Stage Inference (SuStaIn) method was used to identify cognitive progression subtypes based on eight cognitive domains. RESULTS Three cognitive progression subtypes were identified, including verbal learning (Subtype 1), abstract reasoning (Subtype 2), and visual memory (Subtype 3). These subtypes represent different domains of cognitive decline during the progression of AD. Significant differences in age of onset among the different subtypes were also observed. A higher SuStaIn stage was significantly associated with increased mortality risk. DISCUSSION This study provides a characterization of AD heterogeneity in cognitive progression, emphasizing the importance of developing personalized approaches for risk stratification and intervention. Highlights We used the Subtype and Stage Inference (SuStaIn) method to identify three cognitive progression subtypes.Different subtypes have significant variations in age of onset.Higher stages of progression are associated with increased mortality risk.
Collapse
Affiliation(s)
- Huitong Ding
- Department of Anatomy and NeurobiologyBoston University Chobanian & Avedisian School of MedicineBostonMassachusettsUSA
- The Framingham Heart StudyBoston University Chobanian & Avedisian School of MedicineBostonMassachusettsUSA
| | - Biqi Wang
- Department of MedicineUniversity of Massachusetts Chan Medical SchoolWorcesterMassachusettsUSA
| | - Alexander P. Hamel
- Department of MedicineUniversity of Massachusetts Chan Medical SchoolWorcesterMassachusettsUSA
| | - Cody Karjadi
- Department of Anatomy and NeurobiologyBoston University Chobanian & Avedisian School of MedicineBostonMassachusettsUSA
- The Framingham Heart StudyBoston University Chobanian & Avedisian School of MedicineBostonMassachusettsUSA
| | - Ting F. A. Ang
- Department of Anatomy and NeurobiologyBoston University Chobanian & Avedisian School of MedicineBostonMassachusettsUSA
- The Framingham Heart StudyBoston University Chobanian & Avedisian School of MedicineBostonMassachusettsUSA
- Slone Epidemiology CenterBoston University Chobanian & Avedisian School of MedicineBostonMassachusettsUSA
| | - Rhoda Au
- Department of Anatomy and NeurobiologyBoston University Chobanian & Avedisian School of MedicineBostonMassachusettsUSA
- The Framingham Heart StudyBoston University Chobanian & Avedisian School of MedicineBostonMassachusettsUSA
- Slone Epidemiology CenterBoston University Chobanian & Avedisian School of MedicineBostonMassachusettsUSA
- Department of EpidemiologyBoston University School of Public HealthBostonMassachusettsUSA
- Departments of Neurology and MedicineBoston University Chobanian & Avedisian School of MedicineBostonMassachusettsUSA
| | - Honghuang Lin
- Department of MedicineUniversity of Massachusetts Chan Medical SchoolWorcesterMassachusettsUSA
| |
Collapse
|
19
|
Ryoo HG, Choi H, Shi K, Rominger A, Lee DY, Lee DS. Distinct subtypes of spatial brain metabolism patterns in Alzheimer's disease identified by deep learning-based FDG PET clusters. Eur J Nucl Med Mol Imaging 2024; 51:443-454. [PMID: 37735259 DOI: 10.1007/s00259-023-06440-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2023] [Accepted: 09/08/2023] [Indexed: 09/23/2023]
Abstract
PURPOSE Alzheimer's disease (AD) is a heterogeneous disease that presents a broad spectrum of clinicopathologic profiles. To date, objective subtyping of AD independent of disease progression using brain imaging has been required. Our study aimed to extract representations of unique brain metabolism patterns different from disease progression to identify objective subtypes of AD. METHODS A total of 3620 FDG brain PET images with AD, mild cognitive impairment (MCI), and cognitively normal (CN) were obtained from the ADNI database from 1607 participants at enrollment and follow-up visits. A conditional variational autoencoder model was trained on FDG brain PET images of AD patients with the corresponding condition of AD severity score. The k-means algorithm was applied to generate clusters from the encoded representations. The trained deep learning-based cluster model was also transferred to FDG PET of MCI patients and predicted the prognosis of subtypes for conversion from MCI to AD. Spatial metabolism patterns, clinical and biological characteristics, and conversion rate from MCI to AD were compared across the subtypes. RESULTS Four distinct subtypes of spatial metabolism patterns in AD with different brain pathologies and clinical profiles were identified: (i) angular, (ii) occipital, (iii) orbitofrontal, and (iv) minimal hypometabolic patterns. The deep learning model was also successfully transferred for subtyping MCI, and significant differences in frequency (P < 0.001) and risk of conversion (log-rank P < 0.0001) from MCI to AD were observed across the subtypes, highest in S2 (35.7%) followed by S1 (23.4%). CONCLUSION We identified distinct subtypes of AD with different clinicopathologic features. The deep learning-based approach to distinguish AD subtypes on FDG PET could have implications for predicting individual outcomes and provide a clue to understanding the heterogeneous pathophysiology of AD.
Collapse
Affiliation(s)
- Hyun Gee Ryoo
- Department of Nuclear Medicine, Seoul National University Bundang Hospital, Seongnam, Republic of Korea
- Department of Nuclear Medicine, Seoul National University Hospital, 101, Daehak-ro, Jongno-gu, Seoul, 03080, Republic of Korea
- Department of Molecular Medicine and Biopharmaceutical Sciences, Graduate School of Convergence Science and Technology, and College of Medicine or College of Pharmacy, Seoul National University, Seoul, Republic of Korea
| | - Hongyoon Choi
- Department of Nuclear Medicine, Seoul National University Hospital, 101, Daehak-ro, Jongno-gu, Seoul, 03080, Republic of Korea.
- Department of Nuclear Medicine, Seoul National University College of Medicine, Seoul, Republic of Korea.
| | - Kuangyu Shi
- Department of Nuclear Medicine, Inselspital, University of Bern, Freiburgstrasse 18, 3010, Bern, Switzerland
| | - Axel Rominger
- Department of Nuclear Medicine, Inselspital, University of Bern, Freiburgstrasse 18, 3010, Bern, Switzerland
| | - Dong Young Lee
- Department of Neuropsychiatry, Seoul National University Hospital, Seoul, Republic of Korea
- Department of Psychiatry, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Dong Soo Lee
- Department of Nuclear Medicine, Seoul National University Hospital, 101, Daehak-ro, Jongno-gu, Seoul, 03080, Republic of Korea
- Department of Molecular Medicine and Biopharmaceutical Sciences, Graduate School of Convergence Science and Technology, and College of Medicine or College of Pharmacy, Seoul National University, Seoul, Republic of Korea
- Department of Nuclear Medicine, Seoul National University College of Medicine, Seoul, Republic of Korea
| |
Collapse
|
20
|
Jeffs QL, Prather JF, Todd WD. Potential neural substrates underlying circadian and olfactory disruptions in preclinical Alzheimer's disease. Front Neurosci 2023; 17:1295998. [PMID: 38094003 PMCID: PMC10716239 DOI: 10.3389/fnins.2023.1295998] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2023] [Accepted: 11/13/2023] [Indexed: 02/01/2024] Open
Abstract
Alzheimer's disease (AD) is the leading cause of dementia, with over 45 million patients worldwide, and poses significant economic and emotional burdens to both patients and caregivers, significantly raising the number of those affected. Unfortunately, much of the existing research on the disease only addresses a small subset of associated symptomologies and pathologies. In this review, we propose to target the earliest stages of the disease, when symptomology first arises. In these stages, before the onset of hallmark symptoms of AD such as cognitive impairments and memory loss, circadian and olfactory disruptions arise and are detectable. Functional similarities between circadian and olfactory systems provide a basis upon which to seek out common mechanisms in AD which may target them early on in the disease. Existing studies of interactions between these systems, while intriguing, leave open the question of the neural substrates underlying them. Potential substrates for such interactions are proposed in this review, such as indirect projections that may functionally connect the two systems and dopaminergic signaling. These substrates may have significant implications for mechanisms underlying disruptions to circadian and olfactory function in early stages of AD. In this review, we propose early detection of AD using a combination of circadian and olfactory deficits and subsequent early treatment of these deficits may provide profound benefits to both patients and caregivers. Additionally, we suggest that targeting research toward the intersection of these two systems in AD could uncover mechanisms underlying the broader set of symptoms and pathologies that currently elude researchers.
Collapse
Affiliation(s)
| | | | - William D. Todd
- Department of Zoology and Physiology, Program in Neuroscience, University of Wyoming, Laramie, WY, United States
| |
Collapse
|
21
|
Sudwarts A, Thinakaran G. Alzheimer's genes in microglia: a risk worth investigating. Mol Neurodegener 2023; 18:90. [PMID: 37986179 PMCID: PMC10662636 DOI: 10.1186/s13024-023-00679-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2023] [Accepted: 11/07/2023] [Indexed: 11/22/2023] Open
Abstract
Despite expressing many key risk genes, the role of microglia in late-onset Alzheimer's disease pathophysiology is somewhat ambiguous, with various phenotypes reported to be either harmful or protective. Herein, we review some key findings from clinical and animal model investigations, discussing the role of microglial genetics in mediating perturbations from homeostasis. We note that impairment to protective phenotypes may include prolonged or insufficient microglial activation, resulting in dysregulated metabolomic (notably lipid-related) processes, compounded by age-related inflexibility in dynamic responses. Insufficiencies of mouse genetics and aggressive transgenic modelling imply severe limitations in applying current methodologies for aetiological investigations. Despite the shortcomings, widely used amyloidosis and tauopathy models of the disease have proven invaluable in dissecting microglial functional responses to AD pathophysiology. Some recent advances have brought modelling tools closer to human genetics, increasing the validity of both aetiological and translational endeavours.
Collapse
Affiliation(s)
- Ari Sudwarts
- Byrd Alzheimer's Center and Research Institute, University of South Florida, Tampa, FL, 33613, USA.
- Department of Molecular Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL, 33612, USA.
| | - Gopal Thinakaran
- Byrd Alzheimer's Center and Research Institute, University of South Florida, Tampa, FL, 33613, USA.
- Department of Molecular Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL, 33612, USA.
| |
Collapse
|
22
|
Hammers DB, Eloyan A, Taurone A, Thangarajah M, Beckett L, Gao S, Kirby K, Aisen P, Dage JL, Foroud T, Griffin P, Grinberg LT, Jack CR, Kramer J, Koeppe R, Kukull WA, Mundada NS, Joie RL, Soleimani-Meigooni DN, Iaccarino L, Murray ME, Nudelman K, Polsinelli AJ, Rumbaugh M, Toga A, Touroutoglou A, Vemuri P, Atri A, Day GS, Duara R, Graff-Radford NR, Honig LS, Jones DT, Masdeu J, Mendez MF, Womack K, Musiek E, Onyike CU, Riddle M, Rogalski E, Salloway S, Sha SJ, Turner RS, Wingo TS, Wolk DA, Carrillo MC, Dickerson BC, Rabinovici GD, Apostolova LG. Profiling baseline performance on the Longitudinal Early-Onset Alzheimer's Disease Study (LEADS) cohort near the midpoint of data collection. Alzheimers Dement 2023; 19 Suppl 9:S8-S18. [PMID: 37256497 PMCID: PMC10806768 DOI: 10.1002/alz.13160] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Revised: 04/12/2023] [Accepted: 04/13/2023] [Indexed: 06/01/2023]
Abstract
OBJECTIVE The Longitudinal Early-Onset Alzheimer's Disease Study (LEADS) seeks to provide comprehensive understanding of early-onset Alzheimer's disease (EOAD; onset <65 years), with the current study profiling baseline clinical, cognitive, biomarker, and genetic characteristics of the cohort nearing the data-collection mid-point. METHODS Data from 371 LEADS participants were compared based on diagnostic group classification (cognitively normal [n = 89], amyloid-positive EOAD [n = 212], and amyloid-negative early-onset non-Alzheimer's disease [EOnonAD; n = 70]). RESULTS Cognitive performance was worse for EOAD than other groups, and EOAD participants were apolipoprotein E (APOE) ε4 homozygotes at higher rates. An amnestic presentation was common among impaired participants (81%), with several clinical phenotypes present. LEADS participants generally consented at high rates to optional trial procedures. CONCLUSIONS We present the most comprehensive baseline characterization of sporadic EOAD in the United States to date. EOAD presents with widespread cognitive impairment within and across clinical phenotypes, with differences in APOE ε4 allele carrier status appearing to be relevant. HIGHLIGHTS Findings represent the most comprehensive baseline characterization of sporadic early-onset Alzheimer's disease (EOAD) to date. Cognitive impairment was widespread for EOAD participants and more severe than other groups. EOAD participants were homozygous apolipoprotein E (APOE) ε4 carriers at higher rates than the EOnonAD group. Amnestic presentation predominated in EOAD and EOnonAD participants, but other clinical phenotypes were present.
