1
|
Rauchmann BS, Ersözlü E, Luedecke D, Franzmeier N, Perneczky R. Multimodal and longitudinal characterization of distinct tau and atrophy clusters in Alzheimer's disease spectrum. Sci Rep 2025; 15:18142. [PMID: 40415101 DOI: 10.1038/s41598-025-98338-9] [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: 06/02/2024] [Accepted: 04/10/2025] [Indexed: 05/27/2025] Open
Abstract
Neuropathological and neuroimaging studies have identified several (endo-)phenotypes of Alzheimer's disease (AD), suggesting a substantial heterogeneity in cerebral atrophy and tau spreading patterns. We included in our study a total of 320 participants, including healthy controls (N = 154) and patients across the AD spectrum (N = 166). We identified clusters of cerebral atrophy and tau PET uptake using a data-driven and similarity-based clustering approach, aiming to examine regional abnormality patterns in both modalities and differences in the clinical, cognitive, and biomarker characteristics among derived clusters. Abnormality patterns in tau PET and T1-weighted MRI within the same individuals revealed four distinct clusters for each imaging modality as surrogate markers of tau and neurodegeneration, respectively. The tau PET and atrophy clusters mainly showed substantial differences in their clustering allocations. While having the most severe biomarkers burden, the left temporal tau and diffuse atrophy clusters revealed the fastest clinical progression and steepest increase in tau PET uptake. Moreover, the diffuse atrophy cluster showed the fastest cortical volume loss, followed by the limbic-predominant atrophy cluster. Our results suggest characteristic differences between tau PET and atrophy clusters, especially for tau PET clusters, revealing more pronounced differences in cognitive profiles and disease biomarker trajectories than atrophy clusters.
Collapse
Affiliation(s)
- Boris-Stephan Rauchmann
- Department of Neuroradiology, LMU Hospital, LMU Munich, Munich, Germany.
- Division of Mental Health of Older Adults, Department of Psychiatry and Psychotherapy, Ludwig-Maximilians-Universität München, Nußbaumstr. 7, 80336, Munich, Germany.
- German Center for Neurodegenerative Diseases (DZNE) Munich, Munich, Germany.
- Sheffield Institute for Translational Neuroscience (SITraN), University of Sheffield, Sheffield, UK.
| | - Ersin Ersözlü
- Department of Psychiatry and Neurosciences, Charité Universitätsmedizin Berlin, Berlin, Germany
- German Center for Neurodegenerative Diseases (DZNE), Berlin, Germany
| | - Dorothea Luedecke
- Division of Mental Health of Older Adults, Department of Psychiatry and Psychotherapy, Ludwig-Maximilians-Universität München, Nußbaumstr. 7, 80336, Munich, Germany
| | - Nicolai Franzmeier
- Institute for Stroke and Dementia Research (ISD), LMU Hospital, LMU Munich, Munich, Germany
| | - Robert Perneczky
- Division of Mental Health of Older Adults, Department of Psychiatry and Psychotherapy, Ludwig-Maximilians-Universität München, Nußbaumstr. 7, 80336, Munich, Germany
- German Center for Neurodegenerative Diseases (DZNE) Munich, Munich, Germany
- Sheffield Institute for Translational Neuroscience (SITraN), University of Sheffield, Sheffield, UK
- Ageing Epidemiology (AGE) Research Unit, School of Public Health, Imperial College London, London, UK
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| |
Collapse
|
2
|
Gothwal A, Muolokwu CE, Chaulagain B, Mahanta AK, Singh J. Intranasal delivery of pApoE2 via penetratin-mannose multi-functionalized chitosan polymeric micelles to the brain: Reduced total tau and phosphorylated tau burden in transgenic Alzheimer's mouse model. Int J Biol Macromol 2025; 310:143542. [PMID: 40294672 DOI: 10.1016/j.ijbiomac.2025.143542] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2025] [Revised: 04/16/2025] [Accepted: 04/25/2025] [Indexed: 04/30/2025]
Abstract
The phosphorylated tau accumulation is a classical pathological hallmark of future cognitive decline and a cause of neuronal death in Alzheimer's disease (AD). In this study, we developed multi-functionalized chitosan (CS) based polymeric micelles to effectively deliver pApoE2 via intranasal administration to the brain. The CS was modified with caproic acid (CA), cell-penetrating peptide penetratin (PEN), and GLUT-1 transporter ligand mannose (MAN) for selective and enhanced delivery to the brain. The multi-functionalized Cap-g-CS-PEN-MAN polymeric micelles were ≤200 nm in size, cationic in charge, and uniformly distributed (PDI ≤ 0.3). The multi-functionalized polymeric micelles did not exhibit toxicity against bEnd.3 cells and erythrocytes up to polymer concentrations of 500 μg/mL. The Cap-g-CS-PEN-MAN /pDNA polyplex was stable against a DNase rich environment. The Cap-g-CS-PEN-MAN/pAPoE2 polyplex demonstrated elevated expression of ApoE in primary astrocytes and neurons, 9.47 ± 2.13 and 5.67 ± 2.69 ng/mg of protein, respectively. The therapeutic efficacy of the Cap-g-CS-PEN-MAN/pApoE2 polyplex was analyzed against the PS19 tauopathy mice model. Total tau burden was significantly (p ≤ 0.05) reduced by 4.09 ± 1.4 ng/mg of protein in Cap-g-CS-PEN-MAN/pApoE2 polyplex administered mice over the other groups. Phosphorylated tau pT181 level was also significantly (p ≤ 0.05) reduced in Cap-g-CS-PEN-MAN/pApoE2 polyplex administered mice over saline, pApoE2 and Cap-g-CS/pApoE2 treated groups.
Collapse
Affiliation(s)
- Avinash Gothwal
- Department of Pharmaceutical Sciences, School of Pharmacy, College of Health and Human Sciences, North Dakota State University, Fargo, ND 58108-6050, USA
| | - Chinenye Edith Muolokwu
- Department of Pharmaceutical Sciences, School of Pharmacy, College of Health and Human Sciences, North Dakota State University, Fargo, ND 58108-6050, USA
| | - Bivek Chaulagain
- Department of Pharmaceutical Sciences, School of Pharmacy, College of Health and Human Sciences, North Dakota State University, Fargo, ND 58108-6050, USA
| | - Arun Kumar Mahanta
- Department of Pharmaceutical Sciences, School of Pharmacy, College of Health and Human Sciences, North Dakota State University, Fargo, ND 58108-6050, USA
| | - Jagdish Singh
- Department of Pharmaceutical Sciences, School of Pharmacy, College of Health and Human Sciences, North Dakota State University, Fargo, ND 58108-6050, USA.
| |
Collapse
|
3
|
Chang H, Huang C, Huang S, Hsu S, Lin K, Ho T, Wu H, Chang C. Distinct biological property of tau in tau-first cognitive proteinopathy: Evidence by longitudinal clinical neuroimaging profiles and compared with late-onset Alzheimer disease. Psychiatry Clin Neurosci 2024; 78:446-455. [PMID: 38864501 PMCID: PMC11488611 DOI: 10.1111/pcn.13680] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Revised: 03/22/2024] [Accepted: 05/02/2024] [Indexed: 06/13/2024]
Abstract
BACKGROUND Tau-first cognitive proteinopathy (TCP) denotes a clinical phenotype of Alzheimer disease (AD) showing Florzolotau(18F) positron emission tomography (PET) positivity but a negative amyloid status. AIM We explored the biological property of tau using longitudinal cognitive and neuroimaging data in TCP and compared with late-onset AD (LOAD). METHOD We enrolled 56 patients with LOAD, 34 patients with TCP, and 26 cognitive unimpaired controls. All of the participants had historical data of 2 to 4 three-dimensional T1 images and 2 to 6 annual cognitive evaluations over a follow-up period of 7 years. Tau topography was measured using Florzolotau(18F) PET. In the LOAD and TCP groups, we constructed tau or gray matter clusters covarying with the cognitive measurements. We used mediator analysis to explore the regional tau load as predictor, gray matter partitions as mediators, and significant cognitive test scores as outcomes. Longitudinal cognitive decline and cortical thickness degeneration pattern were analyzed using a linear mixed-effects model. RESULTS The TCP group had longitudinal declines in nonexecutive domains. The deterministic factor predicting the short-term memory score in TCP was the hippocampal volume and not directly via the medial and lateral temporal tau load. These features formed the conceptual differences with LOAD. DISCUSSION The biological properties of tau and the longitudinal cognitive-imaging trajectory support the conceptual distinction between TCP and LOAD. TCP represents one specific entity featuring salient short-term memory impairment, declines in nonexecutive domains, a slower gray matter degenerative pattern, and a restricted impact of tau.
Collapse
Affiliation(s)
- Hsin‐I. Chang
- Department of Neurology, Cognition and Aging CenterKaohsiung Chang Gung Memorial Hospital, Chang Gung University College of MedicineKaohsiungTaiwan
- Institute for Translational Research in Biomedicine, Kaohsiung Chang Gung Memorial HospitalKaohsiungTaiwan
| | - Chi‐Wei Huang
- Department of Neurology, Cognition and Aging CenterKaohsiung Chang Gung Memorial Hospital, Chang Gung University College of MedicineKaohsiungTaiwan
- Institute for Translational Research in Biomedicine, Kaohsiung Chang Gung Memorial HospitalKaohsiungTaiwan
| | - Shu‐Hua Huang
- Department of Nuclear MedicineKaohsiung Chang Gung Memorial Hospital, Chang Gung University College of MedicineKaohsiungTaiwan
| | - Shih‐Wei Hsu
- Department of RadiologyKaohsiung Chang Gung Memorial Hospital, Chang Gung University College of MedicineKaohsiungTaiwan
| | - Kun‐Ju Lin
- Department of Nuclear MedicineLin‐Kou Chang Gung Memorial Hospital, Chang Gung UniversityTaoyuanTaiwan
| | - Tsung‐Ying Ho
- Department of Nuclear MedicineLin‐Kou Chang Gung Memorial Hospital, Chang Gung UniversityTaoyuanTaiwan
| | - Hsiu‐Chuan Wu
- Department of NeurologyLin‐Kou Chang Gung Memorial Hospital, Chang Gung UniversityTaoyuanTaiwan
| | - Chiung‐Chih Chang
- Department of Neurology, Cognition and Aging CenterKaohsiung Chang Gung Memorial Hospital, Chang Gung University College of MedicineKaohsiungTaiwan
- Institute for Translational Research in Biomedicine, Kaohsiung Chang Gung Memorial HospitalKaohsiungTaiwan
- School of Medicine, College of Medicine, National Sun Yat‐sen UniversityKaohsiungTaiwan
| |
Collapse
|
4
|
Guo T, Li A, Sun P, He Z, Cai Y, Lan G, Liu L, Li J, Yang J, Zhu Y, Zhao R, Chen X, Shi D, Liu Z, Wang Q, Xu L, Zhou L, Ran P, Wang X, Sun K, Lu J, Han Y. Astrocyte reactivity is associated with tau tangle load and cortical thinning in Alzheimer's disease. Mol Neurodegener 2024; 19:58. [PMID: 39080744 PMCID: PMC11290175 DOI: 10.1186/s13024-024-00750-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2024] [Accepted: 07/25/2024] [Indexed: 08/02/2024] Open
Abstract
BACKGROUND It is not fully established whether plasma β-amyloid(Aβ)42/Aβ40 and phosphorylated Tau181 (p-Tau181) can effectively detect Alzheimer's disease (AD) pathophysiology in older Chinese adults and how these biomarkers correlate with astrocyte reactivity, Aβ plaque deposition, tau tangle aggregation, and neurodegeneration. METHODS We recruited 470 older adults and analyzed plasma Aβ42/Aβ40, p-Tau181, glial fibrillary acidic protein (GFAP), and neurofilament light (NfL) using the Simoa platform. Among them, 301, 195, and 70 underwent magnetic resonance imaging, Aβ and tau positron emission tomography imaging. The plasma Aβ42/Aβ40 and p-Tau181 thresholds were defined as ≤0.0609 and ≥2.418 based on the receiver operating characteristic curve analysis using the Youden index by comparing Aβ-PET negative cognitively unimpaired individuals and Aβ-PET positive cognitively impaired patients. To evaluate the feasibility of using plasma Aβ42/Aβ40 (A) and p-Tau181 (T) to detect AD and understand how astrocyte reactivity affects this process, we compared plasma GFAP, Aβ plaque, tau tangle, plasma NfL, hippocampal volume, and temporal-metaROI cortical thickness between different plasma A/T profiles and explored their relations with each other using general linear models, including age, sex, APOE-ε4, and diagnosis as covariates. RESULTS Plasma A+/T + individuals showed the highest levels of astrocyte reactivity, Aβ plaque, tau tangle, and axonal degeneration, and the lowest hippocampal volume and temporal-metaROI cortical thickness. Lower plasma Aβ42/Aβ40 and higher plasma p-Tau181 were independently and synergistically correlated with higher plasma GFAP and Aβ plaque. Elevated plasma p-Tau181 and GFAP concentrations were directly and interactively associated with more tau tangle formation. Regarding neurodegeneration, higher plasma p-Tau181 and GFAP concentrations strongly correlated with more axonal degeneration, as measured by plasma NfL, and lower plasma Aβ42/Aβ40 and higher plasma p-Tau181 were related to greater hippocampal atrophy. Higher plasma GFAP levels were associated with thinner cortical thickness and significantly interacted with lower plasma Aβ42/Aβ40 and higher plasma p-Tau181 in predicting more temporal-metaROI cortical thinning. Voxel-wise imaging analysis confirmed these findings. DISCUSSION This study provides a valuable reference for using plasma biomarkers to detect AD in the Chinese community population and offers novel insights into how astrocyte reactivity contributes to AD progression, highlighting the importance of targeting reactive astrogliosis to prevent AD.
Collapse
Affiliation(s)
- Tengfei Guo
- Institute of Biomedical Engineering, Shenzhen Bay Laboratory, No.5 Kelian Road, Shenzhen, 518132, China.
- Institute of Biomedical Engineering, Peking University Shenzhen Graduate School, Shenzhen, 518055, China.
| | - Anqi Li
- Institute of Biomedical Engineering, Shenzhen Bay Laboratory, No.5 Kelian Road, Shenzhen, 518132, China
| | - Pan Sun
- Institute of Biomedical Engineering, Shenzhen Bay Laboratory, No.5 Kelian Road, Shenzhen, 518132, China
| | - Zhengbo He
- Institute of Biomedical Engineering, Shenzhen Bay Laboratory, No.5 Kelian Road, Shenzhen, 518132, China
| | - Yue Cai
- Institute of Biomedical Engineering, Shenzhen Bay Laboratory, No.5 Kelian Road, Shenzhen, 518132, China
| | - Guoyu Lan
- Institute of Biomedical Engineering, Shenzhen Bay Laboratory, No.5 Kelian Road, Shenzhen, 518132, China
| | - Lin Liu
- Institute of Biomedical Engineering, Shenzhen Bay Laboratory, No.5 Kelian Road, Shenzhen, 518132, China
| | - Jieyin Li
- Institute of Biomedical Engineering, Shenzhen Bay Laboratory, No.5 Kelian Road, Shenzhen, 518132, China
| | - Jie Yang
- Institute of Biomedical Engineering, Shenzhen Bay Laboratory, No.5 Kelian Road, Shenzhen, 518132, China
- Department of Neurology, Xuanwu Hospital of Capital Medical University, #45 Changchun Street, Xicheng District, Beijing, 100053, China
| | - Yalin Zhu
- Institute of Biomedical Engineering, Shenzhen Bay Laboratory, No.5 Kelian Road, Shenzhen, 518132, China
| | - Ruiyue Zhao
- Department of Nuclear Medicine, The First Affiliated Hospital, Guangzhou Medical University, Guangzhou, 510120, China
| | - Xuhui Chen
- Department of Neurology, Peking University Shenzhen Hospital, Shenzhen, 518000, China
| | - Dai Shi
- Neurology Medicine Center, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, 518000, China
| | - Zhen Liu
- Institute of Biomedical Engineering, Shenzhen Bay Laboratory, No.5 Kelian Road, Shenzhen, 518132, China
| | - Qingyong Wang
- Department of Neurology, Shenzhen Guangming District People's Hospital, Shenzhen, 518107, China
| | - Linsen Xu
- Department of Medical Imaging, Shenzhen Guangming District People's Hospital, Shenzhen, 518106, China
| | - Liemin Zhou
- Neurology Medicine Center, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, 518000, China
| | - Pengcheng Ran
- Department of Nuclear Medicine, Guangdong Hospital of Traditional Chinese Medicine, Guangzhou, 510120, China
| | - Xinlu Wang
- Department of Nuclear Medicine, The First Affiliated Hospital, Guangzhou Medical University, Guangzhou, 510120, China
| | - Kun Sun
- Institute of Cancer Research, Shenzhen Bay Laboratory, Shenzhen, 518132, China
| | - Jie Lu
- Department of Neurology, Xuanwu Hospital of Capital Medical University, #45 Changchun Street, Xicheng District, Beijing, 100053, China.
- Department of Radiology and Nuclear Medicine, Xuanwu Hospital, Capital Medical University, #45 Changchun Street, Xicheng District, Beijing, 100053, China.
| | - Ying Han
- Institute of Biomedical Engineering, Shenzhen Bay Laboratory, No.5 Kelian Road, Shenzhen, 518132, China.
- Department of Neurology, Xuanwu Hospital of Capital Medical University, #45 Changchun Street, Xicheng District, Beijing, 100053, China.