Collapse
Affiliation(s)
- Dustin B. Hammers
- Department of Neurology, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Ani Eloyan
- Department of Biostatistics, Center for Statistical Sciences, Brown University, Providence, Rhode Island, USA
| | - Alexander Taurone
- Department of Biostatistics, Center for Statistical Sciences, Brown University, Providence, Rhode Island, USA
| | - Maryanne Thangarajah
- Department of Biostatistics, Center for Statistical Sciences, Brown University, Providence, Rhode Island, USA
| | - Laurel Beckett
- Department of Public Health Sciences, University of California – Davis, Davis, California, USA
| | - Sujuan Gao
- Department of Biostatistics, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Kala Kirby
- Department of Neurology, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Paul Aisen
- Alzheimer’s Therapeutic Research Institute, University of Southern California, San Diego, California, USA
| | - Jeffrey L. Dage
- Department of Neurology, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Tatiana Foroud
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Percy Griffin
- Medical & Scientific Relations Division, Alzheimer’s Association, Chicago, Illinois, USA
| | - Lea T. Grinberg
- Department of Pathology, University of California – San Francisco, San Francisco, California, USA
- Department of Neurology, University of California – San Francisco, San Francisco, California, USA
| | | | - Joel Kramer
- Department of Neurology, University of California – San Francisco, San Francisco, California, USA
| | - Robert Koeppe
- Department of Radiology, University of Michigan, Ann Arbor, Michigan, USA
| | - Walter A. Kukull
- Department of Epidemiology, University of Washington, Seattle, Washington, USA
| | - Nidhi S Mundada
- Department of Neurology, University of California – San Francisco, San Francisco, California, USA
| | - Renaud La Joie
- Department of Neurology, University of California – San Francisco, San Francisco, California, USA
| | | | - Leonardo Iaccarino
- Department of Neurology, University of California – San Francisco, San Francisco, California, USA
| | | | - Kelly Nudelman
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Angelina J. Polsinelli
- Department of Neurology, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Malia Rumbaugh
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Arthur Toga
- Laboratory of Neuro Imaging, USC Stevens Neuroimaging and Informatics Institute, Keck School of Medicine of USC, Los Angeles, California, USA
| | - Alexandra Touroutoglou
- Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | | | - Alireza Atri
- Banner Sun Health Research Institute, Sun City, Arizona, USA
| | - Gregory S. Day
- Department of Neurology, Mayo Clinic, Jacksonville, Florida, USA
| | - Ranjan Duara
- Wien Center for Alzheimer’s Disease and Memory Disorders, Mount Sinai Medical Center, Miami, Florida, USA
| | | | - Lawrence S. Honig
- Taub Institute and Department of Neurology, Columbia University Irving Medical Center, New York, New York, USA
| | - David T. Jones
- Department of Radiology, Mayo Clinic, Rochester, Minnesota, USA
- Department of Neurology, Mayo Clinic, Rochester, Minnesota, USA
| | - Joseph Masdeu
- Nantz National Alzheimer Center, Houston Methodist and Weill Cornell Medicine, Houston, Texas, USA
| | - Mario F. Mendez
- Department of Neurology, David Geffen School of Medicine at UCLA, Los Angeles, California, USA
| | - Kyle Womack
- Department of Neurology, Washington University in St. Louis, St. Louis, Missouri, USA
| | - Erik Musiek
- Department of Neurology, Washington University in St. Louis, St. Louis, Missouri, USA
| | - Chiadi U. Onyike
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Meghan Riddle
- Department of Neurology, Alpert Medical School, Brown University, Providence, Rhode Island, USA
| | - Emily Rogalski
- Department of Psychiatry and Behavioral Sciences, Mesulam Center for Cognitive Neurology and Alzheimer’s Disease, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Steven Salloway
- Department of Neurology, Alpert Medical School, Brown University, Providence, Rhode Island, USA
| | - Sharon J. Sha
- Department of Neurology & Neurological Sciences, Stanford University, Palo Alto, California, USA
| | | | - Thomas S. Wingo
- Department of Neurology and Human Genetics, Emory University School of Medicine, Atlanta, Georgia, USA
| | - David A. Wolk
- Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Maria C. Carrillo
- Medical & Scientific Relations Division, Alzheimer’s Association, Chicago, Illinois, USA
| | - Bradford C. Dickerson
- Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Gil D. Rabinovici
- Department of Neurology, University of California – San Francisco, San Francisco, California, USA
| | - Liana G. Apostolova
- Department of Neurology, Indiana University School of Medicine, Indianapolis, Indiana, USA
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, Indiana, USA
- Department of Radiology and Imaging Sciences, Center for Neuroimaging, Indiana University School of Medicine Indianapolis, Indianapolis, Indiana, USA
| | | |
Collapse
|
23
|
Ray NR, Ayodele T, Jean-Francois M, Baez P, Fernandez V, Bradley J, Crane PK, Dalgard CL, Kuzma A, Nicaretta H, Sims R, Williams J, Cuccaro ML, Pericak-Vance MA, Mayeux R, Wang LS, Schellenberg GD, Cruchaga C, Beecham GW, Reitz C. The Early-Onset Alzheimer's Disease Whole-Genome Sequencing Project: Study design and methodology. Alzheimers Dement 2023; 19:4187-4195. [PMID: 37390458 PMCID: PMC10527497 DOI: 10.1002/alz.13370] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Revised: 06/01/2023] [Accepted: 06/02/2023] [Indexed: 07/02/2023]
Abstract
INTRODUCTION Sequencing efforts to identify genetic variants and pathways underlying Alzheimer's disease (AD) have largely focused on late-onset AD although early-onset AD (EOAD), accounting for ∼10% of cases, is largely unexplained by known mutations, resulting in a lack of understanding of its molecular etiology. METHODS Whole-genome sequencing and harmonization of clinical, neuropathological, and biomarker data of over 5000 EOAD cases of diverse ancestries. RESULTS A publicly available genomics resource for EOAD with extensive harmonized phenotypes. Primary analysis will (1) identify novel EOAD risk loci and druggable targets; (2) assess local-ancestry effects; (3) create EOAD prediction models; and (4) assess genetic overlap with cardiovascular and other traits. DISCUSSION This novel resource complements over 50,000 control and late-onset AD samples generated through the Alzheimer's Disease Sequencing Project (ADSP). The harmonized EOAD/ADSP joint call will be available through upcoming ADSP data releases and will allow for additional analyses across the full onset range. HIGHLIGHTS Sequencing efforts to identify genetic variants and pathways underlying Alzheimer's disease (AD) have largely focused on late-onset AD although early-onset AD (EOAD), accounting for ∼10% of cases, is largely unexplained by known mutations. This results in a significant lack of understanding of the molecular etiology of this devastating form of the disease. The Early-Onset Alzheimer's Disease Whole-genome Sequencing Project is a collaborative initiative to generate a large-scale genomics resource for early-onset Alzheimer's disease with extensive harmonized phenotype data. Primary analyses are designed to (1) identify novel EOAD risk and protective loci and druggable targets; (2) assess local-ancestry effects; (3) create EOAD prediction models; and (4) assess genetic overlap with cardiovascular and other traits. The harmonized genomic and phenotypic data from this initiative will be available through NIAGADS.
Collapse
Affiliation(s)
- Nicholas R. Ray
- Gertrude H. Sergievsky Center, Columbia University, New
York, NY 10032, USA
- Taub Institute for Research on Alzheimer’s Disease
and the Aging Brain, Columbia University, New York, NY 10032, USA
| | - Temitope Ayodele
- Gertrude H. Sergievsky Center, Columbia University, New
York, NY 10032, USA
| | - Melissa Jean-Francois
- The John P. Hussman Institute for Human Genomics,
University of Miami, Miami, FL 33136, USA
- Dr. John T. MacDonald Foundation Department of Human
Genetics, University of Miami, Coral Gables, FL 33146, USA
| | - Penelope Baez
- Gertrude H. Sergievsky Center, Columbia University, New
York, NY 10032, USA
| | - Victoria Fernandez
- Department of Psychiatry, Neurology and Genetics,
Washington University School of Medicine, St. Louis, MO 63130, USA
- Neurogenomics and Informatic (NGI) Center, Washington
University School of Medicine, St. Louis, MO 63130, USA
| | - Joseph Bradley
- Department of Psychiatry, Neurology and Genetics,
Washington University School of Medicine, St. Louis, MO 63130, USA
- Neurogenomics and Informatic (NGI) Center, Washington
University School of Medicine, St. Louis, MO 63130, USA
| | - Paul K. Crane
- Division of General Internal Medicine, University of
Washington, Seattle, WA 98195, USA
| | - Clifton L. Dalgard
- Department of Anatomy, Physiology & Genetics,
Uniformed Services University of the Health Sciences, Bethesda, MD 20814, USA
- The American Genome Center, Uniformed Services University
of the Health Sciences, Bethesda, MD 20814, USA
| | - Amanda Kuzma
- Penn Neurodegeneration Genomics Center, Department of
Pathology and Laboratory Medicine, University of Pennsylvania Perelman School of
Medicine, Philadelphia, PA 19104, USA
| | - Heather Nicaretta
- Penn Neurodegeneration Genomics Center, Department of
Pathology and Laboratory Medicine, University of Pennsylvania Perelman School of
Medicine, Philadelphia, PA 19104, USA
| | - Rebecca Sims
- Division of Psychological Medicine and Clinical
Neurosciences, School of Medicine, Cardiff University, Cardiff CF10 3AT, UK
| | - Julie Williams
- UK Dementia Research Institute, Cardiff University,
Cardiff CF10 3AT, UK
- Division of Psychological Medicine and Clinical
Neurosciences, School of Medicine, Cardiff University, Cardiff CF10 3AT, UK
| | - Michael L. Cuccaro
- The John P. Hussman Institute for Human Genomics,
University of Miami, Miami, FL 33136, USA
- Dr. John T. MacDonald Foundation Department of Human
Genetics, University of Miami, Coral Gables, FL 33146, USA
| | - Margaret A. Pericak-Vance
- The John P. Hussman Institute for Human Genomics,
University of Miami, Miami, FL 33136, USA
- Dr. John T. MacDonald Foundation Department of Human
Genetics, University of Miami, Coral Gables, FL 33146, USA
| | - Richard Mayeux
- Gertrude H. Sergievsky Center, Columbia University, New
York, NY 10032, USA
- Taub Institute for Research on Alzheimer’s Disease
and the Aging Brain, Columbia University, New York, NY 10032, USA
- Department of Neurology, Columbia University, New York, NY
10032, USA
- Department of Epidemiology, Columbia University, New York,
NY 10032, USA
| | - Li-San Wang
- Penn Neurodegeneration Genomics Center, Department of
Pathology and Laboratory Medicine, University of Pennsylvania Perelman School of
Medicine, Philadelphia, PA 19104, USA
| | - Gerard D. Schellenberg
- Penn Neurodegeneration Genomics Center, Department of
Pathology and Laboratory Medicine, University of Pennsylvania Perelman School of
Medicine, Philadelphia, PA 19104, USA
| | - Carlos Cruchaga
- Department of Psychiatry, Neurology and Genetics,
Washington University School of Medicine, St. Louis, MO 63130, USA
- Neurogenomics and Informatic (NGI) Center, Washington
University School of Medicine, St. Louis, MO 63130, USA
| | - Gary W. Beecham
- The John P. Hussman Institute for Human Genomics,
University of Miami, Miami, FL 33136, USA
- Dr. John T. MacDonald Foundation Department of Human
Genetics, University of Miami, Coral Gables, FL 33146, USA
| | - Christiane Reitz
- Gertrude H. Sergievsky Center, Columbia University, New
York, NY 10032, USA
- Taub Institute for Research on Alzheimer’s Disease
and the Aging Brain, Columbia University, New York, NY 10032, USA
- Department of Neurology, Columbia University, New York, NY
10032, USA
- Department of Epidemiology, Columbia University, New York,
NY 10032, USA
| |
Collapse
|
24
|
Sparks S, Pinto J, Hayes G, Spitschan M, Bulte DP. The impact of Alzheimer's disease risk factors on the pupillary light response. Front Neurosci 2023; 17:1248640. [PMID: 37650103 PMCID: PMC10463762 DOI: 10.3389/fnins.2023.1248640] [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: 06/27/2023] [Accepted: 07/31/2023] [Indexed: 09/01/2023] Open
Abstract
Alzheimer's disease (AD) is the leading cause of dementia, and its prevalence is increasing and is expected to continue to increase over the next few decades. Because of this, there is an urgent requirement to determine a way to diagnose the disease, and to target interventions to delay and ideally stop the onset of symptoms, specifically those impacting cognition and daily livelihood. The pupillary light response (PLR) is controlled by the sympathetic and parasympathetic branches of the autonomic nervous system, and impairments to the pupillary light response (PLR) have been related to AD. However, most of these studies that assess the PLR occur in patients who have already been diagnosed with AD, rather than those who are at a higher risk for the disease but without a diagnosis. Determining whether the PLR is similarly impaired in subjects before an AD diagnosis is made and before cognitive symptoms of the disease begin, is an important step before using the PLR as a diagnostic tool. Specifically, identifying whether the PLR is impaired in specific at-risk groups, considering both genetic and non-genetic risk factors, is imperative. It is possible that the PLR may be impaired in association with some risk factors but not others, potentially indicating different pathways to neurodegeneration that could be distinguished using PLR. In this work, we review the most common genetic and lifestyle-based risk factors for AD and identify established relationships between these risk factors and the PLR. The evidence here shows that many AD risk factors, including traumatic brain injury, ocular and intracranial hypertension, alcohol consumption, depression, and diabetes, are directly related to changes in the PLR. Other risk factors currently lack sufficient literature to make any conclusions relating directly to the PLR but have shown links to impairments in the parasympathetic nervous system; further research should be conducted in these risk factors and their relation to the PLR.
Collapse
Affiliation(s)
- Sierra Sparks
- Department of Engineering Science, Institute of Biomedical Engineering, University of Oxford, Oxford, United Kingdom
| | - Joana Pinto
- Department of Engineering Science, Institute of Biomedical Engineering, University of Oxford, Oxford, United Kingdom
| | - Genevieve Hayes
- Department of Engineering Science, Institute of Biomedical Engineering, University of Oxford, Oxford, United Kingdom
| | - Manuel Spitschan
- TUM Department of Sport and Health Sciences (TUM SG), Chronobiology and Health, Technical University of Munich, Munich, Germany
- TUM Institute for Advanced Study (TUM-IAS), Technical University of Munich, Garching, Germany
- Max Planck Institute for Biological Cybernetics, Translational Sensory and Circadian Neuroscience, Tübingen, Germany
| | - Daniel P. Bulte
- Department of Engineering Science, Institute of Biomedical Engineering, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
25
|
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
|
26
|
Polsinelli AJ, Logan PE, Lane KA, Manchella MK, Nemes S, Sanjay AB, Gao S, Apostolova LG. APOE ε4 carrier status and sex differentiate rates of cognitive decline in early- and late-onset Alzheimer's disease. Alzheimers Dement 2023; 19:1983-1993. [PMID: 36394443 PMCID: PMC10182251 DOI: 10.1002/alz.12831] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2022] [Revised: 07/19/2022] [Accepted: 09/19/2022] [Indexed: 11/19/2022]
Abstract
BACKGROUND We studied the effect of apolipoprotein E (APOE) ε4 status and sex on rates of cognitive decline in early- (EO) and late- (LO) onset Alzheimer's disease (AD). METHOD We ran mixed-effects models with longitudinal cognitive measures as dependent variables, and sex, APOE ε4 carrier status, and interaction terms as predictor variables in 998 EOAD and 2562 LOAD participants from the National Alzheimer's Coordinating Center. RESULTS APOE ε4 carriers showed accelerated cognitive decline relative to non-carriers in both EOAD and LOAD, although the patterns of specific cognitive domains that were affected differed. Female participants showed accelerated cognitive decline relative to male participants in EOAD only. The effect of APOE ε4 was greater in EOAD for executive functioning (p < 0.0001) and greater in LOAD for language (p < 0.0001). CONCLUSION We found APOE ε4 effects on cognitive decline in both EOAD and LOAD and female sex in EOAD only. The specific patterns and magnitude of decline are distinct between the two disease variants. HIGHLIGHTS Apolipoprotein E (APOE) ε4 carrier status and sex differentiate rates of cognitive decline in early-onset (EO) and late-onset (LO) Alzheimer's disease (AD). APOE ε4 in EOAD accelerated decline in memory, executive, and processing speed domains. Female sex in EOAD accelerated decline in language, memory, and global cognition. The effect of APOE ε4 was stronger for language in LOAD and for executive function in EOAD. Sex effects on language and executive function decline differed between EOAD and LOAD.
Collapse
Affiliation(s)
- Angelina J. Polsinelli
- Department of Neurology, Indiana University School of Medicine, Indianapolis, Indiana, USA
- Indiana Alzheimer’s Disease Research Center, Indianapolis, Indiana, USA
| | - Paige E. Logan
- Department of Neurology, Indiana University School of Medicine, Indianapolis, Indiana, USA
- Indiana Alzheimer’s Disease Research Center, Indianapolis, Indiana, USA
| | - Kathleen A. Lane
- Department of Biostatistics and Health Data Science, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Mohit K. Manchella
- Department of Chemistry, University of Southern Indiana Evansville, Indiana, USA
| | - Sára Nemes
- Department of Neurology, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | | | - Sujuan Gao
- Indiana Alzheimer’s Disease Research Center, Indianapolis, Indiana, USA
- Department of Biostatistics and Health Data Science, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Liana G. Apostolova
- Department of Neurology, Indiana University School of Medicine, Indianapolis, Indiana, USA
- Indiana Alzheimer’s Disease Research Center, Indianapolis, Indiana, USA
| |
Collapse
|
27
|
Reitz C, Pericak-Vance MA, Foroud T, Mayeux R. A global view of the genetic basis of Alzheimer disease. Nat Rev Neurol 2023; 19:261-277. [PMID: 37024647 PMCID: PMC10686263 DOI: 10.1038/s41582-023-00789-z] [Citation(s) in RCA: 63] [Impact Index Per Article: 31.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/21/2023] [Indexed: 04/08/2023]
Abstract
The risk of Alzheimer disease (AD) increases with age, family history and informative genetic variants. Sadly, there is still no cure or means of prevention. As in other complex diseases, uncovering genetic causes of AD could identify underlying pathological mechanisms and lead to potential treatments. Rare, autosomal dominant forms of AD occur in middle age as a result of highly penetrant genetic mutations, but the most common form of AD occurs later in life. Large-scale, genome-wide analyses indicate that 70 or more genes or loci contribute to AD. One of the major factors limiting progress is that most genetic data have been obtained from non-Hispanic white individuals in Europe and North America, preventing the development of personalized approaches to AD in individuals of other ethnicities. Fortunately, emerging genetic data from other regions - including Africa, Asia, India and South America - are now providing information on the disease from a broader range of ethnicities. Here, we summarize the current knowledge on AD genetics in populations across the world. We predominantly focus on replicated genetic discoveries but also include studies in ethnic groups where replication might not be feasible. We attempt to identify gaps that need to be addressed to achieve a complete picture of the genetic and molecular factors that drive AD in individuals across the globe.
Collapse
Affiliation(s)
- Christiane Reitz
- The Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University, New York, NY, USA
- The Gertrude H. Sergievsky Center, Columbia University, New York, NY, USA
- Department of Neurology, Columbia University, New York, NY, USA
- Department of Epidemiology, Columbia University, New York, NY, USA
| | - Margaret A Pericak-Vance
- The John P. Hussman Institute for Human Genomics, University of Miami Miller School of Medicine, Miami, FL, USA
- The Dr. John T. Macdonald Foundation Department of Human Genetics, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Tatiana Foroud
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN, USA
- National Centralized Repository for Alzheimer's Disease and Related Dementias, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Richard Mayeux
- The Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University, New York, NY, USA.
- The Gertrude H. Sergievsky Center, Columbia University, New York, NY, USA.
- Department of Neurology, Columbia University, New York, NY, USA.
- Department of Epidemiology, Columbia University, New York, NY, USA.
| |
Collapse
|
28
|
Liang Z, Wu Y, Li C, Liu Z. Clinical and genetic characteristics in a central-southern Chinese cohort of early-onset Alzheimer's disease. Front Neurol 2023; 14:1119326. [PMID: 37051054 PMCID: PMC10084792 DOI: 10.3389/fneur.2023.1119326] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2022] [Accepted: 02/21/2023] [Indexed: 03/29/2023] Open
Abstract
BackgroundMutations in the presenilin-1 (PSEN1), presenilin-2 (PSEN2), and amyloid precursor protein (APP) genes have been commonly identified in early-onset Alzheimer's disease (EOAD). Some of the mutations in the three causative genes, especially the PSEN1 gene, result in variable phenotypes and exhibit clinical heterogeneity among EOAD families.MethodsUsing next-generation sequencing (NGS), we performed genetic screening in a Chinese cohort of 18 patients with EOAD, consisting of five familial EOAD and 13 sporadic cases.ResultsWe identified two likely pathogenic PSEN1 mutations (one novel) and a novel APP mutation in three cases of EOAD, where two are familial and one is sporadic, respectively. In addition, we detected a few variants of uncertain significance (VUS) in several genes, including not only the two known variants in PSEN2 (p.H169N and p.V214L) but also genes causal of other types of dementia or previously identified as risk factors for AD, suggesting the possible involvement of multiple genes in the etiopathology of AD. The patients carrying PSEN1 mutations had an earlier mean age at the onset than those with PSEN2 or APP variants. The initial symptoms varied greatly among patients in the EOAD cohort, from progressive memory impairment and epilepsy to uncommon motor symptoms such as involuntary tremors in the upper extremities.ConclusionsIn conclusion, our study provides further evidence of the genetic profile of patients with EOAD from China and expands the mutation spectrum of both PSEN1 and APP. In addition, our results highlight the clinical heterogeneity in patients with EOAD and mutations in PSEN1, PSEN2, and APP and suggest strong effects of genetic variants on clinical phenotypes. Future functional studies are needed to clarify the interaction between AD-causative gene mutations and phenotypic heterogeneity.
Collapse
Affiliation(s)
- Zhihou Liang
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yan Wu
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Chuanzhou Li
- Department of Medical Genetics, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Chuanzhou Li
| | - Zhijun Liu
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- *Correspondence: Zhijun Liu
| |
Collapse
|
29
|
Forno G, Contador J, Pérez-Millan A, Guillen N, Falgàs N, Sarto J, Tort-Merino A, Castellví M, Bosch B, Fernández-Villullas G, Balasa M, Antonell A, Sala-Llonch R, Sanchez-Valle R, Hornberger M, Lladó A. The APOE4 effect: structural brain differences in Alzheimer's disease according to the age at symptom onset. Eur J Neurol 2023; 30:597-605. [PMID: 36463489 PMCID: PMC10108138 DOI: 10.1111/ene.15657] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Revised: 11/17/2022] [Accepted: 12/01/2022] [Indexed: 12/07/2022]
Abstract
BACKGROUND AND PURPOSE How the APOE genotype can differentially affect cortical and subcortical memory structures in biomarker-confirmed early-onset (EOAD) and late-onset (LOAD) Alzheimer's disease (AD) was assessed. METHOD Eighty-seven cerebrospinal fluid (CSF) biomarker-confirmed AD patients were classified according to their APOE genotype and age at onset. 28 were EOAD APOE4 carriers (+EOAD), 21 EOAD APOE4 non-carriers (-EOAD), 23 LOAD APOE4 carriers (+LOAD) and 15 LOAD APOE4 non-carriers (-LOAD). Grey matter (GM) volume differences were analyzed using voxel-based morphometry in Papez circuit regions. Multiple regression analyses were performed to determine the relation between GM volume loss and cognition. RESULTS Significantly more mammillary body atrophy in +EOAD compared to -EOAD is reported. The medial temporal and posterior cingulate cortex showed less GM in +LOAD compared to -LOAD. Medial temporal GM volume loss was also found in +EOAD compared to -LOAD. With an exception for +EOAD, medial temporal GM was strongly associated with episodic memory in the three groups, whilst posterior cingulate cortex GM volume was more related with visuospatial abilities. Visuospatial abilities and episodic memory were also associated with the anterior thalamic nucleus in -LOAD. CONCLUSIONS Our results show that the APOE genotype has a significant effect on GM integrity as a function of age of disease onset. Specifically, whilst LOAD APOE4 genotype is mostly associated with increased medial temporal and parietal atrophy compared to -LOAD, for EOAD APOE4 might have a more specific effect on subcortical (mammillary body) structures. The findings suggest that APOE genotype needs to be taken into account when classifying patients by age at onset.