- School of Biomedical Engineering, Hainan University, Haikou, 570228, China.
- Center of Alzheimer's Disease, Beijing Institute for Brain Disorders, Beijing, 100053, China.
- National Clinical Research Center for Geriatric Diseases, Beijing, 100053, China.
| |
Collapse
|
5
|
Wang W, Huang J, Qian S, Zheng Y, Yu X, Jiang T, Ai R, Hou J, Ma E, Cai J, He H, Wang X, Xie C. Amyloid-β but not tau accumulation is strongly associated with longitudinal cognitive decline. CNS Neurosci Ther 2024; 30:e14860. [PMID: 39014268 PMCID: PMC11251873 DOI: 10.1111/cns.14860] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Revised: 06/11/2024] [Accepted: 07/03/2024] [Indexed: 07/18/2024] Open
Abstract
OBJECTIVE Alzheimer's disease (AD) pathology is featured by the extracellular accumulation of amyloid-β (Aβ) plaques and intracellular tau neurofibrillary tangles in the brain. We studied whether Aβ and tau accumulation are independently associated with future cognitive decline in the AD continuum. METHODS Data were acquired from the Alzheimer's Disease Neuroimaging Initiative (ADNI) public database. A total of 1272 participants were selected based on the availability of Aβ-PET and CSF tau at baseline and of those 777 participants with follow-up visits. RESULTS We found that Aβ-PET and CSF tau pathology were related to cognitive decline across the AD clinical spectrum, both as potential predictors for dementia progression. Among them, Aβ-PET (A + T- subjects) is an independent reliable predictor of longitudinal cognitive decline in terms of ADAS-13, ADNI-MEM, and MMSE scores rather than tau pathology (A - T+ subjects), indicating tau accumulation is not closely correlated with future cognitive impairment without being driven by Aβ deposition. Of note, a high percentage of APOE ε4 carriers with Aβ pathology (A+) develop poor memory and learning capacity. Interestingly, this condition is not recurrence in terms of the ADNI-MEM domain when adding APOE ε4 status. Finally, the levels of Aβ-PET SUVR related to glucose hypometabolism more strongly in subjects with A + T- than A - T+ both happen at baseline and longitudinal changes. CONCLUSIONS In conclusion, Aβ-PET alone without tau pathology (A + T-) measure is an independent reliable predictor of longitudinal cognitive decline but may nonetheless forecast different status of dementia progression. However, tau accumulation alone without Aβ pathology background (A - T+) was not enough to be an independent predictor of cognitive worsening.
Collapse
Affiliation(s)
- Wenwen Wang
- The Center of Traditional Chinese Medicine, The Second Affiliated HospitalYuying Children's Hospital of Wenzhou Medical UniversityWenzhouChina
| | - Jiani Huang
- Department of NeurologyThe First Affiliated Hospital of Wenzhou Medical UniversityWenzhouChina
| | - Shuangjie Qian
- Department of NeurologyThe First Affiliated Hospital of Wenzhou Medical UniversityWenzhouChina
| | - Yi Zheng
- Department of NeurologyThe First Affiliated Hospital of Wenzhou Medical UniversityWenzhouChina
| | - Xinyue Yu
- Alberta InstituteWenzhou Medical UniversityWenzhouZhejiangChina
| | - Tao Jiang
- Department of NeurologyThe First Affiliated Hospital of Wenzhou Medical UniversityWenzhouChina
| | - Ruixue Ai
- Department of Clinical Molecular Biology, Akershus University HospitalUniversity of OsloLørenskogNorway
| | - Jialong Hou
- Department of NeurologyThe First Affiliated Hospital of Wenzhou Medical UniversityWenzhouChina
| | - Enzi Ma
- Department of NeurologyTraditional Chinese and Western Medicine Hospital of WenzhouWenzhouZhejiangChina
| | - Jinlai Cai
- Department of NeurologyThe First Affiliated Hospital of Wenzhou Medical UniversityWenzhouChina
| | - Haijun He
- Department of NeurologyThe First Affiliated Hospital of Wenzhou Medical UniversityWenzhouChina
| | - XinShi Wang
- Department of NeurologyThe First Affiliated Hospital of Wenzhou Medical UniversityWenzhouChina
| | - Chenglong Xie
- Department of NeurologyThe First Affiliated Hospital of Wenzhou Medical UniversityWenzhouChina
- Oujiang LaboratoryWenzhouZhejiangChina
- Key Laboratory of Alzheimer's Disease of Zhejiang Province, Institute of AgingWenzhou Medical UniversityWenzhouZhejiangChina
- Department of Geriatrics, Geriatric Medical CenterThe First Affiliated Hospital of Wenzhou Medical UniversityWenzhouZhejiangChina
| |
Collapse
|
6
|
Chen H, Yang A, Huang W, Du L, Liu B, Lv K, Luan J, Hu P, Shmuel A, Shu N, Ma G. Associations of quantitative susceptibility mapping with cortical atrophy and brain connectome in Alzheimer's disease: A multi-parametric study. Neuroimage 2024; 290:120555. [PMID: 38447683 DOI: 10.1016/j.neuroimage.2024.120555] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2023] [Revised: 01/07/2024] [Accepted: 02/24/2024] [Indexed: 03/08/2024] Open
Abstract
Aberrant susceptibility due to iron level abnormality and brain network disconnections are observed in Alzheimer's disease (AD), with disrupted iron homeostasis hypothesized to be linked to AD pathology and neuronal loss. However, whether associations exist between abnormal quantitative susceptibility mapping (QSM), brain atrophy, and altered brain connectome in AD remains unclear. Based on multi-parametric brain imaging data from 30 AD patients and 26 healthy controls enrolled at the China-Japan Friendship Hospital, we investigated the abnormality of the QSM signal and volumetric measure across 246 brain regions in AD patients. The structural and functional connectomes were constructed based on diffusion MRI tractography and functional connectivity, respectively. The network topology was quantified using graph theory analyses. We identified seven brain regions with both reduced cortical thickness and abnormal QSM (p < 0.05) in AD, including the right superior frontal gyrus, left superior temporal gyrus, right fusiform gyrus, left superior parietal lobule, right superior parietal lobule, left inferior parietal lobule, and left precuneus. Correlations between cortical thickness and network topology computed across patients in the AD group resulted in statistically significant correlations in five of these regions, with higher correlations in functional compared to structural topology. We computed the correlation between network topological metrics, QSM value and cortical thickness across regions at both individual and group-averaged levels, resulting in a measure we call spatial correlations. We found a decrease in the spatial correlation of QSM and the global efficiency of the structural network in AD patients at the individual level. These findings may provide insights into the complex relationships among QSM, brain atrophy, and brain connectome in AD.
Collapse
Affiliation(s)
- Haojie Chen
- State Key Laboratory of Cognitive Neuroscience and Learning & IDG/McGovern Institute for Brain Research, Beijing Normal University, Beijing 100875, China; BABRI Centre, Beijing Normal University, Beijing, China; Beijing Key Laboratory of Brain Imaging and Connectomics, Beijing Normal University, Beijing, China
| | - Aocai Yang
- Department of Radiology, China-Japan Friendship Hospital, Beijing 100029, China; China-Japan Friendship Hospital (Institute of Clinical Medical Sciences), Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Weijie Huang
- State Key Laboratory of Cognitive Neuroscience and Learning & IDG/McGovern Institute for Brain Research, Beijing Normal University, Beijing 100875, China; BABRI Centre, Beijing Normal University, Beijing, China; Beijing Key Laboratory of Brain Imaging and Connectomics, Beijing Normal University, Beijing, China
| | - Lei Du
- Department of Radiology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital & Institute, Beijing, China
| | - Bing Liu
- Department of Radiology, China-Japan Friendship Hospital, Beijing 100029, China; China-Japan Friendship Hospital (Institute of Clinical Medical Sciences), Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Kuan Lv
- Department of Radiology, China-Japan Friendship Hospital, Beijing 100029, China
| | - Jixin Luan
- Department of Radiology, China-Japan Friendship Hospital, Beijing 100029, China; China-Japan Friendship Hospital (Institute of Clinical Medical Sciences), Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Pianpian Hu
- Department of Radiology, China-Japan Friendship Hospital, Beijing 100029, China
| | - Amir Shmuel
- McConnell Brain Imaging Centre, Montreal Neurological Institute, McGill University, Montreal, QC, Canada; Departments of Neurology and Neurosurgery, Physiology, and Biomedical Engineering, McGill University, Montreal, QC, Canada
| | - Ni Shu
- State Key Laboratory of Cognitive Neuroscience and Learning & IDG/McGovern Institute for Brain Research, Beijing Normal University, Beijing 100875, China; BABRI Centre, Beijing Normal University, Beijing, China; Beijing Key Laboratory of Brain Imaging and Connectomics, Beijing Normal University, Beijing, China.
| | - Guolin Ma
- Department of Radiology, China-Japan Friendship Hospital, Beijing 100029, China; China-Japan Friendship Hospital (Institute of Clinical Medical Sciences), Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China.
| |
Collapse
|
7
|
Lane RM, Darreh-Shori T, Junge C, Li D, Yang Q, Edwards AL, Graham DL, Moore K, Mummery CJ. Onset of Alzheimer disease in apolipoprotein ɛ4 carriers is earlier in butyrylcholinesterase K variant carriers. BMC Neurol 2024; 24:116. [PMID: 38594621 PMCID: PMC11003149 DOI: 10.1186/s12883-024-03611-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Accepted: 03/26/2024] [Indexed: 04/11/2024] Open
Abstract
BACKGROUND The authors sought to examine the impact of the K-variant of butyrylcholinesterase (BCHE-K) carrier status on age-at-diagnosis of Alzheimer disease (AD) in APOE4 carriers. METHODS Patients aged 50-74 years with cerebrospinal fluid (CSF) biomarker-confirmed AD, were recruited to clinical trial (NCT03186989 since June 14, 2017). Baseline demographics, disease characteristics, and biomarkers were evaluated in 45 patients according to BCHE-K and APOE4 allelic status in this post-hoc study. RESULTS In APOE4 carriers (N = 33), the mean age-at-diagnosis of AD in BCHE-K carriers (n = 11) was 6.4 years earlier than in BCHE-K noncarriers (n = 22, P < .001, ANOVA). In APOE4 noncarriers (N = 12) there was no observed influence of BCHE-K. APOE4 carriers with BCHE-K also exhibited slightly higher amyloid and tau accumulations compared to BCHE-K noncarriers. A predominantly amyloid, limited tau, and limbic-amnestic phenotype was exemplified by APOE4 homozygotes with BCHE-K. In the overall population, multiple regression analyses demonstrated an association of amyloid accumulation with APOE4 carrier status (P < .029), larger total brain ventricle volume (P < .021), less synaptic injury (Ng, P < .001), and less tau pathophysiology (p-tau181, P < .005). In contrast, tau pathophysiology was associated with more neuroaxonal damage (NfL, P = .002), more synaptic injury (Ng, P < .001), and higher levels of glial activation (YKL-40, P = .01). CONCLUSION These findings have implications for the genetic architecture of prognosis in early AD, not the genetics of susceptibility to AD. In patients with early AD aged less than 75 years, the mean age-at-diagnosis of AD in APOE4 carriers was reduced by over 6 years in BCHE-K carriers versus noncarriers. The functional status of glia may explain many of the effects of APOE4 and BCHE-K on the early AD phenotype. TRIAL REGISTRATION NCT03186989 since June 14, 2017.
Collapse
Affiliation(s)
- Roger M Lane
- Ionis Pharmaceuticals, 2855 Gazelle Court, Carlsbad, CA, 92010, USA.
| | - Taher Darreh-Shori
- Department of Neurobiology, Care Sciences and Society, Center for Alzheimer Research, Division of Clinical Geriatric, Karolinska Institutet, Stockholm, Sweden
| | - Candice Junge
- Ionis Pharmaceuticals, 2855 Gazelle Court, Carlsbad, CA, 92010, USA
| | - Dan Li
- Ionis Pharmaceuticals, 2855 Gazelle Court, Carlsbad, CA, 92010, USA
| | - Qingqing Yang
- Ionis Pharmaceuticals, 2855 Gazelle Court, Carlsbad, CA, 92010, USA
| | | | | | - Katrina Moore
- Ionis Pharmaceuticals, 2855 Gazelle Court, Carlsbad, CA, 92010, USA
| | | |
Collapse
|
8
|
Koutsodendris N, Blumenfeld J, Agrawal A, Traglia M, Yip O, Rao A, Kim MJ, Nelson MR, Wang YH, Grone B, Hao Y, Thomas R, Zilberter M, Yoon SY, Arriola P, Huang Y. APOE4-promoted gliosis and degeneration in tauopathy are ameliorated by pharmacological inhibition of HMGB1 release. Cell Rep 2023; 42:113252. [PMID: 37863057 PMCID: PMC10873109 DOI: 10.1016/j.celrep.2023.113252] [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: 12/22/2022] [Revised: 07/21/2023] [Accepted: 09/26/2023] [Indexed: 10/22/2023] Open
Abstract
Apolipoprotein E4 (APOE4) is an important driver of Tau pathology, gliosis, and degeneration in Alzheimer's disease (AD). Still, the mechanisms underlying these APOE4-driven pathological effects remain elusive. Here, we report in a tauopathy mouse model that APOE4 promoted the nucleocytoplasmic translocation and release of high-mobility group box 1 (HMGB1) from hippocampal neurons, which correlated with the severity of hippocampal microgliosis and degeneration. Injection of HMGB1 into the hippocampus of young APOE4-tauopathy mice induced considerable and persistent gliosis. Selective removal of neuronal APOE4 reduced HMGB1 translocation and release. Treatment of APOE4-tauopathy mice with HMGB1 inhibitors effectively blocked the intraneuronal translocation and release of HMGB1 and ameliorated the development of APOE4-driven gliosis, Tau pathology, neurodegeneration, and myelin deficits. Single-nucleus RNA sequencing revealed that treatment with HMGB1 inhibitors diminished disease-associated and enriched disease-protective subpopulations of neurons, microglia, and astrocytes in APOE4-tauopathy mice. Thus, HMGB1 inhibitors represent a promising approach for treating APOE4-related AD.
Collapse
Affiliation(s)
- Nicole Koutsodendris
- Gladstone Institute of Neurological Disease, Gladstone Institutes, San Francisco, CA 94158, USA; Developmental and Stem Cell Biology Graduate Program, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Jessica Blumenfeld
- Gladstone Institute of Neurological Disease, Gladstone Institutes, San Francisco, CA 94158, USA; Neuroscience Graduate Program, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Ayushi Agrawal
- Gladstone Institute of Data Science and Biotechnology, Gladstone Institutes, San Francisco, CA 94158, USA
| | - Michela Traglia
- Gladstone Institute of Data Science and Biotechnology, Gladstone Institutes, San Francisco, CA 94158, USA
| | - Oscar Yip
- Gladstone Institute of Neurological Disease, Gladstone Institutes, San Francisco, CA 94158, USA; Biomedical Sciences Graduate Program, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Antara Rao
- Gladstone Institute of Neurological Disease, Gladstone Institutes, San Francisco, CA 94158, USA; Developmental and Stem Cell Biology Graduate Program, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Min Joo Kim
- Gladstone Institute of Neurological Disease, Gladstone Institutes, San Francisco, CA 94158, USA; Biomedical Sciences Graduate Program, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Maxine R Nelson
- Gladstone Institute of Neurological Disease, Gladstone Institutes, San Francisco, CA 94158, USA; Biomedical Sciences Graduate Program, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Yung-Hua Wang
- Gladstone Institute of Neurological Disease, Gladstone Institutes, San Francisco, CA 94158, USA; Gladstone Center for Translational Advancement, Gladstone Institutes, San Francisco, CA 94158, USA
| | - Brian Grone
- Gladstone Institute of Neurological Disease, Gladstone Institutes, San Francisco, CA 94158, USA; Gladstone Center for Translational Advancement, Gladstone Institutes, San Francisco, CA 94158, USA
| | - Yanxia Hao
- Gladstone Institute of Neurological Disease, Gladstone Institutes, San Francisco, CA 94158, USA; Gladstone Center for Translational Advancement, Gladstone Institutes, San Francisco, CA 94158, USA
| | - Reuben Thomas
- Gladstone Institute of Data Science and Biotechnology, Gladstone Institutes, San Francisco, CA 94158, USA
| | - Misha Zilberter
- Gladstone Institute of Neurological Disease, Gladstone Institutes, San Francisco, CA 94158, USA
| | - Seo Yeon Yoon
- Gladstone Institute of Neurological Disease, Gladstone Institutes, San Francisco, CA 94158, USA
| | - Patrick Arriola
- Gladstone Institute of Neurological Disease, Gladstone Institutes, San Francisco, CA 94158, USA
| | - Yadong Huang
- Gladstone Institute of Neurological Disease, Gladstone Institutes, San Francisco, CA 94158, USA; Developmental and Stem Cell Biology Graduate Program, University of California, San Francisco, San Francisco, CA 94143, USA; Neuroscience Graduate Program, University of California, San Francisco, San Francisco, CA 94158, USA; Biomedical Sciences Graduate Program, University of California, San Francisco, San Francisco, CA 94143, USA; Gladstone Center for Translational Advancement, Gladstone Institutes, San Francisco, CA 94158, USA; Departments of Neurology and Pathology, University of California, San Francisco, San Francisco, CA 94143, USA.