Collapse
Affiliation(s)
- Gonzalo Forno
- Alzheimer's Disease and Other Cognitive Disorders Unit, Neurology Service, Hospital Clínic of Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Universitat de Barcelona, Barcelona, Spain
- School of Psychology, Universidad de los Andes, Santiago, Chile
| | - Jose Contador
- Alzheimer's Disease and Other Cognitive Disorders Unit, Neurology Service, Hospital Clínic of Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Universitat de Barcelona, Barcelona, Spain
| | - Agnès Pérez-Millan
- Alzheimer's Disease and Other Cognitive Disorders Unit, Neurology Service, Hospital Clínic of Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Universitat de Barcelona, Barcelona, Spain
- Institute of Neurosciences. Department of Biomedicine, Faculty of Medicine, University of Barcelona, Barcelona, Spain
| | - Nuria Guillen
- Alzheimer's Disease and Other Cognitive Disorders Unit, Neurology Service, Hospital Clínic of Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Universitat de Barcelona, Barcelona, Spain
| | - Neus Falgàs
- Alzheimer's Disease and Other Cognitive Disorders Unit, Neurology Service, Hospital Clínic of Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Universitat de Barcelona, Barcelona, Spain
- Atlantic Fellow for Equity in Brain Health, Global Brain Heath Institute, University of California, San Francisco, California, USA
| | - Jordi Sarto
- Alzheimer's Disease and Other Cognitive Disorders Unit, Neurology Service, Hospital Clínic of Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Universitat de Barcelona, Barcelona, Spain
| | - Adrià Tort-Merino
- Alzheimer's Disease and Other Cognitive Disorders Unit, Neurology Service, Hospital Clínic of Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Universitat de Barcelona, Barcelona, Spain
| | - Magdalena Castellví
- Alzheimer's Disease and Other Cognitive Disorders Unit, Neurology Service, Hospital Clínic of Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Universitat de Barcelona, Barcelona, Spain
| | - Beatriz Bosch
- Alzheimer's Disease and Other Cognitive Disorders Unit, Neurology Service, Hospital Clínic of Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Universitat de Barcelona, Barcelona, Spain
| | - Guadalupe Fernández-Villullas
- Alzheimer's Disease and Other Cognitive Disorders Unit, Neurology Service, Hospital Clínic of Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Universitat de Barcelona, Barcelona, Spain
| | - Mircea Balasa
- Alzheimer's Disease and Other Cognitive Disorders Unit, Neurology Service, Hospital Clínic of Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Universitat de Barcelona, Barcelona, Spain
- Atlantic Fellow for Equity in Brain Health, Global Brain Heath Institute, University of California, San Francisco, California, USA
- Atlantic Fellow for Equity in Brain Health, Global Brain Heath Institute, Trinity College Dublin, Dublin, Irland
| | - Anna Antonell
- Alzheimer's Disease and Other Cognitive Disorders Unit, Neurology Service, Hospital Clínic of Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Universitat de Barcelona, Barcelona, Spain
| | - Roser Sala-Llonch
- Alzheimer's Disease and Other Cognitive Disorders Unit, Neurology Service, Hospital Clínic of Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Universitat de Barcelona, Barcelona, Spain
- Institute of Neurosciences. Department of Biomedicine, Faculty of Medicine, University of Barcelona, Barcelona, Spain
- Biomedical Imaging Group, Biomedical Research Networking Center in Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Barcelona, Spain
| | - Raquel Sanchez-Valle
- Alzheimer's Disease and Other Cognitive Disorders Unit, Neurology Service, Hospital Clínic of Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Universitat de Barcelona, Barcelona, Spain
| | | | - Albert Lladó
- Alzheimer's Disease and Other Cognitive Disorders Unit, Neurology Service, Hospital Clínic of Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Universitat de Barcelona, Barcelona, Spain
| |
Collapse
|
30
|
Affiliation(s)
- George Perry
- Department of Neuroscience, Developmental and Regenerative Biology, University of Texas at San Antonio, San Antonio, TX, USA
| |
Collapse
|
31
|
Kang JM, Shin JH, Kim WR, Seo S, Seo H, Lee SY, Park KH, Na DL, Okamura N, Seong JK, Noh Y. Effects of the APOEɛ4 Allele on the Relationship Between Tau and Amyloid-β in Early- and Late-Onset Alzheimer's Disease. J Alzheimers Dis 2023; 94:1233-1246. [PMID: 37393505 DOI: 10.3233/jad-230339] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/03/2023]
Abstract
BACKGROUND Little is known regarding the differential effects of the apolipoprotein E (APOE) ɛ4 on the regional topography of amyloid and tau in patients with both early-onset (EOAD) and late-onset Alzheimer's disease (LOAD). OBJECTIVE To compare the distribution and association of tau, amyloid, and cortical thickness among groups classified by the presence of APOEɛ4 allele and onset age. METHODS A total of 165 participants including 54 EOAD patients (29 ɛ4-; 25 ɛ4+), 45 LOAD patients (21 ɛ4-; 24 ɛ4+), and 66 age-matched controls underwent 3T MRI, 18F-THK5351 (THK) and 18F-flutemetamol (FLUTE) PET scans, APOE genotyping, and neuropsychological tests. Data for voxel-wise and standardized uptake values from PET scans were analyzed in the context of APOE and age at onset. RESULTS EOAD ɛ4- patients showed greater THK retention in the association cortices, whereas their EOAD ɛ4+ counterparts had more retention in medial temporal areas. THK topography of LOAD ɛ4+ was similar to EOAD ɛ4 + . THK correlated positively with FLUTE and conversely with mean cortical thickness, being lowest in EOAD ɛ4-, highest in LOAD ɛ4-, and modest in ɛ4+ groups. Even in the APOEɛ4+ groups, THK tended to correlate with FLUTE and mean cortical thickness in the inferior parietal region in EOAD and in the medial temporal region in LOAD. LOAD ɛ4- manifested with prevalent small vessel disease markers and the lowest correlation between THK retention and cognition. CONCLUSION Our observations suggest the differential effects of the APOEɛ4 on the relationship between tau and amyloid in EOAD and LOAD.
Collapse
Affiliation(s)
- Jae Myeong Kang
- Department of Psychiatry, Gil Medical Center, Gachon University College of Medicine, Incheon, Republic of Korea
| | - Jeong-Hyeon Shin
- School of Biomedical Engineering, Korea University, Seoul, Republic of Korea
- Bio Medical Research Center, Bio Medical & Health Division, Korea Testing Laboratory, Daegu, Republic of Korea
| | - Woo-Ram Kim
- Neuroscience Research Institute, Gachon University, Incheon, Republic of Korea
| | - Seongho Seo
- Neuroscience Research Institute, Gachon University, Incheon, Republic of Korea
| | - Haeun Seo
- Neuroscience Research Institute, Gachon University, Incheon, Republic of Korea
| | - Sang-Yoon Lee
- Department of Neuroscience, College of Medicine, Gachon University, Incheon, Republic of Korea
| | - Kee Hyung Park
- Department of Neurology, Gil Medical Center, Gachon University College of Medicine, Incheon, Republic of Korea
| | - Duk L Na
- Department of Neurology, Samsung Medical Center, Sungkyunkwan University School of Medicine Seoul, Republic of Korea; Happymind Clinic, Seoul, Republic of Korea
| | - Nobuyuki Okamura
- Division of Pharmacology, Faculty of Medicine, Tohoku Medical and Pharmaceutical University, Sendai, Japan
| | - Joon-Kyoung Seong
- School of Biomedical Engineering, Korea University, Seoul, Republic of Korea
- Department of Artificial Intelligence, Korea University, Seoul, Republic of Korea
| | - Young Noh
- Department of Neurology, Gil Medical Center, Gachon University College of Medicine, Incheon, Republic of Korea
- Department of Health Science and Technology, GAIHST, Gachon University, Incheon, Republic of Korea
| |
Collapse
|
32
|
Huang Y, Xu J, Zhang X, Liu Y, Yu E. Research progress on vestibular dysfunction and visual-spatial cognition in patients with Alzheimer's disease. Front Aging Neurosci 2023; 15:1153918. [PMID: 37151847 PMCID: PMC10158930 DOI: 10.3389/fnagi.2023.1153918] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Accepted: 04/03/2023] [Indexed: 05/09/2023] Open
Abstract
Alzheimer's disease (AD) or vestibular dysfunction may impair visual-spatial cognitive function. Recent studies have shown that vestibular dysfunction is increasingly common in patients with AD, and patients with AD with vestibular impairment show more visual-spatial cognitive impairment. By exploring the relationship and interaction mechanism among the vestibular system, visual-spatial cognitive ability, and AD, this study aims to provide new insights for the screening, diagnosis, and rehabilitation intervention of patients with AD. In contrast, routine vestibular function tests are particularly important for understanding the vestibular function of patients with AD. The efficacy of vestibular function test as a tool for the early screening of patients with AD must also be further studied. Through the visual-spatial cognitive ability test, the "spatial impairment" subtype of patients with AD, which may be significant in caring for patients with AD to prevent loss and falls, can also be determined. Additionally, the visual-spatial cognitive ability test has great benefits in preventing and alleviating cognitive decline of patients with AD.
Collapse
Affiliation(s)
- Yan Huang
- School of Medical Technology and Information Engineering, Zhejiang Chinese Medical University, Hangzhou, China
| | - Jiaxi Xu
- The Second School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, China
| | - Xuehao Zhang
- School of Medical Technology and Information Engineering, Zhejiang Chinese Medical University, Hangzhou, China
| | - Yuhe Liu
- Department of Otolaryngology, Head and Neck Surgery, Beijing Friendship Hospital, Capital Medical University, Beijing, China
- *Correspondence: Yuhe Liu,
| | - Enyan Yu
- Cancer Hospital of the University of Chinese Academy of Sciences, Zhejiang Cancer Hospital, Hangzhou, China
- Enyan Yu,
| |
Collapse
|
33
|
Puris E, Saveleva L, de Sousa Maciel I, Kanninen KM, Auriola S, Fricker G. Protein Expression of Amino Acid Transporters Is Altered in Isolated Cerebral Microvessels of 5xFAD Mouse Model of Alzheimer's Disease. Mol Neurobiol 2023; 60:732-748. [PMID: 36367657 PMCID: PMC9849299 DOI: 10.1007/s12035-022-03111-y] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Accepted: 10/26/2022] [Indexed: 11/13/2022]
Abstract
Membrane transporters such as ATP-binding cassette (ABC) and solute carrier (SLC) transporters expressed at the neurovascular unit (NVU) play an important role in drug delivery to the brain and have been demonstrated to be involved in Alzheimer's disease (AD) pathogenesis. However, our knowledge of quantitative changes in transporter absolute protein expression and functionality in vivo in NVU in AD patients and animal models is limited. The study aim was to investigate alterations in protein expression of ABC and SLC transporters in the isolated brain microvessels and brain prefrontal cortices of a widely used model of familial AD, 5xFAD mice (8 months old), using a sensitive liquid chromatography tandem mass spectrometry-based quantitative targeted absolute proteomic approach. Moreover, we examined alterations in brain prefrontal cortical and plasmatic levels of transporter substrates in 5xFAD mice compared to age-matched wild-type (WT) controls. ASCT1 (encoded by Slc1a4) protein expression in the isolated brain microvessels and brain prefrontal cortices of 5xFAD mice was twice higher compared to WT controls (p = 0.01). Brain cortical levels of ASCT1 substrate, serine, were increased in 5xFAD mice compared to WT animals. LAT1 (encoded by Slc7a5) and 4F2hc (encoded by Slc3a2) protein expressions were significantly altered in the isolated brain microvessels of 5xFAD mice compared to WT controls (p = 0.008 and p = 0.05, respectively). Overall, the study provides important information, which is crucial for the optimal use of the 5xFAD mouse model in AD drug development and for investigating novel drug delivery approaches. In addition, the findings of the study shed light on the novel potential mechanisms underlying AD pathogenesis.