| |
Collapse
|
9
|
Bocancea DI, Svenningsson AL, van Loenhoud AC, Groot C, Barkhof F, Strandberg O, Smith R, La Joie R, Rosen HJ, Pontecorvo MJ, Rabinovici GD, van der Flier WM, Hansson O, Ossenkoppele R. Determinants of cognitive and brain resilience to tau pathology: a longitudinal analysis. Brain 2023; 146:3719-3734. [PMID: 36967222 PMCID: PMC10473572 DOI: 10.1093/brain/awad100] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2022] [Revised: 02/03/2023] [Accepted: 02/23/2023] [Indexed: 09/03/2023] Open
Abstract
Mechanisms of resilience against tau pathology in individuals across the Alzheimer's disease spectrum are insufficiently understood. Longitudinal data are necessary to reveal which factors relate to preserved cognition (i.e. cognitive resilience) and brain structure (i.e. brain resilience) despite abundant tau pathology, and to clarify whether these associations are cross-sectional or longitudinal. We used a longitudinal study design to investigate the role of several demographic, biological and brain structural factors in yielding cognitive and brain resilience to tau pathology as measured with PET. In this multicentre study, we included 366 amyloid-β-positive individuals with mild cognitive impairment or Alzheimer's disease dementia with baseline 18F-flortaucipir-PET and longitudinal cognitive assessments. A subset (n = 200) additionally underwent longitudinal structural MRI. We used linear mixed-effects models with global cognition and cortical thickness as dependent variables to investigate determinants of cognitive resilience and brain resilience, respectively. Models assessed whether age, sex, years of education, APOE-ε4 status, intracranial volume (and cortical thickness for cognitive resilience models) modified the association of tau pathology with cognitive decline or cortical thinning. We found that the association between higher baseline tau-PET levels (quantified in a temporal meta-region of interest) and rate of cognitive decline (measured with repeated Mini-Mental State Examination) was adversely modified by older age (Stβinteraction = -0.062, P = 0.032), higher education level (Stβinteraction = -0.072, P = 0.011) and higher intracranial volume (Stβinteraction = -0.07, P = 0.016). Younger age, higher education and greater cortical thickness were associated with better cognitive performance at baseline. Greater cortical thickness was furthermore associated with slower cognitive decline independent of tau burden. Higher education also modified the negative impact of tau-PET on cortical thinning, while older age was associated with higher baseline cortical thickness and slower rate of cortical thinning independent of tau. Our analyses revealed no (cross-sectional or longitudinal) associations for sex and APOE-ε4 status on cognition and cortical thickness. In this longitudinal study of clinically impaired individuals with underlying Alzheimer's disease neuropathological changes, we identified education as the most robust determinant of both cognitive and brain resilience against tau pathology. The observed interaction with tau burden on cognitive decline suggests that education may be protective against cognitive decline and brain atrophy at lower levels of tau pathology, with a potential depletion of resilience resources with advancing pathology. Finally, we did not find major contributions of sex to brain nor cognitive resilience, suggesting that previous links between sex and resilience might be mainly driven by cross-sectional differences.
Collapse
Affiliation(s)
- Diana I Bocancea
- Alzheimer Center Amsterdam, Neurology, Vrije Universiteit Amsterdam, Amsterdam UMC location VUmc, 1081 HZ Amsterdam, The Netherlands
- Amsterdam Neuroscience, Neurodegeneration, 1081 HV Amsterdam, The Netherlands
| | | | - Anna C van Loenhoud
- Alzheimer Center Amsterdam, Neurology, Vrije Universiteit Amsterdam, Amsterdam UMC location VUmc, 1081 HZ Amsterdam, The Netherlands
- Amsterdam Neuroscience, Neurodegeneration, 1081 HV Amsterdam, The Netherlands
| | - Colin Groot
- Alzheimer Center Amsterdam, Neurology, Vrije Universiteit Amsterdam, Amsterdam UMC location VUmc, 1081 HZ Amsterdam, The Netherlands
- Amsterdam Neuroscience, Neurodegeneration, 1081 HV Amsterdam, The Netherlands
- Clinical Memory Research Unit, Lund University, 211 46 Lund, Sweden
| | - Frederik Barkhof
- Department of Radiology and Nuclear Medicine, Vrije Universiteit Amsterdam, Amsterdam UMC, 1081 HV Amsterdam, The Netherlands
- Queen Square Institute of Neurology and Center for Medical Image Computing, University College London, London WC1N 3BG, UK
| | - Olof Strandberg
- Clinical Memory Research Unit, Lund University, 211 46 Lund, Sweden
| | - Ruben Smith
- Clinical Memory Research Unit, Lund University, 211 46 Lund, Sweden
- Department of Neurology, Skåne University Hospital, 221 84 Lund, Sweden
| | - Renaud La Joie
- Department of Neurology, Memory and Aging Center, University of California, San Francisco, CA 94158, USA
| | - Howard J Rosen
- Department of Neurology, Memory and Aging Center, University of California, San Francisco, CA 94158, USA
| | | | - Gil D Rabinovici
- Department of Neurology, Memory and Aging Center, University of California, San Francisco, CA 94158, USA
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, CA 94143, USA
- Molecular Biophysics and Integrated Bioimaging Division, Lawrence Berkeley National Laboratory, Berkeley, CA 94720, USA
| | - Wiesje M van der Flier
- Alzheimer Center Amsterdam, Neurology, Vrije Universiteit Amsterdam, Amsterdam UMC location VUmc, 1081 HZ Amsterdam, The Netherlands
- Amsterdam Neuroscience, Neurodegeneration, 1081 HV Amsterdam, The Netherlands
- Department of Epidemiology and Biostatistics, Vrije Universiteit Amsterdam, Amsterdam UMC, 1081 HV Amsterdam, The Netherlands
| | - Oskar Hansson
- Clinical Memory Research Unit, Lund University, 211 46 Lund, Sweden
- Memory Clinic, Skåne University Hospital, 214 28 Malmö, Sweden
| | - Rik Ossenkoppele
- Alzheimer Center Amsterdam, Neurology, Vrije Universiteit Amsterdam, Amsterdam UMC location VUmc, 1081 HZ Amsterdam, The Netherlands
- Amsterdam Neuroscience, Neurodegeneration, 1081 HV Amsterdam, The Netherlands
- Clinical Memory Research Unit, Lund University, 211 46 Lund, Sweden
| |
Collapse
|
10
|
Donato L, Mordà D, Scimone C, Alibrandi S, D'Angelo R, Sidoti A. How Many Alzheimer-Perusini's Atypical Forms Do We Still Have to Discover? Biomedicines 2023; 11:2035. [PMID: 37509674 PMCID: PMC10377159 DOI: 10.3390/biomedicines11072035] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2023] [Revised: 07/17/2023] [Accepted: 07/17/2023] [Indexed: 07/30/2023] Open
Abstract
Alzheimer-Perusini's (AD) disease represents the most spread dementia around the world and constitutes a serious problem for public health. It was first described by the two physicians from whom it took its name. Nowadays, we have extensively expanded our knowledge about this disease. Starting from a merely clinical and histopathologic description, we have now reached better molecular comprehension. For instance, we passed from an old conceptualization of the disease based on plaques and tangles to a more modern vision of mixed proteinopathy in a one-to-one relationship with an alteration of specific glial and neuronal phenotypes. However, no disease-modifying therapies are yet available. It is likely that the only way to find a few "magic bullets" is to deepen this aspect more and more until we are able to draw up specific molecular profiles for single AD cases. This review reports the most recent classifications of AD atypical variants in order to summarize all the clinical evidence using several discrimina (for example, post mortem neurofibrillary tangle density, cerebral atrophy, or FDG-PET studies). The better defined four atypical forms are posterior cortical atrophy (PCA), logopenic variant of primary progressive aphasia (LvPPA), behavioral/dysexecutive variant and AD with corticobasal degeneration (CBS). Moreover, we discuss the usefulness of such classifications before outlining the molecular-genetic aspects focusing on microglial activity or, more generally, immune system control of neuroinflammation and neurodegeneration.
Collapse
Affiliation(s)
- Luigi Donato
- Department of Biomedical and Dental Sciences and Morphofunctional Imaging, Division of Medical Biotechnologies and Preventive Medicine, University of Messina, Via Consolare Valeria 1, 98125 Messina, Italy
- Department of Biomolecular Strategies, Genetics, Cutting-Edge Therapies, Euro-Mediterranean Institute of Science and Technology, Via Michele Miraglia, 98139 Palermo, Italy
| | - Domenico Mordà
- Department of Biomedical and Dental Sciences and Morphofunctional Imaging, Division of Medical Biotechnologies and Preventive Medicine, University of Messina, Via Consolare Valeria 1, 98125 Messina, Italy
- Department of Biomolecular Strategies, Genetics, Cutting-Edge Therapies, Euro-Mediterranean Institute of Science and Technology, Via Michele Miraglia, 98139 Palermo, Italy
| | - Concetta Scimone
- Department of Biomedical and Dental Sciences and Morphofunctional Imaging, Division of Medical Biotechnologies and Preventive Medicine, University of Messina, Via Consolare Valeria 1, 98125 Messina, Italy
- Department of Biomolecular Strategies, Genetics, Cutting-Edge Therapies, Euro-Mediterranean Institute of Science and Technology, Via Michele Miraglia, 98139 Palermo, Italy
| | - Simona Alibrandi
- Department of Biomedical and Dental Sciences and Morphofunctional Imaging, Division of Medical Biotechnologies and Preventive Medicine, University of Messina, Via Consolare Valeria 1, 98125 Messina, Italy
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale Ferdinando Stagno D'Alcontres 31, 98166 Messina, Italy
| | - Rosalia D'Angelo
- Department of Biomedical and Dental Sciences and Morphofunctional Imaging, Division of Medical Biotechnologies and Preventive Medicine, University of Messina, Via Consolare Valeria 1, 98125 Messina, Italy
| | - Antonina Sidoti
- Department of Biomedical and Dental Sciences and Morphofunctional Imaging, Division of Medical Biotechnologies and Preventive Medicine, University of Messina, Via Consolare Valeria 1, 98125 Messina, Italy
| |
Collapse
|
11
|
Lamontagne-Kam D, Ulfat AK, Hervé V, Vu TM, Brouillette J. Implication of tau propagation on neurodegeneration in Alzheimer's disease. Front Neurosci 2023; 17:1219299. [PMID: 37483337 PMCID: PMC10360202 DOI: 10.3389/fnins.2023.1219299] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2023] [Accepted: 06/07/2023] [Indexed: 07/25/2023] Open
Abstract
Propagation of tau fibrils correlate closely with neurodegeneration and memory deficits seen during the progression of Alzheimer's disease (AD). Although it is not well-established what drives or attenuates tau spreading, new studies on human brain using positron emission tomography (PET) have shed light on how tau phosphorylation, genetic factors, and the initial epicenter of tau accumulation influence tau accumulation and propagation throughout the brain. Here, we review the latest PET studies performed across the entire AD continuum looking at the impact of amyloid load on tau pathology. We also explore the effects of structural, functional, and proximity connectivity on tau spreading in a stereotypical manner in the brain of AD patients. Since tau propagation can be quite heterogenous between individuals, we then consider how the speed and pattern of propagation are influenced by the starting localization of tau accumulation in connected brain regions. We provide an overview of some genetic variants that were shown to accelerate or slow down tau spreading. Finally, we discuss how phosphorylation of certain tau epitopes affect the spreading of tau fibrils. Since tau pathology is an early event in AD pathogenesis and is one of the best predictors of neurodegeneration and memory impairments, understanding the process by which tau spread from one brain region to another could pave the way to novel therapeutic avenues that are efficient during the early stages of the disease, before neurodegeneration induces permanent brain damage and severe memory loss.
Collapse
|
12
|
Williams T, Bathe T, Vo Q, Sacilotto P, McFarland K, Ruiz AJ, Hery GP, Sullivan P, Borchelt DR, Prokop S, Chakrabarty P. Humanized APOE genotypes influence lifespan independently of tau aggregation in the P301S mouse model of tauopathy. Acta Neuropathol Commun 2023; 11:99. [PMID: 37337279 DOI: 10.1186/s40478-023-01581-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2023] [Accepted: 05/09/2023] [Indexed: 06/21/2023] Open
Abstract
Apolipoprotein (APOE) E4 isoform is a major risk factor of Alzheimer's disease and contributes to metabolic and neuropathological abnormalities during brain aging. To provide insights into whether APOE4 genotype is related to tau-associated neurodegeneration, we have generated human P301S mutant tau transgenic mice (PS19) that carry humanized APOE alleles (APOE2, APOE3 or APOE4). In aging mice that succumbed to paralysis, PS19 mice homozygous for APOE3 had the longest lifespan when compared to APOE4 and APOE2 homozygous mice (APOE3 > APOE4 ~ APOE2). Heterozygous mice with one human APOE and one mouse Apoe allele did not show any variations in lifespan. At end-stage, PS19 mice homozygous for APOE3 and APOE4 showed equivalent levels of phosphorylated tau burden, inflammation levels and ventricular volumes. Compared to these cohorts, PS19 mice homozygous for APOE2 showed lower induction of phosphorylation on selective epitopes, though the effect sizes were small and variable. In spite of this, the APOE2 cohort showed shorter lifespan relative to APOE3 homozygous mice. None of the cohorts accumulated appreciable levels of phosphorylated tau compartmentalized in the insoluble cell fraction. RNAseq analysis showed that the induction of immune gene expression was comparable across all the APOE genotypes in PS19 mice. Notably, the APOE4 homozygous mice showed additional induction of transcripts corresponding to the Alzheimer's disease-related plaque-induced gene signature. In human Alzheimer's disease brain tissues, we found no direct correlation between higher burden of phosphorylated tau and APOE4 genotype. As expected, there was a strong correlation between phosphorylated tau burden with amyloid deposition in APOE4-positive Alzheimer's disease cases. Overall, our results indicate that APOE3 genotype may confer some resilience to tauopathy, while APOE4 and APOE2 may act through multiple pathways to increase the pathogenicity in the context of tauopathy.
Collapse
Affiliation(s)
- Tristan Williams
- Center for Translational Research in Neurodegenerative Disease, University of Florida, Gainesville, FL, 32610, USA
- Department of Neuroscience, University of Florida, Gainesville, FL, 32610, USA
- Eli Lilly & Company, Indianapolis, IN, 46285, USA
| | - Tim Bathe
- Center for Translational Research in Neurodegenerative Disease, University of Florida, Gainesville, FL, 32610, USA
- Department of Neuroscience, University of Florida, Gainesville, FL, 32610, USA
| | - Quan Vo
- Center for Translational Research in Neurodegenerative Disease, University of Florida, Gainesville, FL, 32610, USA
| | - Patricia Sacilotto
- Center for Translational Research in Neurodegenerative Disease, University of Florida, Gainesville, FL, 32610, USA
- Department of Neuroscience, University of Florida, Gainesville, FL, 32610, USA
| | - Karen McFarland
- Center for Translational Research in Neurodegenerative Disease, University of Florida, Gainesville, FL, 32610, USA
- Department of Neurology, University of Florida, Gainesville, FL, 32610, USA
| | - Alejandra Jolie Ruiz
- Center for Translational Research in Neurodegenerative Disease, University of Florida, Gainesville, FL, 32610, USA
| | - Gabriela P Hery
- Center for Translational Research in Neurodegenerative Disease, University of Florida, Gainesville, FL, 32610, USA
| | | | - David R Borchelt
- Center for Translational Research in Neurodegenerative Disease, University of Florida, Gainesville, FL, 32610, USA
- Department of Neuroscience, University of Florida, Gainesville, FL, 32610, USA
- McKnight Brain Institute, University of Florida, Gainesville, FL, 32610, USA
| | - Stefan Prokop
- Center for Translational Research in Neurodegenerative Disease, University of Florida, Gainesville, FL, 32610, USA
- Department of Pathology, Immunology & Laboratory Medicine, University of Florida, Gainesville, FL, 32610, USA
- Fixel Institute for Neurological Diseases, University of Florida, Gainesville, FL, 32608, USA
| | - Paramita Chakrabarty
- Center for Translational Research in Neurodegenerative Disease, University of Florida, Gainesville, FL, 32610, USA.
- Department of Neuroscience, University of Florida, Gainesville, FL, 32610, USA.
- McKnight Brain Institute, University of Florida, Gainesville, FL, 32610, USA.
| |
Collapse
|
13
|
Schilling KG, Archer D, Rheault F, Lyu I, Huo Y, Cai LY, Bunge SA, Weiner KS, Gore JC, Anderson AW, Landman BA. Superficial white matter across development, young adulthood, and aging: volume, thickness, and relationship with cortical features. Brain Struct Funct 2023; 228:1019-1031. [PMID: 37074446 PMCID: PMC10320929 DOI: 10.1007/s00429-023-02642-x] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2022] [Accepted: 04/08/2023] [Indexed: 04/20/2023]
Abstract
Superficial white matter (SWM) represents a significantly understudied part of the human brain, despite comprising a large portion of brain volume and making up a majority of cortico-cortical white matter connections. Using multiple, high-quality datasets with large sample sizes (N = 2421, age range 5-100) in combination with methodological advances in tractography, we quantified features of SWM volume and thickness across the brain and across development, young adulthood, and aging. We had four primary aims: (1) characterize SWM thickness across brain regions (2) describe associations between SWM volume and age (3) describe associations between SWM thickness and age, and (4) quantify relationships between SWM thickness and cortical features. Our main findings are that (1) SWM thickness varies across the brain, with patterns robust across individuals and across the population at the region-level and vertex-level; (2) SWM volume shows unique volumetric trajectories with age that are distinct from gray matter and other white matter trajectories; (3) SWM thickness shows nonlinear cross-sectional changes across the lifespan that vary across regions; and (4) SWM thickness is associated with features of cortical thickness and curvature. For the first time, we show that SWM volume follows a similar trend as overall white matter volume, peaking at a similar time in adolescence, leveling off throughout adulthood, and decreasing with age thereafter. Notably, the relative fraction of total brain volume of SWM continuously increases with age, and consequently takes up a larger proportion of total white matter volume, unlike the other tissue types that decrease with respect to total brain volume. This study represents the first characterization of SWM features across the large portion of the lifespan and provides the background for characterizing normal aging and insight into the mechanisms associated with SWM development and decline.
Collapse
Affiliation(s)
- Kurt G Schilling
- Department of Radiology and Radiological Sciences, Vanderbilt University Medical Center, Nashville, TN, USA.
- Vanderbilt University Institute of Imaging Science, Vanderbilt University, Nashville, TN, USA.
| | - Derek Archer
- Vanderbilt Memory and Alzheimer's Center, Vanderbilt University Medical Center, Nashville, TN, USA
- Department of Neurology, Vanderbilt University Medical Center, Nashville, TN, USA
- Vanderbilt Genetics Institute, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Francois Rheault
- Department of Electrical Engineering and Computer Engineering, Vanderbilt University, Nashville, TN, USA
| | - Ilwoo Lyu
- Computer Science and Engineering, Ulsan National Institute of Science and Technology, Ulsan, South Korea
| | - Yuankai Huo
- Department of Electrical Engineering and Computer Engineering, Vanderbilt University, Nashville, TN, USA
| | - Leon Y Cai
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, USA
| | - Silvia A Bunge
- Department of Psychology, University of California at Berkeley, Berkeley, USA
| | - Kevin S Weiner
- Department of Psychology, University of California at Berkeley, Berkeley, USA
- Helen Wills Neuroscience Institute, University of California at Berkeley, Berkeley, USA
| | - John C Gore
- Department of Radiology and Radiological Sciences, Vanderbilt University Medical Center, Nashville, TN, USA
- Vanderbilt University Institute of Imaging Science, Vanderbilt University, Nashville, TN, USA
| | - Adam W Anderson
- Vanderbilt University Institute of Imaging Science, Vanderbilt University, Nashville, TN, USA
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, USA
| | - Bennett A Landman
- Department of Radiology and Radiological Sciences, Vanderbilt University Medical Center, Nashville, TN, USA
- Vanderbilt University Institute of Imaging Science, Vanderbilt University, Nashville, TN, USA
- Computer Science and Engineering, Ulsan National Institute of Science and Technology, Ulsan, South Korea
| |
Collapse
|
14
|
Singh NA, Tosakulwong N, Graff-Radford J, Machulda MM, Pham NTT, Sintini I, Weigand SD, Schwarz CG, Senjem ML, Carrasquillo MM, Ertekin-Taner N, Jack CR, Lowe VJ, Josephs KA, Whitwell JL. APOE ε4 influences medial temporal atrophy and tau deposition in atypical Alzheimer's disease. Alzheimers Dement 2023; 19:784-796. [PMID: 35691047 PMCID: PMC9742387 DOI: 10.1002/alz.12711] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Revised: 05/02/2022] [Accepted: 05/04/2022] [Indexed: 12/14/2022]
Abstract
INTRODUCTION Apolipoprotein E (APOE) ε4 is an important genetic risk factor for typical Alzheimer's disease (AD), influencing brain volume and tau burden. Little is known about its influence in atypical presentations of AD. METHODS An atypical AD cohort of 140 patients diagnosed with either posterior cortical atrophy or logopenic progressive aphasia underwent magnetic resonance imaging and positron emission tomography. Linear mixed effects models were fit to assess the influence of APOE ε4 on cross-sectional and longitudinal regional metrics. RESULTS At baseline, APOE ε4 carriers had smaller hippocampal and amygdala volumes and greater tau standardized uptake volume ratio in the hippocampus and entorhinal cortex compared to non-carriers while longitudinally, APOE ε4 non-carriers showed faster rates of atrophy and tau accumulation in the entorhinal cortex, with faster tau accumulation in the hippocampus. DISCUSSION APOE ε4 influences patterns of neurodegeneration and tau deposition and was associated with more medial temporal involvement, although there is evidence that non-carriers may be catching up over time.
Collapse
Affiliation(s)
| | | | | | - Mary M. Machulda
- Department of Psychiatry & Psychology, Mayo Clinic, Rochester, MN, USA
| | | | - Irene Sintini
- Department of Radiology, Mayo Clinic, Rochester, MN, USA
| | - Stephen D. Weigand
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN, USA
| | | | | | | | | | | | - Val J. Lowe
- Department of Radiology, Mayo Clinic, Rochester, MN, USA
| | | | | |
Collapse
|
15
|
Dai WZ, Liu L, Zhu MZ, Lu J, Ni JM, Li R, Ma T, Zhu XC. Morphological and Structural Network Analysis of Sporadic Alzheimer's Disease Brains Based on the APOE4 Gene. J Alzheimers Dis 2023; 91:1035-1048. [PMID: 36530087 DOI: 10.3233/jad-220877] [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: 12/23/2022]
Abstract
BACKGROUND Alzheimer's disease (AD) is an increasingly common type of dementia. Apolipoprotein E (APOE) gene is a strong risk factor for AD. OBJECTIVE Here, we explored alterations in grey matter structure (GMV) and networks in AD, as well as the effects of the APOEɛ4 allele on neuroimaging regions based on structural magnetic resonance imaging (sMRI). METHODS All subjects underwent an sMRI scan. GMV and cortical thickness were calculated using voxel-based morphological analysis, and structural networks were constructed based on graph theory analysis to compare differences between AD and normal controls. RESULTS The volumes of grey matter in the bilateral inferior temporal gyrus, right middle temporal gyrus, right inferior parietal lobule, right limbic lobe, right frontal lobe, left anterior cingulate gyrus, and bilateral olfactory cortex of patients with AD were significantly decreased. The cortical thickness in patients with AD was significantly reduced in the left lateral occipital lobe, inferior parietal lobe, orbitofrontal region, precuneus, superior parietal gyrus, right precentral gyrus, middle temporal gyrus, pars opercularis gyrus, insular gyrus, superior marginal gyrus, bilateral fusiform gyrus, and superior frontal gyrus. In terms of local properties, there were significant differences between the AD and control groups in these areas, including the right bank, right temporalis pole, bilateral middle temporal gyrus, right transverse temporal gyrus, left postcentral gyrus, and left parahippocampal gyrus. CONCLUSION There were significant differences in the morphological and structural covariate networks between AD patients and healthy controls under APOEɛ4 allele effects.
Collapse
Affiliation(s)
- Wen-Zhuo Dai
- Department of Neurology, Affiliated Wuxi Clinical College of Nantong University, Wuxi, Jiangsu Province, China.,Department of Neurology, The Affiliated Wuxi No. 2 People's Hospital of Nanjing Medical University, Wuxi, Jiangsu Province, China.,Department of Neurology, Affiliated Wuxi No. 2 Hospital of Jiangnan University, Wuxi, Jiangsu Province, China
| | - Lu Liu
- Department of Neurology, The Affiliated Wuxi No. 2 People's Hospital of Nanjing Medical University, Wuxi, Jiangsu Province, China
| | - Meng-Zhuo Zhu
- Department of Neurology, Affiliated Wuxi Clinical College of Nantong University, Wuxi, Jiangsu Province, China
| | - Jing Lu
- Department of Neurology, The Affiliated Wuxi No. 2 People's Hospital of Nanjing Medical University, Wuxi, Jiangsu Province, China
| | - Jian-Ming Ni
- Radiology Department, Nanjing Medical University, Wuxi, Jiangsu Province, China
| | - Rong Li
- Department of Pharmacy, The Affiliated Wuxi No. 2 People's Hospital of Nanjing Medical University, Wuxi, Jiangsu Province, China
| | - Tao Ma
- Department of Neurology, Affiliated Wuxi Clinical College of Nantong University, Wuxi, Jiangsu Province, China.,Department of Neurology, The Affiliated Wuxi No. 2 People's Hospital of Nanjing Medical University, Wuxi, Jiangsu Province, China.,Department of Neurology, Affiliated Wuxi No. 2 Hospital of Jiangnan University, Wuxi, Jiangsu Province, China
| | - Xi-Chen Zhu
- Department of Neurology, Affiliated Wuxi Clinical College of Nantong University, Wuxi, Jiangsu Province, China.,Department of Neurology, The Affiliated Wuxi No. 2 People's Hospital of Nanjing Medical University, Wuxi, Jiangsu Province, China.,Department of Neurology, Affiliated Wuxi No. 2 Hospital of Jiangnan University, Wuxi, Jiangsu Province, China
| |
Collapse
|
16
|
Hnilicova P, Kantorova E, Sutovsky S, Grofik M, Zelenak K, Kurca E, Zilka N, Parvanovova P, Kolisek M. Imaging Methods Applicable in the Diagnostics of Alzheimer's Disease, Considering the Involvement of Insulin Resistance. Int J Mol Sci 2023; 24:3325. [PMID: 36834741 PMCID: PMC9958721 DOI: 10.3390/ijms24043325] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2022] [Revised: 01/29/2023] [Accepted: 01/30/2023] [Indexed: 02/10/2023] Open
Abstract
Alzheimer's disease (AD) is an incurable neurodegenerative disease and the most frequently diagnosed type of dementia, characterized by (1) perturbed cerebral perfusion, vasculature, and cortical metabolism; (2) induced proinflammatory processes; and (3) the aggregation of amyloid beta and hyperphosphorylated Tau proteins. Subclinical AD changes are commonly detectable by using radiological and nuclear neuroimaging methods such as magnetic resonance imaging (MRI), computed tomography (CT), positron emission tomography (PET), and single-photon emission computed tomography (SPECT). Furthermore, other valuable modalities exist (in particular, structural volumetric, diffusion, perfusion, functional, and metabolic magnetic resonance methods) that can advance the diagnostic algorithm of AD and our understanding of its pathogenesis. Recently, new insights into AD pathoetiology revealed that deranged insulin homeostasis in the brain may play a role in the onset and progression of the disease. AD-related brain insulin resistance is closely linked to systemic insulin homeostasis disorders caused by pancreas and/or liver dysfunction. Indeed, in recent studies, linkages between the development and onset of AD and the liver and/or pancreas have been established. Aside from standard radiological and nuclear neuroimaging methods and clinically fewer common methods of magnetic resonance, this article also discusses the use of new suggestive non-neuronal imaging modalities to assess AD-associated structural changes in the liver and pancreas. Studying these changes might be of great clinical importance because of their possible involvement in AD pathogenesis during the prodromal phase of the disease.
Collapse
Affiliation(s)
- Petra Hnilicova
- Biomedical Center Martin, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, 036 01 Martin, Slovakia
| | - Ema Kantorova
- Clinic of Neurology, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, 036 01 Martin, Slovakia
| | - Stanislav Sutovsky
- 1st Department of Neurology, Faculty of Medicine, Comenius University in Bratislava and University Hospital, 813 67 Bratislava, Slovakia
| | - Milan Grofik
- Clinic of Neurology, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, 036 01 Martin, Slovakia
| | - Kamil Zelenak
- Clinic of Radiology, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, 036 01 Martin, Slovakia
| | - Egon Kurca
- Clinic of Neurology, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, 036 01 Martin, Slovakia
| | - Norbert Zilka
- Institute of Neuroimmunology, Slovak Academy of Sciences, 845 10 Bratislava, Slovakia
| | - Petra Parvanovova
- Department of Medical Biochemistry, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, 036 01 Martin, Slovakia
| | - Martin Kolisek
- Biomedical Center Martin, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, 036 01 Martin, Slovakia
| |
Collapse
|
17
|
Young CB, Johns E, Kennedy G, Belloy ME, Insel PS, Greicius MD, Sperling RA, Johnson KA, Poston KL, Mormino EC. APOE effects on regional tau in preclinical Alzheimer's disease. Mol Neurodegener 2023; 18:1. [PMID: 36597122 PMCID: PMC9811772 DOI: 10.1186/s13024-022-00590-4] [Citation(s) in RCA: 30] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2022] [Accepted: 12/14/2022] [Indexed: 01/05/2023] Open
Abstract
BACKGROUND APOE variants are strongly associated with abnormal amyloid aggregation and additional direct effects of APOE on tau aggregation are reported in animal and human cell models. The degree to which these effects are present in humans when individuals are clinically unimpaired (CU) but have abnormal amyloid (Aβ+) remains unclear. METHODS We analyzed data from CU individuals in the Anti-Amyloid Treatment in Asymptomatic AD (A4) and Longitudinal Evaluation of Amyloid Risk and Neurodegeneration (LEARN) studies. Amyloid PET data were available for 4486 participants (3163 Aβ-, 1323 Aβ+) and tau PET data were available for a subset of 447 participants (55 Aβ-, 392 Aβ+). Linear models examined APOE (number of e2 and e4 alleles) associations with global amyloid and regional tau burden in medial temporal lobe (entorhinal, amygdala) and early neocortical regions (inferior temporal, inferior parietal, precuneus). Consistency of APOE4 effects on regional tau were examined in 220 Aβ + CU and mild cognitive impairment (MCI) participants from the Alzheimer's Disease Neuroimaging Initiative (ADNI). RESULTS APOE2 and APOE4 were associated with lower and higher amyloid positivity rates, respectively. Among Aβ+ CU, e2 and e4 were associated with reduced (-12 centiloids per allele) and greater (+15 centiloids per allele) continuous amyloid burden, respectively. APOE2 was associated with reduced regional tau in all regions (-0.05 to -0.09 SUVR per allele), whereas APOE4 was associated with greater regional tau (+0.02 to +0.07 SUVR per allele). APOE differences were confirmed by contrasting e3/e3 with e2/e3 and e3/e4. Mediation analyses among Aβ+ s showed that direct effects of e2 on regional tau were present in medial temporal lobe and early neocortical regions, beyond an indirect pathway mediated by continuous amyloid burden. For e4, direct effects on regional tau were only significant in medial temporal lobe. The magnitude of protective e2 effects on regional tau was consistent across brain regions, whereas detrimental e4 effects were greatest in medial temporal lobe. APOE4 patterns were confirmed in Aβ+ ADNI participants. CONCLUSIONS APOE influences early regional tau PET burden, above and beyond effects related to cross-sectional amyloid PET burden. Therapeutic strategies targeting underlying mechanisms related to APOE may modify tau accumulation among Aβ+ individuals.
Collapse
Affiliation(s)
- Christina B Young
- Stanford University School of Medicine, 453 Quarry Rd., Palo Alto, Stanford, CA, 94304, USA.
| | - Emily Johns
- Stanford University School of Medicine, 453 Quarry Rd., Palo Alto, Stanford, CA, 94304, USA
| | - Gabriel Kennedy
- Stanford University School of Medicine, 453 Quarry Rd., Palo Alto, Stanford, CA, 94304, USA
| | - Michael E Belloy
- Stanford University School of Medicine, 453 Quarry Rd., Palo Alto, Stanford, CA, 94304, USA
| | - Philip S Insel
- University of California San Francisco, San Francisco, CA, USA
| | - Michael D Greicius
- Stanford University School of Medicine, 453 Quarry Rd., Palo Alto, Stanford, CA, 94304, USA
| | - Reisa A Sperling
- Brigham and Women's Hospital, Boston, MA, USA
- Massachusetts General Hospital, Boston, MA, USA
| | - Keith A Johnson
- Brigham and Women's Hospital, Boston, MA, USA
- Massachusetts General Hospital, Boston, MA, USA
| | - Kathleen L Poston
- Stanford University School of Medicine, 453 Quarry Rd., Palo Alto, Stanford, CA, 94304, USA
| | - Elizabeth C Mormino
- Stanford University School of Medicine, 453 Quarry Rd., Palo Alto, Stanford, CA, 94304, USA
| |
Collapse
|
18
|
Dincer A, Chen CD, McKay NS, Koenig LN, McCullough A, Flores S, Keefe SJ, Schultz SA, Feldman RL, Joseph-Mathurin N, Hornbeck RC, Cruchaga C, Schindler SE, Holtzman DM, Morris JC, Fagan AM, Benzinger TLS, Gordon BA. APOE ε4 genotype, amyloid-β, and sex interact to predict tau in regions of high APOE mRNA expression. Sci Transl Med 2022; 14:eabl7646. [PMID: 36383681 PMCID: PMC9912474 DOI: 10.1126/scitranslmed.abl7646] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
The apolipoprotein E (APOE) ε4 allele is strongly linked with cerebral β-amyloidosis, but its relationship with tauopathy is less established. We investigated the relationship between APOE ε4 carrier status, regional amyloid-β (Aβ), magnetic resonance imaging (MRI) volumetrics, tau positron emission tomography (PET), APOE messenger RNA (mRNA) expression maps, and cerebrospinal fluid phosphorylated tau (CSF ptau181). Three hundred fifty participants underwent imaging, and 270 had ptau181. We used computational models to evaluate the main effect of APOE ε4 carrier status on regional neuroimaging values and then the interaction of ε4 status and global Aβ on regional tau PET and brain volumes as well as CSF ptau181. Separately, we also examined the additional interactive influence of sex. We found that, for the same degree of Aβ burden, APOE ε4 carriers showed greater tau PET signal relative to noncarriers in temporal regions, but no interaction was present for MRI volumes or CSF ptau181. This potentiation of tau aggregation irrespective of sex occurred in brain regions with high APOE mRNA expression, suggesting local vulnerabilities to tauopathy. There were greater effects of APOE genotype in females, although the interactive sex effects did not strongly mirror mRNA expression. Pathology is not homogeneously expressed throughout the brain but mirrors underlying biological patterns such as gene expression.