Collapse
Affiliation(s)
- Elena Puris
- Institute of Pharmacy and Molecular Biotechnology, Ruprecht-Karls-University, Im Neuenheimer Feld 329, 69120 Heidelberg, Germany
| | - Liudmila Saveleva
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, P.O. Box 1627, 70211 Kuopio, Finland
| | - Izaque de Sousa Maciel
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, P.O. Box 1627, 70211 Kuopio, Finland
| | - Katja M. Kanninen
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, P.O. Box 1627, 70211 Kuopio, Finland
| | - Seppo Auriola
- School of Pharmacy, University of Eastern Finland, P.O. Box 1627, 70211 Kuopio, Finland
| | - Gert Fricker
- Institute of Pharmacy and Molecular Biotechnology, Ruprecht-Karls-University, Im Neuenheimer Feld 329, 69120 Heidelberg, Germany
| |
Collapse
|
34
|
Park D, Son KJ, Jeong E, Kim H, Lee SY, Kim JH, Kim HS. Effects of Socioeconomic Status and Residence Areas on Long-Term Survival in Patients With Early-Onset Dementia: The Korean National Health Insurance Service Database Study. J Korean Med Sci 2022; 37:e354. [PMID: 36536548 PMCID: PMC9763705 DOI: 10.3346/jkms.2022.37.e354] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Accepted: 11/30/2022] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Early-onset dementia (EOD) is still insufficiently considered for healthcare policies. We investigated the effect of socio-environmental factors on the long-term survival of patients with EOD. METHODS This retrospective cohort study utilized the Korean National Health Insurance Database from 2007 to 2018. We enrolled 3,825 patients aged 40 to 65 years old with all types of dementia newly diagnosed in 2009 as EOD cases. We defined socioeconomic status using the national health insurance premium (NHIP) levels. Residential areas were classified into capital, metropolitan, city, and county levels. All-cause mortality was the primary outcome. Kaplan-Meier curves and log-rank tests were employed. Further, Cox-proportional hazards models were established. RESULTS The mean survival of the fourth NHIP level group was 96.31 ± 1.20 months, whereas that of the medical-aid group was 85.53 ± 1.30 months (P < 0.001). The patients living in the capital had a mean survival of 95.73 ± 1.34 months, whereas those living in the county had 89.66 ± 1.75 months (P = 0.035). In the Cox-proportional hazards model, the medical-aid (adjusted hazard ratio [aHR], 1.67; P < 0.001), first NHIP level (aHR, 1.26; P = 0.012), and second NHIP level (aHR, 1.26; P = 0.008) groups were significantly associated with a higher long-term mortality risk. The capital residents exhibited a significantly lower long-term mortality risk than did the county residents (aHR, 0.82; P = 0.041). CONCLUSION Socioeconomic status and residential area are associated with long-term survival in patients with EOD. This study provides a rational basis for establishing a healthcare policy for patients with EOD.
Collapse
Affiliation(s)
- Dougho Park
- Department of Rehabilitation Medicine, Pohang Stroke and Spine Hospital, Pohang, Korea
- Department of Medical Science and Engineering, School of Convergence Science and Technology, Pohang University of Science and Technology, Pohang, Korea
| | - Kang Ju Son
- Department of Research and Analysis, National Health Insurance Service Ilsan Hospital, Goyang, Korea
- Department of Biostatistics and Computing, Yonsei University Graduate School, Seoul, Korea
| | - Eunhwan Jeong
- Department of Neurology, Pohang Stroke and Spine Hospital, Pohang, Korea
| | - Haejong Kim
- Department of Neurology, Pohang Stroke and Spine Hospital, Pohang, Korea
| | - Su Yun Lee
- Department of Neurology, Pohang Stroke and Spine Hospital, Pohang, Korea
| | - Jong Hun Kim
- Department of Neurology, National Health Insurance Service Ilsan Hospital, Goyang, Korea.
| | - Hyoung Seop Kim
- Department of Physical Medicine and Rehabilitation, National Health Insurance Service Ilsan Hospital, Goyang, Korea.
| |
Collapse
|
35
|
Arezoumandan S, Xie SX, Cousins KAQ, Mechanic-Hamilton DJ, Peterson CS, Huang CY, Ohm DT, Ittyerah R, McMillan CT, Wolk DA, Yushkevich P, Trojanowski JQ, Lee EB, Grossman M, Phillips JS, Irwin DJ. Regional distribution and maturation of tau pathology among phenotypic variants of Alzheimer's disease. Acta Neuropathol 2022; 144:1103-1116. [PMID: 35871112 PMCID: PMC9936795 DOI: 10.1007/s00401-022-02472-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Revised: 07/02/2022] [Accepted: 07/14/2022] [Indexed: 01/26/2023]
Abstract
Alzheimer's disease neuropathologic change (ADNC) is clinically heterogenous and can present with a classic multidomain amnestic syndrome or focal non-amnestic syndromes. Here, we investigated the distribution and burden of phosphorylated and C-terminally cleaved tau pathologies across hippocampal subfields and cortical regions among phenotypic variants of Alzheimer's disease (AD). In this study, autopsy-confirmed patients with ADNC, were classified into amnestic (aAD, N = 40) and non-amnestic (naAD, N = 39) groups based on clinical criteria. We performed digital assessment of tissue sections immunostained for phosphorylated-tau (AT8 detects pretangles and mature tangles), D421-truncated tau (TauC3, a marker for mature tangles and ghost tangles), and E391-truncated tau (MN423, a marker that primarily detects ghost tangles), in hippocampal subfields and three cortical regions. Linear mixed-effect models were used to test regional and group differences while adjusting for demographics. Both groups showed AT8-reactivity across hippocampal subfields that mirrored traditional Braak staging with higher burden of phosphorylated-tau in subregions implicated as affected early in Braak staging. The burden of phosphorylated-tau and TauC3-immunoreactive tau in the hippocampus was largely similar between the aAD and naAD groups. In contrast, the naAD group had lower relative distribution of MN423-reactive tangles in CA1 (β = - 0.2, SE = 0.09, p = 0.001) and CA2 (β = - 0.25, SE = 0.09, p = 0.005) compared to the aAD. While the two groups had similar levels of phosphorylated-tau pathology in cortical regions, there was higher burden of TauC3 reactivity in sup/mid temporal cortex (β = 0.16, SE = 0.07, p = 0.02) and MN423 reactivity in all cortical regions (β = 0.4-0.43, SE = 0.09, p < 0.001) in the naAD compared to aAD. In conclusion, AD clinical variants may have a signature distribution of overall phosphorylated-tau pathology within the hippocampus reflecting traditional Braak staging; however, non-amnestic AD has greater relative mature tangle pathology in the neocortex compared to patients with clinical amnestic AD, where the hippocampus had greatest relative burden of C-terminally cleaved tau reactivity. Thus, varying neuronal susceptibility to tau-mediated neurodegeneration may influence the clinical expression of ADNC.
Collapse
Affiliation(s)
- Sanaz Arezoumandan
- Digital Neuropathology Laboratory, Department of Neurology, Perelman School of Medicine, Philadelphia, PA, 19104, USA
- Penn Frontotemporal Degeneration Center, Perelman School of Medicine, Philadelphia, PA, 19104, USA
| | - Sharon X Xie
- Department of Biostatistics, Epidemiology, and Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Katheryn A Q Cousins
- Penn Frontotemporal Degeneration Center, Perelman School of Medicine, Philadelphia, PA, 19104, USA
| | - Dawn J Mechanic-Hamilton
- Department of Neurology, Penn Memory Center, Perelman School of Medicine, Philadelphia, PA, 19104, USA
- Department of Neurology, Penn Alzheimer's Disease Research Center, Perelman School of Medicine, Philadelphia, PA, 19104, USA
| | - Claire S Peterson
- Digital Neuropathology Laboratory, Department of Neurology, Perelman School of Medicine, Philadelphia, PA, 19104, USA
- Penn Frontotemporal Degeneration Center, Perelman School of Medicine, Philadelphia, PA, 19104, USA
| | - Camille Y Huang
- Digital Neuropathology Laboratory, Department of Neurology, Perelman School of Medicine, Philadelphia, PA, 19104, USA
| | - Daniel T Ohm
- Digital Neuropathology Laboratory, Department of Neurology, Perelman School of Medicine, Philadelphia, PA, 19104, USA
- Penn Frontotemporal Degeneration Center, Perelman School of Medicine, Philadelphia, PA, 19104, USA
| | - Ranjit Ittyerah
- Penn Image Computing and Science Lab, Department of Radiology, Perelman School of Medicine, Philadelphia, PA, 19104, USA
| | - Corey T McMillan
- Penn Frontotemporal Degeneration Center, Perelman School of Medicine, Philadelphia, PA, 19104, USA
- Department of Neurology, Penn Alzheimer's Disease Research Center, Perelman School of Medicine, Philadelphia, PA, 19104, USA
| | - David A Wolk
- Department of Neurology, Penn Memory Center, Perelman School of Medicine, Philadelphia, PA, 19104, USA
- Department of Neurology, Penn Alzheimer's Disease Research Center, Perelman School of Medicine, Philadelphia, PA, 19104, USA
| | - Paul Yushkevich
- Department of Neurology, Penn Alzheimer's Disease Research Center, Perelman School of Medicine, Philadelphia, PA, 19104, USA
- Penn Image Computing and Science Lab, Department of Radiology, Perelman School of Medicine, Philadelphia, PA, 19104, USA
| | - John Q Trojanowski
- Department of Neurology, Penn Alzheimer's Disease Research Center, Perelman School of Medicine, Philadelphia, PA, 19104, USA
- Center for Neurodegenerative Disease Research, Department of Pathology and Laboratory Medicine, Perelman School of Medicine, Philadelphia, PA, 19104, USA
| | - Edward B Lee
- Department of Neurology, Penn Alzheimer's Disease Research Center, Perelman School of Medicine, Philadelphia, PA, 19104, USA
- Center for Neurodegenerative Disease Research, Department of Pathology and Laboratory Medicine, Perelman School of Medicine, Philadelphia, PA, 19104, USA
- Translational Neuropathology Research Laboratory, Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Murray Grossman
- Penn Frontotemporal Degeneration Center, Perelman School of Medicine, Philadelphia, PA, 19104, USA
| | - Jeffrey S Phillips
- Penn Frontotemporal Degeneration Center, Perelman School of Medicine, Philadelphia, PA, 19104, USA
| | - David J Irwin
- Digital Neuropathology Laboratory, Department of Neurology, Perelman School of Medicine, Philadelphia, PA, 19104, USA.
- Penn Frontotemporal Degeneration Center, Perelman School of Medicine, Philadelphia, PA, 19104, USA.
| |
Collapse
|
36
|
Thu NT, Graff-Radford J, Machulda MM, Spychalla AJ, Schwarz CG, Senjem ML, Lowe VJ, Vemuri P, Kantarci K, Knopman DS, Petersen RC, Jack CR, Josephs KA, Whitwell JL. Regional white matter hyperintensities in posterior cortical atrophy and logopenic progressive aphasia. Neurobiol Aging 2022; 119:46-55. [PMID: 35970009 PMCID: PMC9886198 DOI: 10.1016/j.neurobiolaging.2022.07.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Revised: 07/13/2022] [Accepted: 07/23/2022] [Indexed: 02/01/2023]
Abstract
White matter hyperintensities (WMH) are markers of cerebral small vessel disease and are associated with higher risk of typical amnestic Alzheimer's disease (tAD). Little is known about the frequency and distribution of WMH in atypical variants of AD, including logopenic progressive aphasia (LPA) and posterior cortical atrophy (PCA). We investigated WMHs in 75 LPA, 39 PCA, and 50 tAD patients and associations with age, beta-amyloid PET burden, and cognition. PCA had greater subcortical WMHs in right occipital, parietal, and temporal lobes compared to LPA, and greater parieto-occipital subcortical and occipital periventricular WMHs than tAD. LPA had greater subcortical WMHs in left parietal lobe and deep white matter WMHs than PCA, and greater fronto-occipital subcortical and occipital periventricular WMHs than tAD. Total WMH increased with increasing age but was not related to beta-amyloid burden. Greater WMH was associated with visuoperceptual performance in LPA and PCA after correcting for atrophy. WMH topography differs across AD variants. Further work is needed to determine whether they reflect cerebrovascular disease or regionally specific neurodegenerative changes.
Collapse
Affiliation(s)
- Nha Trang Thu
- Department of Radiology, Mayo Clinic, Rochester, MN, USA
| | | | - Mary M. Machulda
- Department of Psychiatry and Psychology, Mayo Clinic, Rochester, MN, USA
| | | | | | | | - Val J. Lowe
- Department of Radiology, Mayo Clinic, Rochester, MN, USA
| | | | - Kejal Kantarci
- Department of Radiology, Mayo Clinic, Rochester, MN, USA
| | | | | | | | | | | |
Collapse
|
37
|
Regmi S, Liu DD, Shen M, Kevadiya BD, Ganguly A, Primavera R, Chetty S, Yarani R, Thakor AS. Mesenchymal stromal cells for the treatment of Alzheimer’s disease: Strategies and limitations. Front Mol Neurosci 2022; 15:1011225. [PMID: 36277497 PMCID: PMC9584646 DOI: 10.3389/fnmol.2022.1011225] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2022] [Accepted: 09/20/2022] [Indexed: 11/27/2022] Open
Abstract
Alzheimer’s disease (AD) is a major cause of age-related dementia and is characterized by progressive brain damage that gradually destroys memory and the ability to learn, which ultimately leads to the decline of a patient’s ability to perform daily activities. Although some of the pharmacological treatments of AD are available for symptomatic relief, they are not able to limit the progression of AD and have several side effects. Mesenchymal stem/stromal cells (MSCs) could be a potential therapeutic option for treating AD due to their immunomodulatory, anti-inflammatory, regenerative, antioxidant, anti-apoptotic, and neuroprotective effects. MSCs not only secret neuroprotective and anti-inflammatory factors to promote the survival of neurons, but they also transfer functional mitochondria and miRNAs to boost their bioenergetic profile as well as improve microglial clearance of accumulated protein aggregates. This review focuses on different clinical and preclinical studies using MSC as a therapy for treating AD, their outcomes, limitations and the strategies to potentiate their clinical translation.