Collapse
Affiliation(s)
- Aylin Dincer
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, Saint Louis, MO, USA.,Knight Alzheimer Disease Research Center, Washington University School of Medicine, Saint Louis, MO, USA
| | - Charles D Chen
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, Saint Louis, MO, USA.,Knight Alzheimer Disease Research Center, Washington University School of Medicine, Saint Louis, MO, USA
| | - Nicole S McKay
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, Saint Louis, MO, USA.,Knight Alzheimer Disease Research Center, Washington University School of Medicine, Saint Louis, MO, USA
| | - Lauren N Koenig
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, Saint Louis, MO, USA.,Knight Alzheimer Disease Research Center, Washington University School of Medicine, Saint Louis, MO, USA
| | - Austin McCullough
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, Saint Louis, MO, USA.,Knight Alzheimer Disease Research Center, Washington University School of Medicine, Saint Louis, MO, USA
| | - Shaney Flores
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, Saint Louis, MO, USA.,Knight Alzheimer Disease Research Center, Washington University School of Medicine, Saint Louis, MO, USA
| | - Sarah J Keefe
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, Saint Louis, MO, USA.,Knight Alzheimer Disease Research Center, Washington University School of Medicine, Saint Louis, MO, USA
| | - Stephanie A Schultz
- Knight Alzheimer Disease Research Center, Washington University School of Medicine, Saint Louis, MO, USA
| | - Rebecca L Feldman
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, Saint Louis, MO, USA.,Knight Alzheimer Disease Research Center, Washington University School of Medicine, Saint Louis, MO, USA
| | - Nelly Joseph-Mathurin
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, Saint Louis, MO, USA.,Knight Alzheimer Disease Research Center, Washington University School of Medicine, Saint Louis, MO, USA
| | - Russ C Hornbeck
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, Saint Louis, MO, USA.,Knight Alzheimer Disease Research Center, Washington University School of Medicine, Saint Louis, MO, USA
| | - Carlos Cruchaga
- Knight Alzheimer Disease Research Center, Washington University School of Medicine, Saint Louis, MO, USA.,Department of Psychiatry, Washington University School of Medicine, Saint Louis, MO, USA
| | - Suzanne E Schindler
- Knight Alzheimer Disease Research Center, Washington University School of Medicine, Saint Louis, MO, USA.,Department of Neurology, Washington University School of Medicine, Saint Louis, MO, USA
| | - David M Holtzman
- Knight Alzheimer Disease Research Center, Washington University School of Medicine, Saint Louis, MO, USA.,Department of Neurology, Washington University School of Medicine, Saint Louis, MO, USA.,Hope Center for Neurological Disorders, Washington University School of Medicine, Saint Louis, MO, USA
| | - John C Morris
- Knight Alzheimer Disease Research Center, Washington University School of Medicine, Saint Louis, MO, USA.,Department of Neurology, Washington University School of Medicine, Saint Louis, MO, USA
| | - Anne M Fagan
- Knight Alzheimer Disease Research Center, Washington University School of Medicine, Saint Louis, MO, USA.,Department of Neurology, Washington University School of Medicine, Saint Louis, MO, USA
| | - Tammie LS Benzinger
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, Saint Louis, MO, USA.,Knight Alzheimer Disease Research Center, Washington University School of Medicine, Saint Louis, MO, USA
| | - Brian A Gordon
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, Saint Louis, MO, USA.,Knight Alzheimer Disease Research Center, Washington University School of Medicine, Saint Louis, MO, USA.,Hope Center for Neurological Disorders, Washington University School of Medicine, Saint Louis, MO, USA.,Department of Psychological & Brain Sciences, Washington University, Saint Louis, MO, USA
| |
Collapse
|
19
|
Harerimana NV, Goate AM, Bowles KR. The influence of 17q21.31 and APOE genetic ancestry on neurodegenerative disease risk. Front Aging Neurosci 2022; 14:1021918. [PMID: 36337698 PMCID: PMC9632173 DOI: 10.3389/fnagi.2022.1021918] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2022] [Accepted: 09/26/2022] [Indexed: 09/08/2024] Open
Abstract
Advances in genomic research over the last two decades have greatly enhanced our knowledge concerning the genetic landscape and pathophysiological processes involved in multiple neurodegenerative diseases. However, current insights arise almost exclusively from studies on individuals of European ancestry. Despite this, studies have revealed that genetic variation differentially impacts risk for, and clinical presentation of neurodegenerative disease in non-European populations, conveying the importance of ancestry in predicting disease risk and understanding the biological mechanisms contributing to neurodegeneration. We review the genetic influence of two important disease-associated loci, 17q21.31 (the "MAPT locus") and APOE, to neurodegenerative disease risk in non-European populations, touching on global population differences and evolutionary genetics by ancestry that may underlie some of these differences. We conclude there is a need to increase representation of non-European ancestry individuals in genome-wide association studies (GWAS) and biomarker analyses in order to help resolve existing disparities in understanding risk for, diagnosis of, and treatment for neurodegenerative diseases in diverse populations.
Collapse
Affiliation(s)
- Nadia V. Harerimana
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, United States
- Ronald M. Loeb Center for Alzheimer’s Disease, Icahn School of Medicine at Mount Sinai, New York, NY, United States
- Icahn Genomics Institute, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Alison M. Goate
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, United States
- Ronald M. Loeb Center for Alzheimer’s Disease, Icahn School of Medicine at Mount Sinai, New York, NY, United States
- Icahn Genomics Institute, Icahn School of Medicine at Mount Sinai, New York, NY, United States
- Estelle and Daniel Maggin Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Kathryn R. Bowles
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, United States
- Ronald M. Loeb Center for Alzheimer’s Disease, Icahn School of Medicine at Mount Sinai, New York, NY, United States
- Icahn Genomics Institute, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| |
Collapse
|
20
|
Li L, Yu X, Sheng C, Jiang X, Zhang Q, Han Y, Jiang J. A review of brain imaging biomarker genomics in Alzheimer’s disease: implementation and perspectives. Transl Neurodegener 2022; 11:42. [PMID: 36109823 PMCID: PMC9476275 DOI: 10.1186/s40035-022-00315-z] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2022] [Accepted: 08/24/2022] [Indexed: 11/25/2022] Open
Abstract
Alzheimer’s disease (AD) is a progressive neurodegenerative disease with phenotypic changes closely associated with both genetic variants and imaging pathology. Brain imaging biomarker genomics has been developed in recent years to reveal potential AD pathological mechanisms and provide early diagnoses. This technique integrates multimodal imaging phenotypes with genetic data in a noninvasive and high-throughput manner. In this review, we summarize the basic analytical framework of brain imaging biomarker genomics and elucidate two main implementation scenarios of this technique in AD studies: (1) exploring novel biomarkers and seeking mutual interpretability and (2) providing a diagnosis and prognosis for AD with combined use of machine learning methods and brain imaging biomarker genomics. Importantly, we highlight the necessity of brain imaging biomarker genomics, discuss the strengths and limitations of current methods, and propose directions for development of this research field.
Collapse
|
21
|
Yang A, Du L, Gao W, Liu B, Chen Y, Wang Y, Liu X, Lv K, Zhang W, Xia H, Wu K, Ma G. Associations of cortical iron accumulation with cognition and cerebral atrophy in Alzheimer's disease. Quant Imaging Med Surg 2022; 12:4570-4586. [PMID: 36060596 PMCID: PMC9403583 DOI: 10.21037/qims-22-7] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Accepted: 06/10/2022] [Indexed: 11/06/2022]
Abstract
BACKGROUND In Alzheimer's disease (AD), cerebral iron accumulation colocalizes with the pathological proteins amyloid-β (Aβ) and tau. Furthermore, tau-induced cortical thinning is associated with cognitive decline. In this study, quantitative susceptibility mapping (QSM) was used to investigate the whole-brain distribution pattern of cortical iron deposition and its relationships with cognition and cortical thickness in AD. METHODS This cross-sectional study prospectively recruited 30 participants with AD and 26 age- and sex-matched healthy controls (HCs). All participants underwent QSM and T1-weighted examinations on a 3.0T MRI scanner. Global cognition was assessed using the Mini-Mental State Examination (MMSE) and Montreal Cognitive Assessment (MoCA). Whole-brain cross-sectional QSM analysis and whole-brain QSM regression analyses against the MMSE and MoCA scores were performed. Surface-based morphometry analysis was also performed. Subsequently, in regions with significant atrophy, magnetic susceptibility was compared between the AD and HC groups, and the association between magnetic susceptibility and cortical thickness was assessed. RESULTS Whole-brain QSM cross-sectional analysis in the AD group demonstrated widespread increased susceptibility across the cortical ribbon, asymmetrically covering the left hemisphere cerebral cortex, caudate nucleus, putamen, and partial cerebellar cortex. Whole-brain QSM regression analyses in the AD group showed that increased susceptibility covaried with lower MMSE and MoCA scores, and was predominantly located in the right parietal cortex and lateral occipital cortex. In the AD group, cortical thickness was reduced in the left superior temporal gyrus, right frontal pole, fusiform gyus, and pars opercularis, and there were increases in susceptibility in the right frontal pole (AD: mean ± SD 0.034±0.007 ppm, 95% CI: 0.032-0.037 ppm; HC: 0.030±0.005 ppm, 95% CI: 0.028-0.032 ppm; P=0.016) and pars opercularis (AD: 0.020±0.003 ppm, 95% CI: 0.018-0.021 ppm; HC: 0.017±0.002 ppm, 95% CI: 0.017-0.018 ppm; P=0.002). Susceptibility was negatively correlated with cortical thickness in the right pars opercularis in the entire cohort (r=-0.521, P<0.001) and AD group (r=-0.510, P=0.005). CONCLUSIONS Widespread cortical iron, as measured by QSM, accumulated in AD and iron deposition was associated with poor cognitive performance. Increased iron content was also associated with brain atrophy. Our study suggests QSM may be a useful imaging biomarker for monitoring the neurodegenerative progression of AD.
Collapse
Affiliation(s)
- Aocai Yang
- Department of Radiology, China-Japan Friendship Hospital, Beijing, China
- Graduate School of Peking Union Medical College, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Lei Du
- Department of Radiology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital & Institute, Beijing, China
| | - Wenwen Gao
- Department of Radiology, China-Japan Friendship Hospital, Beijing, China
- Peking University China-Japan Friendship School of Clinical Medicine, Beijing, China
| | - Bing Liu
- Department of Radiology, China-Japan Friendship Hospital, Beijing, China
- Graduate School of Peking Union Medical College, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yue Chen
- Department of Radiology, China-Japan Friendship Hospital, Beijing, China
- Peking University China-Japan Friendship School of Clinical Medicine, Beijing, China
| | - Yige Wang
- Department of Radiology, China-Japan Friendship Hospital, Beijing, China
- Graduate School of Peking Union Medical College, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xiuxiu Liu
- Department of Radiology, China-Japan Friendship Hospital, Beijing, China
- Peking University China-Japan Friendship School of Clinical Medicine, Beijing, China
| | - Kuan Lv
- Department of Radiology, China-Japan Friendship Hospital, Beijing, China
- Peking University China-Japan Friendship School of Clinical Medicine, Beijing, China
| | - Wenwei Zhang
- Institute of Electrical Engineering, Chinese Academy of Sciences, Beijing, China
| | - Hui Xia
- Institute of Electrical Engineering, Chinese Academy of Sciences, Beijing, China
| | - Kai Wu
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou, China
| | - Guolin Ma
- Department of Radiology, China-Japan Friendship Hospital, Beijing, China
- Graduate School of Peking Union Medical College, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
22
|
Saroja SR, Gorbachev K, TCW J, Goate AM, Pereira AC. Astrocyte-secreted glypican-4 drives APOE4-dependent tau hyperphosphorylation. Proc Natl Acad Sci U S A 2022; 119:e2108870119. [PMID: 35969759 PMCID: PMC9407658 DOI: 10.1073/pnas.2108870119] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2021] [Accepted: 06/07/2022] [Indexed: 01/03/2023] Open
Abstract
Tau protein aggregates are a major driver of neurodegeneration and behavioral impairments in tauopathies, including in Alzheimer's disease (AD). Apolipoprotein E4 (APOE4), the highest genetic risk factor for late-onset AD, has been shown to exacerbate tau hyperphosphorylation in mouse models. However, the exact mechanisms through which APOE4 induces tau hyperphosphorylation remains unknown. Here, we report that the astrocyte-secreted protein glypican-4 (GPC-4), which we identify as a binding partner of APOE4, drives tau hyperphosphorylation. We discovered that first, GPC-4 preferentially interacts with APOE4 in comparison to APOE2, considered to be a protective allele to AD, and second, that postmortem APOE4-carrying AD brains highly express GPC-4 in neurotoxic astrocytes. Furthermore, the astrocyte-secreted GPC-4 induced both tau accumulation and propagation in vitro. CRISPR/dCas9-mediated activation of GPC-4 in a tauopathy mouse model robustly induced tau hyperphosphorylation. In the absence of GPC4, APOE4-induced tau hyperphosphorylation was largely diminished using in vitro tau fluorescence resonance energy transfer-biosensor cells, in human-induced pluripotent stem cell-derived astrocytes and in an in vivo mouse model. We further show that APOE4-mediated surface trafficking of APOE receptor low-density lipoprotein receptor-related protein 1 through GPC-4 can be a gateway to tau spreading. Collectively, these data support that APOE4-induced tau hyperphosphorylation is directly mediated by GPC-4.
Collapse
Affiliation(s)
- Sivaprakasam R. Saroja
- Department of Neurology, Icahn School of Medicine, Mount Sinai, New York, NY 10029
- Nash Family Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029
| | - Kirill Gorbachev
- Department of Neurology, Icahn School of Medicine, Mount Sinai, New York, NY 10029
- Nash Family Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029
| | - Julia TCW
- Nash Family Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029
- Ronald M. Loeb Center for Alzheimer's Disease, Icahn School of Medicine at Mount Sinai, New York, NY 10029
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029
| | - Alison M. Goate
- Nash Family Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029
- Ronald M. Loeb Center for Alzheimer's Disease, Icahn School of Medicine at Mount Sinai, New York, NY 10029
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029
| | - Ana C. Pereira
- Department of Neurology, Icahn School of Medicine, Mount Sinai, New York, NY 10029
- Nash Family Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029
- Ronald M. Loeb Center for Alzheimer's Disease, Icahn School of Medicine at Mount Sinai, New York, NY 10029
| |
Collapse
|
23
|
Frontzkowski L, Ewers M, Brendel M, Biel D, Ossenkoppele R, Hager P, Steward A, Dewenter A, Römer S, Rubinski A, Buerger K, Janowitz D, Binette AP, Smith R, Strandberg O, Carlgren NM, Dichgans M, Hansson O, Franzmeier N. Earlier Alzheimer’s disease onset is associated with tau pathology in brain hub regions and facilitated tau spreading. Nat Commun 2022; 13:4899. [PMID: 35987901 PMCID: PMC9392750 DOI: 10.1038/s41467-022-32592-7] [Citation(s) in RCA: 42] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2021] [Accepted: 08/08/2022] [Indexed: 12/20/2022] Open
Abstract
AbstractIn Alzheimer’s disease (AD), younger symptom onset is associated with accelerated disease progression and tau spreading, yet the mechanisms underlying faster disease manifestation are unknown. To address this, we combined resting-state fMRI and longitudinal tau-PET in two independent samples of controls and biomarker-confirmed AD patients (ADNI/BioFINDER, n = 240/57). Consistent across both samples, we found that younger symptomatic AD patients showed stronger tau-PET in globally connected fronto-parietal hubs, i.e., regions that are critical for maintaining cognition in AD. Stronger tau-PET in hubs predicted faster subsequent tau accumulation, suggesting that tau in globally connected regions facilitates connectivity-mediated tau spreading. Further, stronger tau-PET in hubs mediated the association between younger age and faster tau accumulation in symptomatic AD patients, which predicted faster cognitive decline. These independently validated findings suggest that younger AD symptom onset is associated with stronger tau pathology in brain hubs, and accelerated tau spreading throughout connected brain regions and cognitive decline.
Collapse
|
24
|
Stage E, Risacher SL, Lane KA, Gao S, Nho K, Saykin AJ, Apostolova LG, for the Alzheimer's Disease Neuroimaging Initiative. Association of the top 20 Alzheimer's disease risk genes with [ 18F]flortaucipir PET. ALZHEIMER'S & DEMENTIA (AMSTERDAM, NETHERLANDS) 2022; 14:e12308. [PMID: 35592828 PMCID: PMC9092485 DOI: 10.1002/dad2.12308] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Revised: 03/03/2022] [Accepted: 03/07/2022] [Indexed: 04/12/2023]
Abstract
Introduction We previously reported genetic associations of the top Alzheimer's disease (AD) risk alleles with amyloid deposition and neurodegeneration. Here, we report the association of these variants with [18F]flortaucipir standardized uptake value ratio (SUVR). Methods We analyzed the [18F]flortaucipir scans of 352 cognitively normal (CN), 160 mild cognitive impairment (MCI), and 54 dementia (DEM) participants from Alzheimer's Disease Neuroimaging Initiative (ADNI)2 and 3. We ran step-wise regression with log-transformed [18F]flortaucipir meta-region of interest SUVR as the outcome measure and genetic variants, age, sex, and apolipoprotein E (APOE) ε4 as predictors. The results were visualized using parametric mapping at familywise error cluster-level-corrected P < .05. Results APOE ε4 showed significant (P < .05) associations with tau deposition across all disease stages. Other significantly associated genes include variants in ABCA7 in CN, CR1 in MCI, BIN1 and CASS4 in MCI and dementia participants. Discussion We found significant associations to tau deposition for ABCA7, BIN1, CASS4, and CR1, in addition to APOE ε4. These four variants have been previously associated with tau metabolism through model systems.
Collapse
Affiliation(s)
- Eddie Stage
- Department of NeurologyIndiana University School of MedicineIndianapolisIndianaUSA
| | - Shannon L. Risacher
- Department of Radiology and Imaging SciencesIndiana University School of MedicineIndianapolisIndianaUSA
| | - Kathleen A. Lane
- Department of BiostatisticsIndiana University School of MedicineIndianapolisIndianaUSA
| | - Sujuan Gao
- Department of BiostatisticsIndiana University School of MedicineIndianapolisIndianaUSA
| | - Kwangsik Nho
- Department of Radiology and Imaging SciencesIndiana University School of MedicineIndianapolisIndianaUSA
| | - Andrew J. Saykin
- Department of NeurologyIndiana University School of MedicineIndianapolisIndianaUSA
- Department of Radiology and Imaging SciencesIndiana University School of MedicineIndianapolisIndianaUSA
| | - Liana G. Apostolova
- Department of NeurologyIndiana University School of MedicineIndianapolisIndianaUSA
- Department of Radiology and Imaging SciencesIndiana University School of MedicineIndianapolisIndianaUSA
- Department of Medical and Molecular GeneticsIndiana University School of MedicineIndianapolisIndianaUSA
| | | |
Collapse
|
25
|
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
|
26
|
Frisoni GB, Altomare D, Thal DR, Ribaldi F, van der Kant R, Ossenkoppele R, Blennow K, Cummings J, van Duijn C, Nilsson PM, Dietrich PY, Scheltens P, Dubois B. The probabilistic model of Alzheimer disease: the amyloid hypothesis revised. Nat Rev Neurosci 2022; 23:53-66. [PMID: 34815562 PMCID: PMC8840505 DOI: 10.1038/s41583-021-00533-w] [Citation(s) in RCA: 251] [Impact Index Per Article: 83.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/04/2021] [Indexed: 01/03/2023]
Abstract
The current conceptualization of Alzheimer disease (AD) is driven by the amyloid hypothesis, in which a deterministic chain of events leads from amyloid deposition and then tau deposition to neurodegeneration and progressive cognitive impairment. This model fits autosomal dominant AD but is less applicable to sporadic AD. Owing to emerging information regarding the complex biology of AD and the challenges of developing amyloid-targeting drugs, the amyloid hypothesis needs to be reconsidered. Here we propose a probabilistic model of AD in which three variants of AD (autosomal dominant AD, APOE ε4-related sporadic AD and APOE ε4-unrelated sporadic AD) feature decreasing penetrance and decreasing weight of the amyloid pathophysiological cascade, and increasing weight of stochastic factors (environmental exposures and lower-risk genes). Together, these variants account for a large share of the neuropathological and clinical variability observed in people with AD. The implementation of this model in research might lead to a better understanding of disease pathophysiology, a revision of the current clinical taxonomy and accelerated development of strategies to prevent and treat AD.
Collapse
Affiliation(s)
- Giovanni B Frisoni
- Laboratory of Neuroimaging of Aging (LANVIE), University of Geneva, Geneva, Switzerland.
- Memory Clinic, Geneva University Hospitals, Geneva, Switzerland.
| | - Daniele Altomare
- Laboratory of Neuroimaging of Aging (LANVIE), University of Geneva, Geneva, Switzerland
- Memory Clinic, Geneva University Hospitals, Geneva, Switzerland
| | - Dietmar Rudolf Thal
- Laboratory for Neuropathology, Department of Imaging and Pathology, and Leuven Brain Institute, University of Leuven, Leuven, Belgium
- Department of Pathology, University Hospital Leuven, Leuven, Belgium
| | - Federica Ribaldi
- Laboratory of Neuroimaging of Aging (LANVIE), University of Geneva, Geneva, Switzerland
- Memory Clinic, Geneva University Hospitals, Geneva, Switzerland
- Laboratory of Alzheimer's Neuroimaging and Epidemiology (LANE), IRCCS Centro S. Giovanni di Dio Fatebenefratelli, Brescia, Italy
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Rik van der Kant
- Alzheimer Center Amsterdam, Department of Neurology, Amsterdam Neuroscience, Amsterdam UMC, Amsterdam, Netherlands
- Center for Neurogenomics and Cognitive Research, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam, Netherlands
| | - Rik Ossenkoppele
- Alzheimer Center Amsterdam, Department of Neurology, Amsterdam Neuroscience, Amsterdam UMC, Amsterdam, Netherlands
- Clinical Memory Research Unit, Lund University, Lund, Sweden
| | - Kaj Blennow
- Cinical Neurochemistry Laboratory, Institute of Neuroscience and Physiology, University of Gothenburg, Sahlgrenska University Hospital, Mölndal, Sweden
| | - Jeffrey Cummings
- Chambers-Grundy Center for Transformative Neuroscience, Department of Brain Health, School of Integrated Health Sciences; University of Nevada, Las Vegas, Las Vegas, NV, USA
| | - Cornelia van Duijn
- Department of Epidemiology, Erasmus University Medical Center, Rotterdam, Netherlands
- Clinical Trial Service Unit and Epidemiological Studies Unit, Nuffield Department of Population Health, University of Oxford, Oxford, UK
| | - Peter M Nilsson
- Department of Clinical Sciences, Lund University, Skåne University Hospital, Malmö, Sweden
| | | | - Philip Scheltens
- Alzheimer Center Amsterdam, Department of Neurology, Amsterdam Neuroscience, Amsterdam UMC, Amsterdam, Netherlands
- Life Science Partners, Amsterdam, Netherlands
| | - Bruno Dubois
- Institut de la Mémoire et de la Maladie d'Alzheimer, IM2A, Groupe Hospitalier Pitié-Salpêtrière, Sorbonne Université, Paris, France
- Institut du Cerveau et de la Moelle Épinière, UMR-S975, INSERM, Paris, France
| |
Collapse
|
27
|
Jellinger KA. Recent update on the heterogeneity of the Alzheimer’s disease spectrum. J Neural Transm (Vienna) 2021; 129:1-24. [DOI: 10.1007/s00702-021-02449-2] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2021] [Accepted: 11/25/2021] [Indexed: 02/03/2023]
|
28
|
Whitwell JL, Tosakulwong N, Weigand SD, Graff-Radford J, Ertekin-Taner N, Machulda MM, Duffy JR, Schwarz CG, Senjem ML, Jack CR, Lowe VJ, Josephs KA. Relationship of APOE, age at onset, amyloid and clinical phenotype in Alzheimer disease. Neurobiol Aging 2021; 108:90-98. [PMID: 34551374 DOI: 10.1016/j.neurobiolaging.2021.08.012] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Revised: 08/13/2021] [Accepted: 08/18/2021] [Indexed: 11/26/2022]
Abstract
The apolipoprotein E (APOE) ε4 allele is the most well-established risk factor for Alzheimer's disease (AD), although its relationship to age at onset and clinical phenotype is unclear. We aimed to assess relationships between APOE genotype and age at onset, amyloid-beta (Aβ) deposition and typical versus atypical clinical presentations in AD. Frequency of APOE ε4 carriers by age at onset was assessed in 447 AD patients, 138 atypical AD patients recruited by the Neurodegenerative Research Group at Mayo Clinic, and 309 with typical AD from ADNI. APOE ε4 frequency increased with age at onset in atypical AD but showed a bell-shaped curve in typical AD where highest frequencies were observed between 65 and 70 years. Typical AD showed higher APOE ε4 frequencies than atypical AD only between the ages of 57 and 69 years. Global Aβ standard uptake value ratios did not differ according to APOE e4 status in either group. APOE genotype varies by both age at onset and clinical phenotype in AD, highlighting the heterogeneous nature of AD.
Collapse
Affiliation(s)
| | | | - Stephen D Weigand
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN, USA
| | | | | | - Mary M Machulda
- Department of Psychology and Psychiatry, Mayo Clinic, Rochester, MN, USA
| | - Joseph R Duffy
- Department of Neurology, Mayo Clinic, Rochester, MN, USA
| | | | - Matthew L Senjem
- Department of Radiology, Mayo Clinic, Rochester, MN, USA; Department of Information Technology, Mayo Clinic, Rochester, MN, USA
| | | | - Val J Lowe
- Department of Radiology, Mayo Clinic, Rochester, MN, USA
| | | | | |
Collapse
|
29
|
Koutsodendris N, Nelson MR, Rao A, Huang Y. Apolipoprotein E and Alzheimer's Disease: Findings, Hypotheses, and Potential Mechanisms. ANNUAL REVIEW OF PATHOLOGY-MECHANISMS OF DISEASE 2021; 17:73-99. [PMID: 34460318 DOI: 10.1146/annurev-pathmechdis-030421-112756] [Citation(s) in RCA: 123] [Impact Index Per Article: 30.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Alzheimer's disease (AD) is a multifactorial neurodegenerative disorder that involves dysregulation of many cellular and molecular processes. It is notoriously difficult to develop therapeutics for AD due to its complex nature. Nevertheless, recent advancements in imaging technology and the development of innovative experimental techniques have allowed researchers to perform in-depth analyses to uncover the pathogenic mechanisms of AD. An important consideration when studying late-onset AD is its major genetic risk factor, apolipoprotein E4 (apoE4). Although the exact mechanisms underlying apoE4 effects on AD initiation and progression are not fully understood, recent studies have revealed critical insights into the apoE4-induced deficits that occur in AD. In this review, we highlight notable studies that detail apoE4 effects on prominent AD pathologies, including amyloid-β, tau pathology, neuroinflammation, and neural network dysfunction. We also discuss evidence that defines the physiological functions of apoE and outlines how these functions are disrupted in apoE4-related AD. Expected final online publication date for the Annual Review of Pathology: Mechanisms of Disease, Volume 17 is January 2022. Please see http://www.annualreviews.org/page/journal/pubdates for revised estimates.
Collapse
Affiliation(s)
- Nicole Koutsodendris
- Developmental and Stem Cell Biology Graduate Program, University of California, San Francisco, California 94131, USA; , .,Gladstone Institutes of Neurological Disease, San Francisco, California 94158, USA
| | - Maxine R Nelson
- Gladstone Institutes of Neurological Disease, San Francisco, California 94158, USA.,Biomedical Sciences Graduate Program, University of California, San Francisco, California 94143, USA
| | - Antara Rao
- Developmental and Stem Cell Biology Graduate Program, University of California, San Francisco, California 94131, USA; , .,Gladstone Institutes of Neurological Disease, San Francisco, California 94158, USA
| | - Yadong Huang
- Developmental and Stem Cell Biology Graduate Program, University of California, San Francisco, California 94131, USA; , .,Gladstone Institutes of Neurological Disease, San Francisco, California 94158, USA.,Biomedical Sciences Graduate Program, University of California, San Francisco, California 94143, USA.,Department of Neurology, University of California, San Francisco, California 94158, USA
| |
Collapse
|
30
|
Iannopollo E, Garcia K, Alzheimer' Disease Neuroimaging Initiative. Enhanced detection of cortical atrophy in Alzheimer's disease using structural MRI with anatomically constrained longitudinal registration. Hum Brain Mapp 2021; 42:3576-3592. [PMID: 33988265 PMCID: PMC8249882 DOI: 10.1002/hbm.25455] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2020] [Revised: 04/08/2021] [Accepted: 04/11/2021] [Indexed: 12/14/2022] Open
Abstract
Cortical atrophy is a defining feature of Alzheimer's disease (AD), often detectable before symptoms arise. In surface-based analyses, studies have commonly focused on cortical thinning while overlooking the impact of loss in surface area. To capture the impact of both cortical thinning and surface area loss, we used anatomically constrained Multimodal Surface Matching (aMSM), a recently developed tool for mapping change in surface area. We examined cortical atrophy over 2 years in cognitively normal subjects and subjects with diagnoses of stable mild cognitive impairment, mild cognitive impairment that converted to AD, and AD. Magnetic resonance imaging scans were segmented and registered to a common atlas using previously described techniques (FreeSurfer and ciftify), then longitudinally registered with aMSM. Changes in cortical thickness, surface area, and volume were mapped within each diagnostic group, and groups were compared statistically. Changes in thickness and surface area detected atrophy at similar levels of significance, though regions of atrophy somewhat differed. Furthermore, we found that surface area maps offered greater consistency across scanners (3.0 vs. 1.5 T). Comparisons to the FreeSurfer longitudinal pipeline and parcellation-based (region-of-interest) analysis suggest that aMSM may allow more robust detection of atrophy, particularly in earlier disease stages and using smaller sample sizes.
Collapse
Affiliation(s)
- Emily Iannopollo
- Department of Radiology and Imaging SciencesIndiana University School of MedicineEvansvilleIndianaUSA
| | - Kara Garcia
- Department of Radiology and Imaging SciencesIndiana University School of MedicineEvansvilleIndianaUSA
| | | |
Collapse
|
31
|
Wolters EE, Dodich A, Boccardi M, Corre J, Drzezga A, Hansson O, Nordberg A, Frisoni GB, Garibotto V, Ossenkoppele R. Clinical validity of increased cortical uptake of [ 18F]flortaucipir on PET as a biomarker for Alzheimer's disease in the context of a structured 5-phase biomarker development framework. Eur J Nucl Med Mol Imaging 2021; 48:2097-2109. [PMID: 33547556 PMCID: PMC8175307 DOI: 10.1007/s00259-020-05118-w] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2020] [Accepted: 11/15/2020] [Indexed: 12/14/2022]
Abstract
PURPOSE In 2017, the Geneva Alzheimer's disease (AD) Biomarker Roadmap initiative adapted the framework of the systematic validation of oncological diagnostic biomarkers to AD biomarkers, with the aim to accelerate their development and implementation in clinical practice. With this work, we assess the maturity of [18F]flortaucipir PET and define its research priorities. METHODS The level of maturity of [18F]flortaucipir was assessed based on the AD Biomarker Roadmap. The framework assesses analytical validity (phases 1-2), clinical validity (phases 3-4), and clinical utility (phase 5). RESULTS The main aims of phases 1 (rationale for use) and 2 (discriminative ability) have been achieved. [18F]Flortaucipir binds with high affinity to paired helical filaments of tau and has favorable kinetic properties and excellent discriminative accuracy for AD. The majority of secondary aims of phase 2 were fully achieved. Multiple studies showed high correlations between ante-mortem [18F]flortaucipir PET and post-mortem tau (as assessed by histopathology), and also the effects of covariates on tracer binding are well studied. The aims of phase 3 (early detection ability) were only partially or preliminarily achieved, and the aims of phases 4 and 5 were not achieved. CONCLUSION Current literature provides partial evidence for clinical utility of [18F]flortaucipir PET. The aims for phases 1 and 2 were mostly achieved. Phase 3 studies are currently ongoing. Future studies including representative MCI populations and a focus on healthcare outcomes are required to establish full maturity of phases 4 and 5.
Collapse
Affiliation(s)
- E E Wolters
- Department of Radiology & Nuclear Medicine, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam UMC, location VUmc, PO Box 7057, 1007 MB, Amsterdam, The Netherlands.