Collapse
|
38
|
Fray S, Achouri-Rassas A, Belal S, Messaoud T. Missing apolipoprotein E ɛ4 allele associated with nonamnestic Alzheimer’s disease in a Tunisian population. J Genet 2022. [DOI: 10.1007/s12041-022-01384-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/14/2022]
|
39
|
Bolton CJ, Tam JW. Differential Involvement of the Locus Coeruleus in Early- and Late-Onset Alzheimer's Disease: A Potential Mechanism of Clinical Differences? J Geriatr Psychiatry Neurol 2022; 35:733-739. [PMID: 34496652 PMCID: PMC12023724 DOI: 10.1177/08919887211044755] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
Sporadic early-onset Alzheimer's disease (sEOAD) is often associated with atypical clinical features, yet the cause of this heterogeneity remains unclear. This study investigated post-mortem atrophy of the locus coeruleus (LC) in sEOAD and late-onset Alzheimer's disease (LOAD). Levels of LC atrophy, as estimated by pathologist-rating of hypopigmentation, were compared between sEOAD (n = 115) and LOAD (n = 672) participants while controlling for other measures of pathological progression. Subsequent analyses compared low vs. high LC atrophy sEOAD subgroups on neuropsychological test performance. Results show nearly 4 times greater likelihood of higher LC atrophy in sEOAD as compared to LOAD (p < .005). sEOAD participants with greater LC atrophy displayed significantly worse performance on various baseline measures of attentional functioning (p < .05), despite similar global cognition (p = .25). These findings suggest the LC is an important potential driver of clinical and pathological heterogeneity in sEOAD.
Collapse
Affiliation(s)
- Corey J. Bolton
- Vanderbilt Memory and Alzheimer’s Center, Vanderbilt University Medical Center, Nashville, TN, USA
- Department of Neurology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Joyce W. Tam
- Department of Psychiatry and Behavioral Sciences, Rush University Medical Center, Chicago, IL, USA
| |
Collapse
|
40
|
Role of Chemokines in the Development and Progression of Alzheimer's Disease. J Mol Neurosci 2022; 72:1929-1951. [PMID: 35821178 PMCID: PMC9392685 DOI: 10.1007/s12031-022-02047-1] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2022] [Accepted: 07/04/2022] [Indexed: 11/24/2022]
Abstract
Alzheimer’s disease (AD) is a progressive neurogenerative disorder manifested by gradual memory loss and cognitive decline due to profound damage of cholinergic neurons. The neuropathological hallmarks of AD are intracellular deposits of neurofibrillary tangles (NFTs) and extracellular aggregates of amyloid β (Aβ). Mounting evidence indicates that intensified neuroinflammatory processes play a pivotal role in the pathogenesis of AD. Chemokines serve as signaling molecules in immune cells but also in nerve cells. Under normal conditions, neuroinflammation plays a neuroprotective role against various harmful factors. However, overexpression of chemokines initiates disruption of the integrity of the blood–brain barrier, facilitating immune cells infiltration into the brain. Then activated adjacent glial cells–astrocytes and microglia, release massive amounts of chemokines. Prolonged inflammation loses its protective role and drives an increase in Aβ production and aggregation, impairment of its clearance, or enhancement of tau hyperphosphorylation, contributing to neuronal loss and exacerbation of AD. Moreover, chemokines can be further released in response to growing deposits of toxic forms of Aβ. On the other hand, chemokines seem to exert multidimensional effects on brain functioning, including regulation of neurogenesis and synaptic plasticity in regions responsible for memory and cognitive abilities. Therefore, underexpression or complete genetic ablation of some chemokines can worsen the course of AD. This review covers the current state of knowledge on the role of particular chemokines and their receptors in the development and progression of AD. Special emphasis is given to their impact on forming Aβ and NFTs in humans and in transgenic murine models of AD.
Collapse
|
41
|
Bagaria J, Moon Y, Bagyinszky E, Shim KH, An SSA, Kim S, Han SH. Whole Exome Sequencing Reveals a Novel APOE Mutation in a Patient With Sporadic Early-Onset Alzheimer's Disease. Front Neurol 2022; 13:899644. [PMID: 35756922 PMCID: PMC9226417 DOI: 10.3389/fneur.2022.899644] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Accepted: 05/04/2022] [Indexed: 11/13/2022] Open
Abstract
Apolipoprotein (APOE) is implicated and verified as the main risk factor for early-onset Alzheimer's disease (AD). APOE is a protein that binds to lipids and is involved in cholesterol stability. Our paper reports a case of a sporadic early-onset AD (sEOAD) patient of a 54-year-old Korean man, where a novel APOE Leu159Pro heterozygous mutation was revealed upon Whole Exome Sequence analysis. The proband's CSF showed downregulated levels of Aβ42, with unchanged Tau levels. The mutation is in the Low-Density Lipoprotein Receptor (LDLR) region of the APOE gene, which mediates the clearance of APOE lipoproteins. LDLR works as a high-affinity point for APOE. Studies suggest that APOE-LDLR interplay could have varying effects. The LDLR receptor pathway has been previously suggested as a therapeutic target to treat tauopathy. However, the APOE-LDLR interaction has also shown a significant correlation with memory retention. Leu159Pro could be an interesting mutation that could be responsible for a less damaging pattern of AD by suppressing tau-association neurodegeneration while affecting the patient's memory retention and cognitive performance.
Collapse
Affiliation(s)
- Jaya Bagaria
- Department of Bionanotechnology, Gachon University, Seongnam-si, South Korea
| | - Yeonsil Moon
- Department of Neurology, Konkuk University School of Medicine and Konkuk University Medical Center, Seoul, South Korea
| | - Eva Bagyinszky
- Department of Industrial and Environmental Engineering, Graduate School of Environment, Gachon University, Seongnam, South Korea
| | - Kyu Hwan Shim
- Department of Bionanotechnology, Gachon University, Seongnam-si, South Korea
| | - Seong Soo A An
- Department of Bionanotechnology, Gachon University, Seongnam-si, South Korea
| | - SangYun Kim
- Department of Neurology, Seoul National University College of Medicine and Seoul National University Budang Hospital, Seongnam-si, South Korea
| | - Seol Heui Han
- Department of Neurology, Konkuk University School of Medicine and Konkuk University Medical Center, Seoul, South Korea
| |
Collapse
|
42
|
Sarto J, Mayà G, Molina-Porcel L, Balasa M, Gelpi E, Aldecoa I, Borrego-Écija S, Contador J, Ximelis T, Vergara M, Antonell A, Sánchez-Valle R, Albert L. Evolution of Clinical-Pathological Correlations in Early-Onset Alzheimer's Disease Over a 25-Year Period in an Academic Brain Bank. J Alzheimers Dis 2022; 87:1659-1669. [PMID: 35723108 DOI: 10.3233/jad-220045] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
BACKGROUND Early onset Alzheimer's disease (EOAD) represents a diagnostic challenge and is associated with a high diagnostic delay and misdiagnosis. OBJECTIVE To describe clinical and pathological data from a pathologically confirmed EOAD cohort and evaluate evolving trends in clinical-pathological correlation accuracy. METHODS Retrospective review of clinical and neuropathological data of pathologically confirmed EOAD patients (age at onset [AAO] < 60). Comparison between two periods: 1994- 2009 and 2010- 2018. RESULTS Eighty brain donors were included. Mean AAO, age at death, and diagnostic delay was 55, 66, and 3 years, respectively. Twenty-nine percent had a nonamnestic presentation. Sixteen percent were given a non-AD initial clinical diagnosis (initial misdiagnosis) and 14% received a final misdiagnosis. Nonamnestic presentation patients received more misdiagnoses than amnestic presentation ones (39% versus 7% and 39% versus 3.5%, on initial and final misdiagnosis, respectively). When comparing both time periods, a trend towards a higher diagnostic accuracy in the 2010- 2018 period was observed, mainly on initial misdiagnosis in nonamnestic presentation patients (53% versus 13%, p = 0.069). Diagnostic delay was similar between both periods. Cerebral amyloid angiopathy (96%) and Lewy body co-pathology (55%) were very frequent, while limbic-predominant age-related TDP-43 encephalopathy pathologic changes were only present in 12.5%. CONCLUSION In the last decade, there has been a trend towards improved diagnostic accuracy in EOAD, which might be explained by improved diagnostic criteria, increasing experience on EOAD and the beginning of the use of biomarkers, although diagnostic delay remains similar. Concomitant neuropathology was very frequent despite the relatively young age of brain donors.
Collapse
Affiliation(s)
- Jordi Sarto
- Alzheimer's Disease and Other Cognitive Disorders Unit, Neurology Service, Hospital Clínic de Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), University of Barcelona, Barcelona, Spain.,Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - Gerard Mayà
- Alzheimer's Disease and Other Cognitive Disorders Unit, Neurology Service, Hospital Clínic de Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), University of Barcelona, Barcelona, Spain
| | - Laura Molina-Porcel
- Alzheimer's Disease and Other Cognitive Disorders Unit, Neurology Service, Hospital Clínic de Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), University of Barcelona, Barcelona, Spain.,Neurological Tissue Bank, Biobanc-Hospital Clínic Barcelona-IDIBAPS, Barcelona, Spain
| | - Mircea Balasa
- Alzheimer's Disease and Other Cognitive Disorders Unit, Neurology Service, Hospital Clínic de Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), University of Barcelona, Barcelona, Spain
| | - Ellen Gelpi
- Neurological Tissue Bank, Biobanc-Hospital Clínic Barcelona-IDIBAPS, Barcelona, Spain.,Division of Neuropathology and Neurochemistry, Department of Neurology, Medical University of Vienna, Vienna, Austria
| | - Iban Aldecoa
- Neurological Tissue Bank, Biobanc-Hospital Clínic Barcelona-IDIBAPS, Barcelona, Spain.,Pathology Service, Biomedical Diagnostic Center, Hospital Clínic de Barcelona, University of Barcelona, Barcelona, Spain
| | - Sergi Borrego-Écija
- Alzheimer's Disease and Other Cognitive Disorders Unit, Neurology Service, Hospital Clínic de Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), University of Barcelona, Barcelona, Spain.,Neurological Tissue Bank, Biobanc-Hospital Clínic Barcelona-IDIBAPS, Barcelona, Spain
| | - Jose Contador
- Alzheimer's Disease and Other Cognitive Disorders Unit, Neurology Service, Hospital Clínic de Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), University of Barcelona, Barcelona, Spain
| | - Teresa Ximelis
- Neurological Tissue Bank, Biobanc-Hospital Clínic Barcelona-IDIBAPS, Barcelona, Spain
| | - Miguel Vergara
- Alzheimer's Disease and Other Cognitive Disorders Unit, Neurology Service, Hospital Clínic de Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), University of Barcelona, Barcelona, Spain
| | - Anna Antonell
- Alzheimer's Disease and Other Cognitive Disorders Unit, Neurology Service, Hospital Clínic de Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), University of Barcelona, Barcelona, Spain
| | - Raquel Sánchez-Valle
- Alzheimer's Disease and Other Cognitive Disorders Unit, Neurology Service, Hospital Clínic de Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), University of Barcelona, Barcelona, Spain.,Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain.,Neurological Tissue Bank, Biobanc-Hospital Clínic Barcelona-IDIBAPS, Barcelona, Spain
| | - Lladó Albert
- Alzheimer's Disease and Other Cognitive Disorders Unit, Neurology Service, Hospital Clínic de Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), University of Barcelona, Barcelona, Spain.,Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | | |
Collapse
|
43
|
Sirkis DW, Bonham LW, Johnson TP, La Joie R, Yokoyama JS. Dissecting the clinical heterogeneity of early-onset Alzheimer's disease. Mol Psychiatry 2022; 27:2674-2688. [PMID: 35393555 PMCID: PMC9156414 DOI: 10.1038/s41380-022-01531-9] [Citation(s) in RCA: 61] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/11/2021] [Revised: 03/07/2022] [Accepted: 03/16/2022] [Indexed: 12/14/2022]
Abstract
Early-onset Alzheimer's disease (EOAD) is a rare but particularly devastating form of AD. Though notable for its high degree of clinical heterogeneity, EOAD is defined by the same neuropathological hallmarks underlying the more common, late-onset form of AD. In this review, we describe the various clinical syndromes associated with EOAD, including the typical amnestic phenotype as well as atypical variants affecting visuospatial, language, executive, behavioral, and motor functions. We go on to highlight advances in fluid biomarker research and describe how molecular, structural, and functional neuroimaging can be used not only to improve EOAD diagnostic acumen but also enhance our understanding of fundamental pathobiological changes occurring years (and even decades) before the onset of symptoms. In addition, we discuss genetic variation underlying EOAD, including pathogenic variants responsible for the well-known mendelian forms of EOAD as well as variants that may increase risk for the much more common forms of EOAD that are either considered to be sporadic or lack a clear autosomal-dominant inheritance pattern. Intriguingly, specific pathogenic variants in PRNP and MAPT-genes which are more commonly associated with other neurodegenerative diseases-may provide unexpectedly important insights into the formation of AD tau pathology. Genetic analysis of the atypical clinical syndromes associated with EOAD will continue to be challenging given their rarity, but integration of fluid biomarker data, multimodal imaging, and various 'omics techniques and their application to the study of large, multicenter cohorts will enable future discoveries of fundamental mechanisms underlying the development of EOAD and its varied clinical presentations.
Collapse
Affiliation(s)
- Daniel W Sirkis
- Memory and Aging Center, Department of Neurology, Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, 94158, USA
| | - Luke W Bonham
- Memory and Aging Center, Department of Neurology, Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, 94158, USA
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, San Francisco, CA, 94158, USA
| | - Taylor P Johnson
- Memory and Aging Center, Department of Neurology, Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, 94158, USA
| | - Renaud La Joie
- Memory and Aging Center, Department of Neurology, Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, 94158, USA
| | - Jennifer S Yokoyama
- Memory and Aging Center, Department of Neurology, Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, 94158, USA.
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, San Francisco, CA, 94158, USA.
| |
Collapse
|
44
|
Qiao Z, Wang G, Zhao X, Wang K, Fan D, Chen Q, Ai L. Neuropsychological Performance Is Correlated With Tau Protein Deposition and Glucose Metabolism in Patients With Alzheimer’s Disease. Front Aging Neurosci 2022; 14:841942. [PMID: 35663582 PMCID: PMC9158435 DOI: 10.3389/fnagi.2022.841942] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Accepted: 03/07/2022] [Indexed: 11/13/2022] Open
Abstract
Objective This study characterizes glucose metabolism and tau protein deposition distribution in patients with Alzheimer’s disease (AD) and to evaluate the relationships between neuropsychological performance and tau protein deposition or glucose metabolism using 18F-FDG and 18F-AV1451 positron emission tomography/computed tomography (PET/CT). Methods Sixty-four patients with β-amyloid-positive (Aβ+) AD and twenty-five healthy participants were enrolled in this study. All participants underwent 18F-FDG and 18F-AV1451 PET/CT. Clinical data and neuropsychological scores were collected. Patients with AD were divided into mild, moderate, and severe groups according to mini-mental state examination (MMSE) scores. The standardized uptake value ratios (SUVRs) for both FDG and AV1451 PET images were calculated using the cerebellar vermis as reference. The SUVRs of the whole cerebral cortex and each brain region were calculated. The volume of interest (VOI) was obtained using automated anatomical atlas (AAL) and Brodmann regions. Student’s t-test was used to perform intergroup comparisons of SUVR. The partial correlation coefficient between SUVR and neuropsychological scores was computed in an inter-subject manner using age and education as covariates. Results The mild subgroup showed a reduction in glucose metabolism and aggregation of tau protein in the temporoparietal cortex. With a decline in neuropsychiatric performance, the SUVR on FDG PET decreased and SUVR on tau PET increased gradually. The areas of glucose metabolism reduction and tau protein deposition appeared first in the parietal cortex, followed by the temporal and frontal cortex, successively. Both FDG and tau SUVRs significantly correlated with MMSE, Montreal cognitive assessment (MOCA), auditory verbal learning test (AVLT), Boston naming test (BNT), clock drawing task (CDT), and verbal fluency test (VFT) (p < 0.05). The SUVR on FDG PET significantly correlated with activities of daily living (ADL) and the Hamilton depression scale (HAMD). There was no significant correlation between the tau SUVRs and ADL or HAMD. Conclusion The extension of tau protein deposition was similar but not exactly consistent with the area of glucose metabolism reduction. Both tau and FDG SUVRs correlated with cognitive function in domain-specific patterns, and the results of FDG PET more closely correlated with neuropsychological function than tau PET results did.
Collapse
Affiliation(s)
- Zhen Qiao
- Department of Nuclear Medicine, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Guihong Wang
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- Guihong Wang,
| | - Xiaobin Zhao
- Department of Nuclear Medicine, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Kai Wang
- Department of Nuclear Medicine, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Di Fan
- Department of Nuclear Medicine, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Qian Chen
- Department of Nuclear Medicine, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Lin Ai
- Department of Nuclear Medicine, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- *Correspondence: Lin Ai,
| |
Collapse
|
45
|
Nabi M, Tabassum N. Role of Environmental Toxicants on Neurodegenerative Disorders. FRONTIERS IN TOXICOLOGY 2022; 4:837579. [PMID: 35647576 PMCID: PMC9131020 DOI: 10.3389/ftox.2022.837579] [Citation(s) in RCA: 64] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Accepted: 04/22/2022] [Indexed: 12/22/2022] Open
Abstract
Neurodegeneration leads to the loss of structural and functioning components of neurons over time. Various studies have related neurodegeneration to a number of degenerative disorders. Neurological repercussions of neurodegeneration can have severe impacts on the physical and mental health of patients. In the recent past, various neurodegenerative ailments such as Alzheimer’s and Parkinson’s illnesses have received global consideration owing to their global occurrence. Environmental attributes have been regarded as the main contributors to neural dysfunction-related disorders. The majority of neurological diseases are mainly related to prenatal and postnatal exposure to industrially produced environmental toxins. Some neurotoxic metals, like lead (Pb), aluminium (Al), Mercury (Hg), manganese (Mn), cadmium (Cd), and arsenic (As), and also pesticides and metal-based nanoparticles, have been implicated in Parkinson’s and Alzheimer’s disease. The contaminants are known for their ability to produce senile or amyloid plaques and neurofibrillary tangles (NFTs), which are the key features of these neurological dysfunctions. Besides, solvent exposure is also a significant contributor to neurological diseases. This study recapitulates the role of environmental neurotoxins on neurodegeneration with special emphasis on major neurodegenerative disorders such as Alzheimer’s and Parkinson’s disease.
Collapse
Affiliation(s)
- Masarat Nabi
- Department of Environmental Science, University of Kashmir, Srinagar, India
- *Correspondence: Masarat Nabi, , orcid.org/0000-0003-1677-6498; Nahida Tabassum,
| | - Nahida Tabassum
- Department of Pharmaceutical Sciences, University of Kashmir, Srinagar, India
- *Correspondence: Masarat Nabi, , orcid.org/0000-0003-1677-6498; Nahida Tabassum,
| |
Collapse
|
46
|
Uchida Y, Kan H, Sakurai K, Horimoto Y, Hayashi E, Iida A, Okamura N, Oishi K, Matsukawa N. APOE ɛ4 dose associates with increased brain iron and β-amyloid via blood-brain barrier dysfunction. J Neurol Neurosurg Psychiatry 2022; 93:jnnp-2021-328519. [PMID: 35483916 DOI: 10.1136/jnnp-2021-328519] [Citation(s) in RCA: 41] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Accepted: 03/23/2022] [Indexed: 01/09/2023]
Abstract
OBJECTIVE To examine the effect of apolipoprotein E (APOE) ɛ4 dose on blood-brain barrier (BBB) clearance function, evaluated using an advanced MRI technique and analyse its correlation with brain iron and β-amyloid accumulation in the early stages of the Alzheimer's continuum. METHODS In this single-centre observational prospective cohort study, 24 APOE ɛ4 non-carriers, 22 heterozygotes and 20 homozygotes in the early stages of the Alzheimer's continuum were scanned with diffusion-prepared arterial spin labelling, which estimates the water exchange rate across the BBB (kw). Participants also underwent quantitative susceptibility mapping, [11C]Pittsburgh compound B-positron emission tomography and neuropsychological testing. Using an atlas-based approach, we compared the regional kw of the whole brain among the groups and analysed its correlation with the neuroradiological and neuropsychological findings. RESULTS The BBB kw values in the neocortices differed significantly among the groups (APOE ɛ4 non-carriers>heterozygotes>homozygotes). These values correlated with brain iron levels (frontal lobe: r=-0.476, 95% CI=-0.644 to -0.264, p=0.011; medial temporal lobe: r=-0.455, 95% CI=-0.628 to -0.239, p=0.017), β-amyloid loads (frontal lobe: r=-0.504, 95% CI=-0.731 to -0.176, p=0.015; medial temporal lobe: r=-0.452, 95% CI=-0.699 to -0.110, p=0.036) and neuropsychological scores, after adjusting for age, sex and APOE ɛ4 dose. INTERPRETATION Our results suggest that an increased APOE ɛ4 dose is associated with decreased effective brain-waste clearance, such as iron and β-amyloid, through the BBB.
Collapse
Affiliation(s)
- Yuto Uchida
- Department of Neurology, Nagoya City University, Nagoya, Japan
- Department of Neurology, Toyokawa City Hospital, Toyokawa, Japan
| | - Hirohito Kan
- Department of Integrated Health Sciences, Nagoya University, Nagoya, Japan
| | - Keita Sakurai
- Department of Radiology, National Center for Geriatrics and Gerontology, Obu, Japan
| | - Yoshihiko Horimoto
- Department of Neurology, Nagoya City Rehabilitation Center Group, Nagoya, Japan
| | - Emi Hayashi
- Department of Radiology, Nagoya City Rehabilitation Center Group, Nagoya, Japan
| | - Akihiko Iida
- Department of Radiology, Nagoya City Rehabilitation Center Group, Nagoya, Japan
| | - Nobuyuki Okamura
- Division of Pharmacology, Faculty of Medicine, Tohoku Medical and Pharmaceutical University, Sendai, Japan
| | - Kenichi Oishi
- Department of Radiology and Radiological Science, Johns Hopkins University, Baltimore, Maryland, USA
| | | |
Collapse
|
47
|
Smirnov DS, Salmon DP, Galasko D, Goodwill VS, Hansen LA, Zhao Y, Edland SD, Léger GC, Peavy GM, Jacobs DM, Rissman R, Pizzo DP, Hiniker A. Association of Neurofibrillary Tangle Distribution With Age at Onset-Related Clinical Heterogeneity in Alzheimer Disease: An Autopsy Study. Neurology 2022; 98:e506-e517. [PMID: 34810247 PMCID: PMC8826459 DOI: 10.1212/wnl.0000000000013107] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2021] [Accepted: 11/04/2021] [Indexed: 02/03/2023] Open
Abstract
BACKGROUND AND OBJECTIVE Patients with earlier age at onset of sporadic Alzheimer disease (AD) are more likely than those with later onset to present with atypical clinical and cognitive features. We sought to determine whether this age-related clinical and cognitive heterogeneity is mediated by different topographic distributions of tau-aggregate neurofibrillary tangles (NFTs) or by variable amounts of concomitant non-AD neuropathology. METHODS The relative distribution of NFT density in hippocampus and midfrontal neocortex was calculated, and α-synuclein, TAR DNA binding protein 43 (TDP-43), and microvascular copathologies were staged, in patients with severe AD and age at onset of 51-60 (n = 40), 61-70 (n = 41), and >70 (n = 40) years. Regression, mediation, and mixed effects models examined relationships of pathologic findings with clinical features and longitudinal cognitive decline. RESULTS Patients with later age at onset of AD were less likely to present with nonmemory complaints (odds ratio [OR] 0.46 per decade, 95% confidence interval [CI] 0.22-0.88), psychiatric symptoms (β = -0.66, 95% CI -1.15 to -0.17), and functional impairment (β = -1.25, 95% CI -2.34 to -0.16). TDP-43 (OR 2.00, 95% CI 1.23-3.35) and microvascular copathology (OR 2.02, 95% CI 1.24-3.40) were more common in later onset AD, and α-synuclein copathology was not related to age at onset. NFT density in midfrontal cortex (β = -0.51, 95% CI -0.72 to -0.31) and midfrontal/hippocampal NFT ratio (β = -0.18, 95% CI -0.26 to -0.10) were lower in those with later age at onset. Executive function (β = 0.48, 95% CI 0.09-0.90) and visuospatial cognitive deficits (β = 0.97, 95% CI 0.46-1.46) were less impaired in patients with later age at onset. Mediation analyses showed that the effect of age at onset on severity of executive function deficits was mediated by midfrontal/hippocampal NFT ratio (β = 0.21, 95% CI 0.08-0.38) and not by concomitant non-AD pathologies. Midfrontal/hippocampal NFT ratio also mediated an association between earlier age at onset and faster decline on tests of global cognition, executive function, and visuospatial abilities. DISCUSSION Worse executive dysfunction and faster cognitive decline in people with sporadic AD with earlier rather than later age at onset is mediated by greater relative midfrontal neocortical to hippocampal NFT burden and not by concomitant non-AD neuropathology.