- Alzheimer Center Amsterdam, Department of Neurology, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam, The Netherlands.
| | - A Dodich
- NIMTlab - Neuroimaging and Innovative Molecular Tracers Laboratory, University of Geneva, Geneva, Switzerland
- Centre for Mind/Brain Sciences-CIMeC, University of Trento, Rovereto, Italy
| | - M Boccardi
- Late Translational Dementia Studies Group, German Center for Neurodegenerative Diseases (DZNE), Rostock-Greifswald site, Rostock, Germany
| | - J Corre
- NIMTlab - Neuroimaging and Innovative Molecular Tracers Laboratory, University of Geneva, Geneva, Switzerland
- CURIC, Centre Universitaire Romand d'Implants Cochléaires, Department of Clinical Neurosciences, University of Geneva, Geneva, Switzerland
| | - A Drzezga
- Faculty of Medicine, University of Cologne, Cologne, Germany
- Institute of Neuroscience and Medicine (INM-2), Molecular Organization of the Brain, Research Center Jülich, Jülich, Germany
- German Center for Neurodegenerative Diseases (DZNE), Bonn-Cologne, Germany
| | - O Hansson
- Memory Clinic, Skåne University Hospital, Malmö, Sweden
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, Lund, Sweden
| | - A Nordberg
- Division of Clinical Geriatrics, Center for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden
| | - G B Frisoni
- LANVIE - Laboratory of Neuroimaging of Aging, University of Geneva, Geneva, Switzerland
- Memory Clinic, University Hospital, Geneva, Switzerland
| | - V Garibotto
- NIMTlab - Neuroimaging and Innovative Molecular Tracers Laboratory, University of Geneva, Geneva, Switzerland
| | - R Ossenkoppele
- Alzheimer Center Amsterdam, Department of Neurology, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam, The Netherlands
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, Lund, Sweden
| |
Collapse
|
32
|
Piersson AD, Mohamad M, Suppiah S, Rajab NF. Topographical patterns of whole-brain structural alterations in association with genetic risk, cerebrospinal fluid, positron emission tomography biomarkers of Alzheimer’s disease, and neuropsychological measures. Clin Transl Imaging 2021. [DOI: 10.1007/s40336-021-00440-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
33
|
Ossenkoppele R, Leuzy A, Cho H, Sudre CH, Strandberg O, Smith R, Palmqvist S, Mattsson-Carlgren N, Olsson T, Jögi J, Stormrud E, Ryu YH, Choi JY, Boxer AL, Gorno-Tempini ML, Miller BL, Soleimani-Meigooni D, Iaccarino L, La Joie R, Borroni E, Klein G, Pontecorvo MJ, Devous MD, Villeneuve S, Lyoo CH, Rabinovici GD, Hansson O. The impact of demographic, clinical, genetic, and imaging variables on tau PET status. Eur J Nucl Med Mol Imaging 2021; 48:2245-2258. [PMID: 33215319 PMCID: PMC8131404 DOI: 10.1007/s00259-020-05099-w] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2020] [Accepted: 10/27/2020] [Indexed: 12/14/2022]
Abstract
PURPOSE A substantial proportion of amyloid-β (Aβ)+ patients with clinically diagnosed Alzheimer's disease (AD) dementia and mild cognitive impairment (MCI) are tau PET-negative, while some clinically diagnosed non-AD neurodegenerative disorder (non-AD) patients or cognitively unimpaired (CU) subjects are tau PET-positive. We investigated which demographic, clinical, genetic, and imaging variables contributed to tau PET status. METHODS We included 2338 participants (430 Aβ+ AD dementia, 381 Aβ+ MCI, 370 non-AD, and 1157 CU) who underwent [18F]flortaucipir (n = 1944) or [18F]RO948 (n = 719) PET. Tau PET positivity was determined in the entorhinal cortex, temporal meta-ROI, and Braak V-VI regions using previously established cutoffs. We performed bivariate binary logistic regression models with tau PET status (positive/negative) as dependent variable and age, sex, APOEε4, Aβ status (only in CU and non-AD analyses), MMSE, global white matter hyperintensities (WMH), and AD-signature cortical thickness as predictors. Additionally, we performed multivariable binary logistic regression models to account for all other predictors in the same model. RESULTS Tau PET positivity in the temporal meta-ROI was 88.6% for AD dementia, 46.5% for MCI, 9.5% for non-AD, and 6.1% for CU. Among Aβ+ participants with AD dementia and MCI, lower age, MMSE score, and AD-signature cortical thickness showed the strongest associations with tau PET positivity. In non-AD and CU participants, presence of Aβ was the strongest predictor of a positive tau PET scan. CONCLUSION We identified several demographic, clinical, and neurobiological factors that are important to explain the variance in tau PET retention observed across the AD pathological continuum, non-AD neurodegenerative disorders, and cognitively unimpaired persons.
Collapse
Affiliation(s)
- Rik Ossenkoppele
- Clinical Memory Research Unit, Lund University, Lund, Sweden.
- Alzheimer Center Amsterdam, Department of Neurology, Amsterdam Neuroscience, Amsterdam UMC, VU University Medical Center, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands.
| | - Antoine Leuzy
- Clinical Memory Research Unit, Lund University, Lund, Sweden
| | - Hanna Cho
- Department of Neurology, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, South Korea
| | - Carole H Sudre
- School of Biomedical Engineering and Imaging Sciences, King's College London, London, UK
- Dementia Research Centre, Department of Neurodegenerative Disease, UCL Institute of Neurology, London, UK
- Centre for Medical Image Computing, Department of Medical Physics, University College London, London, UK
| | - Olof Strandberg
- Clinical Memory Research Unit, Lund University, Lund, Sweden
| | - Ruben Smith
- Clinical Memory Research Unit, Lund University, Lund, Sweden
| | | | - Niklas Mattsson-Carlgren
- Clinical Memory Research Unit, Lund University, Lund, Sweden
- Department of Neurology, Skåne University Hospital, Lund, Sweden
- Wallenberg Centre for Molecular Medicine, Lund University, Lund, Sweden
| | - Tomas Olsson
- Department of Radiation Physics, Skåne University Hospital, Lund, Sweden
| | - Jonas Jögi
- Department of Clinical Physiology and Nuclear Medicine, Skåne University Hospital, Lund, Sweden
| | - Erik Stormrud
- Clinical Memory Research Unit, Lund University, Lund, Sweden
- Memory Clinic, Skåne University Hospital, Malmö, Sweden
| | - Young Hoon Ryu
- Department of Nuclear Medicine, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, South Korea
| | - Jae Yong Choi
- Department of Nuclear Medicine, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, South Korea
- Division of applied RI, Korea Institute Radiological and Medical Sciences, Seoul, South Korea
| | - Adam L Boxer
- Department of Neurology, Memory and Aging Center, University of California San Francisco, San Francisco, USA
| | - Maria L Gorno-Tempini
- Department of Neurology, Memory and Aging Center, University of California San Francisco, San Francisco, USA
| | - Bruce L Miller
- Department of Neurology, Memory and Aging Center, University of California San Francisco, San Francisco, USA
| | - David Soleimani-Meigooni
- Department of Neurology, Memory and Aging Center, University of California San Francisco, San Francisco, USA
| | - Leonardo Iaccarino
- Department of Neurology, Memory and Aging Center, University of California San Francisco, San Francisco, USA
| | - Renaud La Joie
- Department of Neurology, Memory and Aging Center, University of California San Francisco, San Francisco, USA
| | | | | | | | | | - Sylvia Villeneuve
- Departments of Psychiatry and Neurology & Neurosurgery, Douglas Mental Health University Institute, McGill University, Montreal, Quebec, Canada
| | - Chul Hyoung Lyoo
- Department of Neurology, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, South Korea
| | - Gil D Rabinovici
- F. Hoffmann-La Roche Ltd, Basel, Switzerland
- Department of Radiology and Biomedical Imaging, University of California San Francisco, San Francisco, USA
- Molecular Biophysics and Integrated Bioimaging Division, Lawrence Berkeley National Laboratory, Berkeley, CA, USA
| | - Oskar Hansson
- Clinical Memory Research Unit, Lund University, Lund, Sweden.
- Memory Clinic, Skåne University Hospital, Malmö, Sweden.
| |
Collapse
|
34
|
Salvadó G, Grothe MJ, Groot C, Moscoso A, Schöll M, Gispert JD, Ossenkoppele R. Differential associations of APOE-ε2 and APOE-ε4 alleles with PET-measured amyloid-β and tau deposition in older individuals without dementia. Eur J Nucl Med Mol Imaging 2021; 48:2212-2224. [PMID: 33521872 PMCID: PMC8175302 DOI: 10.1007/s00259-021-05192-8] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2020] [Accepted: 01/03/2021] [Indexed: 01/07/2023]
Abstract
PURPOSE To examine associations between the APOE-ε2 and APOE-ε4 alleles and core Alzheimer's disease (AD) pathological hallmarks as measured by amyloid-β (Aβ) and tau PET in older individuals without dementia. METHODS We analyzed data from 462 ADNI participants without dementia who underwent Aβ ([18F]florbetapir or [18F]florbetaben) and tau ([18F]flortaucipir) PET, structural MRI, and cognitive testing. Employing APOE-ε3 homozygotes as the reference group, associations between APOE-ε2 and APOE-ε4 carriership with global Aβ PET and regional tau PET measures (entorhinal cortex (ERC), inferior temporal cortex, and Braak-V/VI neocortical composite regions) were investigated using linear regression models. In a subset of 156 participants, we also investigated associations between APOE genotype and regional tau accumulation over time using linear mixed models. Finally, we assessed whether Aβ mediated the cross-sectional and longitudinal associations between APOE genotype and tau. RESULTS Compared to APOE-ε3 homozygotes, APOE-ε2 carriers had lower global Aβ burden (βstd [95% confidence interval (CI)]: - 0.31 [- 0.45, - 0.16], p = 0.034) but did not differ on regional tau burden or tau accumulation over time. APOE-ε4 participants showed higher Aβ (βstd [95%CI]: 0.64 [0.42, 0.82], p < 0.001) and tau burden (βstd range: 0.27-0.51, all p < 0.006). In mediation analyses, APOE-ε4 only retained an Aβ-independent effect on tau in the ERC. APOE-ε4 showed a trend towards increased tau accumulation over time in Braak-V/VI compared to APOE-ε3 homozygotes (βstd [95%CI]: 0.10 [- 0.02, 0.18], p = 0.11), and this association was fully mediated by baseline Aβ. CONCLUSION Our data suggest that the established protective effect of the APOE-ε2 allele against developing clinical AD is primarily linked to resistance against Aβ deposition rather than tau pathology.
Collapse
Affiliation(s)
- Gemma Salvadó
- Alzheimer Prevention Program, Barcelonaβeta Brain Research Center (BBRC), Pasqual Maragall Foundation, C/ Wellington, 30 08005, Barcelona, Spain.
- IMIM (Hospital del Mar Medical Research Institute), Barcelona, Spain.
- Alzheimer Center Amsterdam, Department of Neurology, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam, The Netherlands.
| | - Michel J Grothe
- Wallenberg Centre for Molecular and Translational Medicine, Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, University of Gothenburg, Gothenburg, Sweden.
- Unidad de Trastornos del Movimiento, Servicio de Neurología y Neurofisiología Clínica, Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Avda. Manuel Siurot, s/n 41013, Seville, Spain.
| | - Colin Groot
- Alzheimer Center Amsterdam, Department of Neurology, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam, The Netherlands
| | - Alexis Moscoso
- Wallenberg Centre for Molecular and Translational Medicine, Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, University of Gothenburg, Gothenburg, Sweden
| | - Michael Schöll
- Wallenberg Centre for Molecular and Translational Medicine, Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, University of Gothenburg, Gothenburg, Sweden
- Dementia Research Centre, Institute of Neurology, University College London, London, UK
| | - Juan Domingo Gispert
- Alzheimer Prevention Program, Barcelonaβeta Brain Research Center (BBRC), Pasqual Maragall Foundation, C/ Wellington, 30 08005, Barcelona, Spain
- IMIM (Hospital del Mar Medical Research Institute), Barcelona, Spain
- Universitat Pompeu Fabra, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Madrid, Spain
| | - Rik Ossenkoppele
- Alzheimer Center Amsterdam, Department of Neurology, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam, The Netherlands
- Clinical Memory Research Unit, Lund University, Lund, Sweden
| |
Collapse
|
35
|
Jellinger KA. Pathobiological Subtypes of Alzheimer Disease. Dement Geriatr Cogn Disord 2021; 49:321-333. [PMID: 33429401 DOI: 10.1159/000508625] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Accepted: 05/11/2020] [Indexed: 11/19/2022] Open
Abstract
Alzheimer disease (AD), the most common form of dementia, is a heterogenous disorder with various pathobiological subtypes. In addition to the 4 major subtypes based on the distribution of tau pathology and brain atrophy (typical, limbic predominant, hippocampal sparing, and minimal atrophy [MA]), several other clinical variants showing distinct regional patterns of tau burden have been identified: nonamnestic, corticobasal syndromal, primary progressive aphasia, posterior cortical atrophy, behavioral/dysexecutive, and mild dementia variants. Among the subtypes, differences were found in age at onset, sex distribution, cognitive status, disease duration, APOE genotype, and biomarker levels. The patterns of key network destructions parallel the tau and atrophy patterns of the AD subgroups essentially. Interruption of key networks, in particular the default-mode network that is responsible for cognitive decline, is consistent in hetero-genous AD groups. AD pathology is often associated with co-pathologies: cerebrovascular lesions, Lewy pathology, and TDP-43 proteinopathies. These mixed pathologies essentially influence the clinical picture of AD and may accel-erate disease progression. Unraveling the heterogeneity among the AD spectrum entities is important for opening a window to pathogenic mechanisms affecting the brain and enabling precision medicine approaches as a basis for developing preventive and ultimately successful disease-modifying therapies for AD.
Collapse
|
36
|
Ossenkoppele R, Hansson O. Towards clinical application of tau PET tracers for diagnosing dementia due to Alzheimer's disease. Alzheimers Dement 2021; 17:1998-2008. [PMID: 33984177 DOI: 10.1002/alz.12356] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Revised: 03/22/2021] [Accepted: 03/28/2021] [Indexed: 11/07/2022]
Abstract
The recent development of several tau positron emission tomography (PET) tracers represents a major milestone for the Alzheimer's disease (AD) field. These tau PET tracers bind tau neurofibrillary tangles, a key neuropathological characteristic of AD that is tightly linked to synaptic loss, brain atrophy, and cognitive decline. It is notable that these tau PET tracers show low uptake in most non-AD tauopathies and other neurodegenerative disorders, resulting in a diagnostic specificity that is superior to that of amyloid beta (Aβ) PET and biofluid markers, especially at an older age when incidental Aβ pathology is common. Furthermore, tau PET tracers diagnostically outperform widely used MRI markers. Given its excellent diagnostic performance due to the combination of high sensitivity and specificity for detecting tau pathology in AD dementia, we hypothesize that tau PET can become an important diagnostic tool in specialized clinics for the differential diagnosis of dementia syndromes where AD is among the major possible underlying diseases.
Collapse
Affiliation(s)
- Rik Ossenkoppele
- Lund University, Clinical Memory Research Unit, Lund, Sweden.,Department of Neurology, Amsterdam Neuroscience, Alzheimer Center Amsterdam, Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam, The Netherlands
| | - Oskar Hansson
- Lund University, Clinical Memory Research Unit, Lund, Sweden.,Memory Clinic, Skåne University Hospital, Malmö, Sweden
| |
Collapse
|
37
|
Yan S, Zheng C, Paranjpe MD, Li Y, Li W, Wang X, Benzinger TLS, Lu J, Zhou Y. Sex modifies APOE ε4 dose effect on brain tau deposition in cognitively impaired individuals. Brain 2021; 144:3201-3211. [PMID: 33876815 PMCID: PMC8634082 DOI: 10.1093/brain/awab160] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Revised: 03/16/2021] [Accepted: 04/08/2021] [Indexed: 11/23/2022] Open
Abstract
Recent studies in cognitively unimpaired elderly individuals suggest that the APOE ε4 allele exerts a dosage-dependent effect on brain tau deposition. The aim of this study was to investigate sex differences in APOE ε4 gene dosage effects on brain tau deposition in cognitively impaired individuals using quantitative 18F-flortaucipir PET. Preprocessed 18F-flortaucipir tau PET images, T1-weighted structural MRI, demographic information, global cortical amyloid-β burden measured by 18F-florbetapir PET, CSF total tau and phosphorylated tau measurements were obtained from the Alzheimer’s Disease Neuroimaging Initiative database. Two hundred and sixty-eight cognitively impaired individuals with 146 APOE ε4 non-carriers and 122 carriers (85 heterozygotes and 37 homozygotes) were included in the study. An iterative reblurred Van Cittert iteration partial volume correction method was applied to all downloaded PET images. Magnetic resonance images were used for PET spatial normalization. Twelve regional standardized uptake value ratios relative to the cerebellum were computed in standard space. APOE ε4 dosage × sex interaction effect on 18F-flortaucipir standardized uptake value ratios was assessed using generalized linear models and sex-stratified analysis. We observed a significant APOE ε4 dosage × sex interaction effect on tau deposition in the lateral temporal, posterior cingulate, medial temporal, inferior temporal, entorhinal cortex, amygdala, parahippocampal gyrus regions after adjusting for age and education level (P < 0.05). The medial temporal, entorhinal cortex, amygdala and parahippocampal gyrus regions retained a significant APOE ε4 dosage × sex interaction effect on tau deposition after adjusting for global cortical amyloid-β (P < 0.05). In sex-stratified analysis, there was no significant difference in tau deposition between female homozygotes and heterozygotes (P > 0.05). In contrast, male homozygotes standardized uptake value ratios were significantly greater than heterozygotes or non-carriers throughout all 12 regions of interest (P < 0.05). Female heterozygotes exhibited significantly increased tau deposition compared to male heterozygotes in the orbitofrontal, posterior cingulate, lateral temporal, inferior temporal, entorhinal cortex, amygdala and parahippocampal gyrus (P < 0.05). Results from voxel-wise analysis were similar to the ones obtained from regions of interest analysis. Our findings indicate that an APOE ε4 dosage effect on brain region-specific tau deposition exists in males, but not females. These results have important clinical implications towards developing sex and genotype-guided therapeutics in Alzheimer’s disease and uncovers a potential explanation underlying differential APOE ε4-associated Alzheimer’s risk in males and females.