Collapse
Affiliation(s)
- Denis S Smirnov
- From the Departments of Neurosciences (D.S.S., D.P.S., D.G., G.C.L., G.M.P., D.M.J., R.R., A.H.), Pathology (V.S.G., L.A.H., D.P.P., A.H.), and Family Medicine and Public Health (Y.Z., S.D.E.), University of California, San Diego; and VA San Diego Healthcare System (D.G., R.R., A.H.), CA
| | - David P Salmon
- From the Departments of Neurosciences (D.S.S., D.P.S., D.G., G.C.L., G.M.P., D.M.J., R.R., A.H.), Pathology (V.S.G., L.A.H., D.P.P., A.H.), and Family Medicine and Public Health (Y.Z., S.D.E.), University of California, San Diego; and VA San Diego Healthcare System (D.G., R.R., A.H.), CA
| | - Douglas Galasko
- From the Departments of Neurosciences (D.S.S., D.P.S., D.G., G.C.L., G.M.P., D.M.J., R.R., A.H.), Pathology (V.S.G., L.A.H., D.P.P., A.H.), and Family Medicine and Public Health (Y.Z., S.D.E.), University of California, San Diego; and VA San Diego Healthcare System (D.G., R.R., A.H.), CA
| | - Vanessa S Goodwill
- From the Departments of Neurosciences (D.S.S., D.P.S., D.G., G.C.L., G.M.P., D.M.J., R.R., A.H.), Pathology (V.S.G., L.A.H., D.P.P., A.H.), and Family Medicine and Public Health (Y.Z., S.D.E.), University of California, San Diego; and VA San Diego Healthcare System (D.G., R.R., A.H.), CA
| | - Lawrence A Hansen
- From the Departments of Neurosciences (D.S.S., D.P.S., D.G., G.C.L., G.M.P., D.M.J., R.R., A.H.), Pathology (V.S.G., L.A.H., D.P.P., A.H.), and Family Medicine and Public Health (Y.Z., S.D.E.), University of California, San Diego; and VA San Diego Healthcare System (D.G., R.R., A.H.), CA
| | - Yu Zhao
- From the Departments of Neurosciences (D.S.S., D.P.S., D.G., G.C.L., G.M.P., D.M.J., R.R., A.H.), Pathology (V.S.G., L.A.H., D.P.P., A.H.), and Family Medicine and Public Health (Y.Z., S.D.E.), University of California, San Diego; and VA San Diego Healthcare System (D.G., R.R., A.H.), CA
| | - Steven D Edland
- From the Departments of Neurosciences (D.S.S., D.P.S., D.G., G.C.L., G.M.P., D.M.J., R.R., A.H.), Pathology (V.S.G., L.A.H., D.P.P., A.H.), and Family Medicine and Public Health (Y.Z., S.D.E.), University of California, San Diego; and VA San Diego Healthcare System (D.G., R.R., A.H.), CA
| | - Gabriel C Léger
- From the Departments of Neurosciences (D.S.S., D.P.S., D.G., G.C.L., G.M.P., D.M.J., R.R., A.H.), Pathology (V.S.G., L.A.H., D.P.P., A.H.), and Family Medicine and Public Health (Y.Z., S.D.E.), University of California, San Diego; and VA San Diego Healthcare System (D.G., R.R., A.H.), CA
| | - Guerry M Peavy
- From the Departments of Neurosciences (D.S.S., D.P.S., D.G., G.C.L., G.M.P., D.M.J., R.R., A.H.), Pathology (V.S.G., L.A.H., D.P.P., A.H.), and Family Medicine and Public Health (Y.Z., S.D.E.), University of California, San Diego; and VA San Diego Healthcare System (D.G., R.R., A.H.), CA
| | - Diane M Jacobs
- From the Departments of Neurosciences (D.S.S., D.P.S., D.G., G.C.L., G.M.P., D.M.J., R.R., A.H.), Pathology (V.S.G., L.A.H., D.P.P., A.H.), and Family Medicine and Public Health (Y.Z., S.D.E.), University of California, San Diego; and VA San Diego Healthcare System (D.G., R.R., A.H.), CA
| | - Robert Rissman
- From the Departments of Neurosciences (D.S.S., D.P.S., D.G., G.C.L., G.M.P., D.M.J., R.R., A.H.), Pathology (V.S.G., L.A.H., D.P.P., A.H.), and Family Medicine and Public Health (Y.Z., S.D.E.), University of California, San Diego; and VA San Diego Healthcare System (D.G., R.R., A.H.), CA
| | - Donald P Pizzo
- From the Departments of Neurosciences (D.S.S., D.P.S., D.G., G.C.L., G.M.P., D.M.J., R.R., A.H.), Pathology (V.S.G., L.A.H., D.P.P., A.H.), and Family Medicine and Public Health (Y.Z., S.D.E.), University of California, San Diego; and VA San Diego Healthcare System (D.G., R.R., A.H.), CA
| | - Annie Hiniker
- From the Departments of Neurosciences (D.S.S., D.P.S., D.G., G.C.L., G.M.P., D.M.J., R.R., A.H.), Pathology (V.S.G., L.A.H., D.P.P., A.H.), and Family Medicine and Public Health (Y.Z., S.D.E.), University of California, San Diego; and VA San Diego Healthcare System (D.G., R.R., A.H.), CA.
| |
Collapse
|
48
|
Differential associations between neocortical tau pathology and blood flow with cognitive deficits in early-onset vs late-onset Alzheimer's disease. Eur J Nucl Med Mol Imaging 2022; 49:1951-1963. [PMID: 34997294 PMCID: PMC9016024 DOI: 10.1007/s00259-021-05669-6] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Accepted: 12/20/2021] [Indexed: 12/23/2022]
Abstract
Purpose Early-onset Alzheimer’s disease (EOAD) and late-onset Alzheimer’s disease (LOAD) differ in neuropathological burden and type of cognitive deficits. Assessing tau pathology and relative cerebral blood flow (rCBF) measured with [18F]flortaucipir PET in relation to cognition may help explain these differences between EOAD and LOAD. Methods Seventy-nine amyloid-positive individuals with a clinical diagnosis of AD (EOAD: n = 35, age-at-PET = 59 ± 5, MMSE = 23 ± 4; LOAD: n = 44, age-at-PET = 71 ± 5, MMSE = 23 ± 4) underwent a 130-min dynamic [18F]flortaucipir PET scan and extensive neuropsychological assessment. We extracted binding potentials (BPND) and R1 (proxy of rCBF) from parametric images using receptor parametric mapping, in medial and lateral temporal, parietal, occipital, and frontal regions-of-interest and used nine neuropsychological tests covering memory, attention, language, and executive functioning. We first examined differences between EOAD and LOAD in BPND or R1 using ANOVA (region-of-interest analysis) and voxel-wise contrasts. Next, we performed linear regression models to test for potential interaction effects between age-at-onset and BPND/R1 on cognition. Results Both region-of-interest and voxel-wise contrasts showed higher [18F]flortaucipir BPND values across all neocortical regions in EOAD. By contrast, LOAD patients had lower R1 values (indicative of more reduced rCBF) in medial temporal regions. For both tau and flow in lateral temporal, and occipitoparietal regions, associations with cognitive impairment were stronger in EOAD than in LOAD (EOAD BPND − 0.76 ≤ stβ ≤ − 0.48 vs LOAD − 0.18 ≤ stβ ≤ − 0.02; EOAD R1 0.37 ≤ stβ ≤ 0.84 vs LOAD − 0.25 ≤ stβ ≤ 0.16). Conclusions Compared to LOAD, the degree of lateral temporal and occipitoparietal tau pathology and relative cerebral blood-flow is more strongly associated with cognition in EOAD. Supplementary Information The online version contains supplementary material available at 10.1007/s00259-021-05669-6.
Collapse
|
49
|
Duara R, Barker W. Heterogeneity in Alzheimer's Disease Diagnosis and Progression Rates: Implications for Therapeutic Trials. Neurotherapeutics 2022; 19:8-25. [PMID: 35084721 PMCID: PMC9130395 DOI: 10.1007/s13311-022-01185-z] [Citation(s) in RCA: 48] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/05/2022] [Indexed: 01/03/2023] Open
Abstract
The clinical presentation and the pathological processes underlying Alzheimer's disease (AD) can be very heterogeneous in severity, location, and composition including the amount and distribution of AB deposition and spread of neurofibrillary tangles in different brain regions resulting in atypical clinical patterns and the existence of distinct AD variants. Heterogeneity in AD may be related to demographic factors (such as age, sex, educational and socioeconomic level) and genetic factors, which influence underlying pathology, the cognitive and behavioral phenotype, rate of progression, the occurrence of neuropsychiatric features, and the presence of comorbidities (e.g., vascular disease, neuroinflammation). Heterogeneity is also manifest in the individual resilience to the development of neuropathology (brain reserve) and the ability to compensate for its cognitive and functional impact (cognitive and functional reserve). The variability in specific cognitive profiles and types of functional impairment may be associated with different progression rates, and standard measures assessing progression may not be equivalent for individual cognitive and functional profiles. Other factors, which may govern the presence, rate, and type of progression of AD, include the individuals' general medical health, the presence of specific systemic conditions, and lifestyle factors, including physical exercise, cognitive and social stimulation, amount of leisure activities, environmental stressors, such as toxins and pollution, and the effects of medications used to treat medical and behavioral conditions. These factors that affect progression are important to consider while designing a clinical trial to ensure, as far as possible, well-balanced treatment and control groups.
Collapse
Affiliation(s)
- Ranjan Duara
- Wien Center for Alzheimer's Disease and Memory Disorders, Mount Sinai Medical Center, Miami Beach, FL, USA
- Departments of Neurology, University of Florida College of Medicine, Gainesville, FL, USA
- Herbert Wertheim College of Medicine, Florida International University, Miami, FL, USA
| | - Warren Barker
- Wien Center for Alzheimer's Disease and Memory Disorders, Mount Sinai Medical Center, Miami Beach, FL, USA.
| |
Collapse
|
50
|
Moser VA, Workman MJ, Hurwitz SJ, Lipman RM, Pike CJ, Svendsen CN. Microglial transcription profiles in mouse and human are driven by APOE4 and sex. iScience 2021; 24:103238. [PMID: 34746703 PMCID: PMC8551075 DOI: 10.1016/j.isci.2021.103238] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2021] [Revised: 06/28/2021] [Accepted: 10/04/2021] [Indexed: 11/22/2022] Open
Abstract
Apolipoprotein E4 (APOE4) is the strongest genetic risk factor for sporadic Alzheimer's disease (AD). APOE4 is known to affect the function of microglia, but to what extent this gene drives microglial gene expression has thus far not been examined. Using a transgenic mouse model of AD that expresses human APOE, we identify a unique transcriptional profile associated with APOE4 expression. We also show a sex and APOE interaction, such that both female sex and APOE4 drive expression of this gene profile. We confirm these findings in human cells, using microglia derived from induced pluripotent stem cells (iMGL). Moreover, we find that these interactions are driven in part by genes related to metal processing, and we show that zinc treatment has APOE genotype-dependent effects on iMGL. These data identify a sex- and APOE4-associated microglial transcription profile and highlight the importance of considering interactive risk factors such as sex and environmental exposures.
Collapse
Affiliation(s)
- V. Alexandra Moser
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Michael J. Workman
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Samantha J. Hurwitz
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Rachel M. Lipman
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Christian J. Pike
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA, USA
| | - Clive N. Svendsen
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| |
Collapse
|