Collapse
Affiliation(s)
- Shaozhen Yan
- Department of Radiology and Nuclear Medicine, Xuanwu Hospital, Capital Medical University, Beijing, China.,Mallinckrodt Institute of Radiology, Washington University in St. Louis School of Medicine, St. Louis, MO, USA
| | - Chaojie Zheng
- Mallinckrodt Institute of Radiology, Washington University in St. Louis School of Medicine, St. Louis, MO, USA.,Central Research Institute, United Imaging Healthcare Group Co., Ltd, Shanghai, China
| | - Manish D Paranjpe
- Harvard-MIT Program in Health Sciences and Technology, Harvard Medical School, Boston, MA, USA
| | - Yanxiao Li
- Central Research Institute, United Imaging Healthcare Group Co., Ltd, Shanghai, China.,School of Computer Science, the University of Sydney, NSW 2006, Australia
| | - Weihua Li
- Department of Radiology and Nuclear Medicine, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Xiuying Wang
- School of Computer Science, the University of Sydney, NSW 2006, Australia
| | - Tammie L S Benzinger
- Mallinckrodt Institute of Radiology, Washington University in St. Louis School of Medicine, St. Louis, MO, USA.,Department of Neurology, Washington University in St. Louis School of Medicine, Saint Louis, MO, USA
| | - Jie Lu
- Department of Radiology and Nuclear Medicine, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Yun Zhou
- Mallinckrodt Institute of Radiology, Washington University in St. Louis School of Medicine, St. Louis, MO, USA.,Central Research Institute, United Imaging Healthcare Group Co., Ltd, Shanghai, China
| | | |
Collapse
|
38
|
Buckley RF. Recent Advances in Imaging of Preclinical, Sporadic, and Autosomal Dominant Alzheimer's Disease. Neurotherapeutics 2021; 18:709-727. [PMID: 33782864 PMCID: PMC8423933 DOI: 10.1007/s13311-021-01026-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/15/2021] [Indexed: 12/25/2022] Open
Abstract
Observing Alzheimer's disease (AD) pathological changes in vivo with neuroimaging provides invaluable opportunities to understand and predict the course of disease. Neuroimaging AD biomarkers also allow for real-time tracking of disease-modifying treatment in clinical trials. With recent neuroimaging advances, along with the burgeoning availability of longitudinal neuroimaging data and big-data harmonization approaches, a more comprehensive evaluation of the disease has shed light on the topographical staging and temporal sequencing of the disease. Multimodal imaging approaches have also promoted the development of data-driven models of AD-associated pathological propagation of tau proteinopathies. Studies of autosomal dominant, early sporadic, and late sporadic courses of the disease have shed unique insights into the AD pathological cascade, particularly with regard to genetic vulnerabilities and the identification of potential drug targets. Further, neuroimaging markers of b-amyloid, tau, and neurodegeneration have provided a powerful tool for validation of novel fluid cerebrospinal and plasma markers. This review highlights some of the latest advances in the field of human neuroimaging in AD across these topics, particularly with respect to positron emission tomography and structural and functional magnetic resonance imaging.
Collapse
Affiliation(s)
- Rachel F Buckley
- Department of Neurology, Massachusetts General Hospital & Brigham and Women's, Harvard Medical School, Boston, MA, USA.
- Melbourne School of Psychological Sciences and Florey Institutes, University of Melbourne, Melbourne, VIC, Australia.
- Department of Neurology, Massachusetts General Hospital, 149 13th St, Charlestown, MA, 02129, USA.
| |
Collapse
|
39
|
Doust YV, King AE, Ziebell JM. Implications for microglial sex differences in tau-related neurodegenerative diseases. Neurobiol Aging 2021; 105:340-348. [PMID: 34174592 DOI: 10.1016/j.neurobiolaging.2021.03.010] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Revised: 03/18/2021] [Accepted: 03/19/2021] [Indexed: 11/26/2022]
Abstract
Tauopathies are a group of neurodegenerative diseases that involve pathological changes to the tau protein. Neuroinflammation is a commonly reported feature of tauopathies that has been demonstrated to exacerbate tau pathology and, hence, neurodegeneration. Microglia can mediate the inflammatory response in order to maintain brain homeostasis. In the aged brain, microglia are reported to undergo morphological and functional changes, adopting a pro-inflammatory profile and loss of homeostatic functions. Dystrophic and dysfunctional microglia are associated with tau pathology in the healthy and diseased brain which is proposed to contribute to disease development and progression. Microglia have also been recently demonstrated to possess sexually dimorphic roles in the developing, adult and aged brain. The sex differences in microglial functionality suggest that microglia may contribute to tauopathies which may differ between sexes. This review highlights the detrimental loop between age-related microglial changes and tau pathology with implications for microglial sexual dichotomy.
Collapse
Affiliation(s)
- Yasmine V Doust
- Wicking Dementia Research and Education Centre, College of Health and Medicine, University of Tasmania, Hobart, Tasmania, Australia
| | - Anna E King
- Wicking Dementia Research and Education Centre, College of Health and Medicine, University of Tasmania, Hobart, Tasmania, Australia
| | - Jenna M Ziebell
- Wicking Dementia Research and Education Centre, College of Health and Medicine, University of Tasmania, Hobart, Tasmania, Australia.
| |
Collapse
|
40
|
Smith R, Strandberg O, Mattsson-Carlgren N, Leuzy A, Palmqvist S, Pontecorvo MJ, Devous MD, Ossenkoppele R, Hansson O. The accumulation rate of tau aggregates is higher in females and younger amyloid-positive subjects. Brain 2021; 143:3805-3815. [PMID: 33439987 PMCID: PMC7805812 DOI: 10.1093/brain/awaa327] [Citation(s) in RCA: 73] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2020] [Revised: 05/21/2020] [Accepted: 08/06/2020] [Indexed: 02/06/2023] Open
Abstract
The development of tau-PET allows paired helical filament tau pathology to be visualized in vivo. Increased knowledge about conditions affecting the rate of tau accumulation could guide the development of therapies halting the progression of Alzheimer’s disease. However, the factors modifying the rate of tau accumulation over time in Alzheimer’s disease are still largely unknown. Large-scale longitudinal cohort studies, adjusting for baseline tau load, are needed to establish such risk factors. In the present longitudinal study, 419 participants from four cohorts in the USA (Avid 05e, n = 157; Expedition-3, n = 82; ADNI, n = 123) and Sweden (BioFINDER, n = 57) were scanned repeatedly with tau-PET. The study participants were cognitively unimpaired (n = 153), or patients with mild cognitive impairment (n = 139) or Alzheimer’s disease dementia (n = 127). Participants underwent two to four tau-PET (18F-flortaucipir) scans with a mean (± standard deviation) of 537 (±163) days between the first and last scan. The change in tau-PET signal was estimated in temporal meta- and neocortical regions of interest. Subject specific tau-PET slopes were predicted simultaneously by age, sex, amyloid status (determined by amyloid-β PET), APOE ε4 genotype, study cohort, diagnosis and baseline tau load. We found that accelerated increase in tau-PET signal was observed in amyloid-β-positive mild cognitive impairment (3.0 ± 5.3%) and Alzheimer’s disease dementia (2.9 ± 5.7%), respectively, when compared to either amyloid-β-negative cognitively unimpaired (0.4 ± 2.7%), amyloid-β-negative mild cognitive impairment (−0.4 ± 2.3%) or amyloid-β-positive cognitively unimpaired (1.2 ± 2.8%). Tau-PET uptake was accelerated in females (temporal region of interest: t = 2.86, P = 0.005; neocortical region of interest: t = 2.90, P = 0.004), younger individuals (temporal region of interest: t = −2.49, P = 0.013), and individuals with higher baseline tau-PET signal (temporal region of interest: t = 3.83, P < 0.001; neocortical region of interest: t = 5.01, P < 0.001). Tau-PET slopes decreased with age in amyloid-β-positive subjects, but were stable by age in amyloid-β-negative subjects (age × amyloid-β status interaction: t = −2.39, P = 0.018). There were no effects of study cohort or APOE ε4 positivity. In a similar analysis on longitudinal amyloid-β-PET (in ADNI subjects only, n = 639), we found significant associations between the rate of amyloid-β accumulation and APOE ε4 positivity, older age and baseline amyloid-β positivity, but no effect of sex. In conclusion, in this longitudinal PET study comprising four cohorts, we found that the tau accumulation rate is greater in females and younger amyloid-β-positive individuals, while amyloid-β accumulation is greater in APOE ε4 carriers and older individuals. These findings are important considerations for the design of clinical trials, and might improve our understanding of factors associated with faster tau aggregation and spread.
Collapse
Affiliation(s)
- Ruben Smith
- Clinical Memory Research Unit, Department of Clinical Sciences, Lund University, Malmö, Sweden.,Department of Neurology, Skåne University Hospital, Lund, Sweden
| | - Olof Strandberg
- Clinical Memory Research Unit, Department of Clinical Sciences, Lund University, Malmö, Sweden
| | - Niklas Mattsson-Carlgren
- Clinical Memory Research Unit, Department of Clinical Sciences, Lund University, Malmö, Sweden.,Department of Neurology, Skåne University Hospital, Lund, Sweden.,Wallenberg Centre for Molecular Medicine, Lund University, Lund, Sweden
| | - Antoine Leuzy
- Clinical Memory Research Unit, Department of Clinical Sciences, Lund University, Malmö, Sweden
| | - Sebastian Palmqvist
- Clinical Memory Research Unit, Department of Clinical Sciences, Lund University, Malmö, Sweden.,Memory Clinic, Skåne University Hospital, Lund, Sweden
| | | | | | - Rik Ossenkoppele
- Clinical Memory Research Unit, Department of Clinical Sciences, Lund University, Malmö, Sweden.,Amsterdam University Medical Center, Alzheimercenter, Neuroscience Campus Amsterdam, Amsterdam, The Netherlands
| | - Oskar Hansson
- Clinical Memory Research Unit, Department of Clinical Sciences, Lund University, Malmö, Sweden.,Memory Clinic, Skåne University Hospital, Lund, Sweden
| |
Collapse
|
41
|
Ossenkoppele R, Lyoo CH, Jester-Broms J, Sudre CH, Cho H, Ryu YH, Choi JY, Smith R, Strandberg O, Palmqvist S, Kramer J, Boxer AL, Gorno-Tempini ML, Miller BL, La Joie R, Rabinovici GD, Hansson O. Assessment of Demographic, Genetic, and Imaging Variables Associated With Brain Resilience and Cognitive Resilience to Pathological Tau in Patients With Alzheimer Disease. JAMA Neurol 2021; 77:632-642. [PMID: 32091549 PMCID: PMC7042808 DOI: 10.1001/jamaneurol.2019.5154] [Citation(s) in RCA: 86] [Impact Index Per Article: 21.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Question Which demographic, genetic, and neuroimaging factors are associated with cognitive and brain resilience to pathological tau in patients with Alzheimer disease? Findings In this multicenter, cross-sectional, longitudinal study of 260 cognitively impaired amyloid-β–positive participants, young age and female sex were associated with greater brain resilience, whereas higher educational level and cortical thickness were associated with greater cognitive resilience. Meaning Cognitive and brain resilience may be associated with differential mechanisms, which may help explain interindividual differences in how well patients tolerate pathological tau. Importance Better understanding is needed of the degree to which individuals tolerate Alzheimer disease (AD)–like pathological tau with respect to brain structure (brain resilience) and cognition (cognitive resilience). Objective To examine the demographic (age, sex, and educational level), genetic (APOE-ε4 status), and neuroimaging (white matter hyperintensities and cortical thickness) factors associated with interindividual differences in brain and cognitive resilience to tau positron emission tomography (PET) load and to changes in global cognition over time. Design, Setting, an Participants In this cross-sectional, longitudinal study, tau PET was performed from June 1, 2014, to November 30, 2017, and global cognition monitored for a mean [SD] interval of 2.0 [1.8] years at 3 dementia centers in South Korea, Sweden, and the United States. The study included amyloid-β–positive participants with mild cognitive impairment or AD dementia. Data analysis was performed from October 26, 2018, to December 11, 2019. Exposures Standard dementia screening, cognitive testing, brain magnetic resonance imaging, amyloid-β PET and cerebrospinal fluid analysis, and flortaucipir (tau) labeled with fluor-18 (18F) PET. Main Outcomes and Measures Separate linear regression models were performed between whole cortex [18F]flortaucipir uptake and cortical thickness, and standardized residuals were used to obtain a measure of brain resilience. The same procedure was performed for whole cortex [18F]flortaucipir uptake vs Mini-Mental State Examination (MMSE) as a measure of cognitive resilience. Bivariate and multivariable linear regression models were conducted with age, sex, educational level, APOE-ε4 status, white matter hyperintensity volumes, and cortical thickness as independent variables and brain and cognitive resilience measures as dependent variables. Linear mixed models were performed to examine whether changes in MMSE scores over time differed as a function of a combined brain and cognitive resilience variable. Results A total of 260 participants (145 [55.8%] female; mean [SD] age, 69.2 [9.5] years; mean [SD] MMSE score, 21.9 [5.5]) were included in the study. In multivariable models, women (standardized β = −0.15, P = .02) and young patients (standardized β = −0.20, P = .006) had greater brain resilience to pathological tau. Higher educational level (standardized β = 0.23, P < .001) and global cortical thickness (standardized β = 0.23, P < .001) were associated with greater cognitive resilience to pathological tau. Linear mixed models indicated a significant interaction of brain resilience × cognitive resilience × time on MMSE (β [SE] = −0.235 [0.111], P = .03), with steepest slopes for individuals with both low brain and cognitive resilience. Conclusions and Relevance Results of this study suggest that women and young patients with AD have relative preservation of brain structure when exposed to neocortical pathological tau. Interindividual differences in resilience to pathological tau may be important to disease progression because participants with both low brain and cognitive resilience had the most rapid cognitive decline over time.
Collapse
Affiliation(s)
- Rik Ossenkoppele
- Lund University, Clinical Memory Research Unit, Lund, Sweden.,Alzheimer Center Amsterdam, Department of Neurology, Amsterdam Neuroscience, Amsterdam University Medical Center, Amsterdam, the Netherlands
| | - Chul Hyoung Lyoo
- Gangnam Severance Hospital, Department of Neurology, Yonsei University College of Medicine, Seoul, South Korea
| | | | - Carole H Sudre
- King's College London School of Biomedical Engineering and Imaging Sciences, London, United Kingdom.,Dementia Research Centre, Department of Neurodegenerative Disease, University College London Institute of Neurology, London, United Kingdom.,Centre for Medical Image Computing, Department of Medical Physics, University College London, London, United Kingdom
| | - Hanna Cho
- Dementia Research Centre, Department of Neurodegenerative Disease, University College London Institute of Neurology, London, United Kingdom
| | - Young Hoon Ryu
- Gangnam Severance Hospital, Department of Nuclear Medicine, Yonsei University College of Medicine, Seoul, South Korea
| | - Jae Yong Choi
- Gangnam Severance Hospital, Department of Nuclear Medicine, Yonsei University College of Medicine, Seoul, South Korea.,Division of Applied RI, Korea Institute Radiological and Medical Sciences, Seoul, South Korea
| | - Ruben Smith
- Lund University, Clinical Memory Research Unit, Lund, Sweden
| | - Olof Strandberg
- Lund University, Clinical Memory Research Unit, Lund, Sweden
| | | | - Joel Kramer
- Memory and Aging Center, Department of Neurology, University of California, San Francisco
| | - Adam L Boxer
- Memory and Aging Center, Department of Neurology, University of California, San Francisco
| | - Maria L Gorno-Tempini
- Memory and Aging Center, Department of Neurology, University of California, San Francisco
| | - Bruce L Miller
- Memory and Aging Center, Department of Neurology, University of California, San Francisco
| | - Renaud La Joie
- Memory and Aging Center, Department of Neurology, University of California, San Francisco
| | - Gil D Rabinovici
- Memory and Aging Center, Department of Neurology, University of California, San Francisco.,Department of Radiology and Biomedical Imaging, University of California, San Francisco.,Molecular Biophysics and Integrated Bioimaging Division, Lawrence Berkeley National Laboratory, Berkeley, California.,Associate Editor
| | - Oskar Hansson
- Lund University, Clinical Memory Research Unit, Lund, Sweden.,Memory Clinic, Skåne University Hospital, Malmö, Sweden
| |
Collapse
|
42
|
Palmqvist S, Eshaghi A. Spatial Distribution of Tau and β-Amyloid Pathologies and Their Role in Different Alzheimer Disease Phenotypes. Neurology 2020; 96:191-192. [PMID: 33262229 DOI: 10.1212/wnl.0000000000011272] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Affiliation(s)
- Sebastian Palmqvist
- From the Clinical Memory Research Unit (S.P.), Department of Clinical Sciences, Lund University, Memory Clinic (S.P.), Skåne University Hospital, Malmö, Sweden; and Queen Square Multiple Sclerosis Centre (A.E.), UCL Queen Square Institute of Neurology, Faculty of Brain Sciences, and Centre for Medical Image Computing (A.E.), Department of Computer Science, University College London. UK.
| | - Arman Eshaghi
- From the Clinical Memory Research Unit (S.P.), Department of Clinical Sciences, Lund University, Memory Clinic (S.P.), Skåne University Hospital, Malmö, Sweden; and Queen Square Multiple Sclerosis Centre (A.E.), UCL Queen Square Institute of Neurology, Faculty of Brain Sciences, and Centre for Medical Image Computing (A.E.), Department of Computer Science, University College London. UK
| |
Collapse
|
43
|
La Joie R, Visani AV, Lesman-Segev OH, Baker SL, Edwards L, Iaccarino L, Soleimani-Meigooni DN, Mellinger T, Janabi M, Miller ZA, Perry DC, Pham J, Strom A, Gorno-Tempini ML, Rosen HJ, Miller BL, Jagust WJ, Rabinovici GD. Association of APOE4 and Clinical Variability in Alzheimer Disease With the Pattern of Tau- and Amyloid-PET. Neurology 2020; 96:e650-e661. [PMID: 33262228 DOI: 10.1212/wnl.0000000000011270] [Citation(s) in RCA: 83] [Impact Index Per Article: 16.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2020] [Accepted: 09/11/2020] [Indexed: 02/06/2023] Open
Abstract
OBJECTIVE To assess whether Alzheimer disease (AD) clinical presentation and APOE4 relate to the burden and topography of β-amyloid (Aβ) and tau pathologies using in vivo PET imaging. METHODS We studied 119 Aβ-positive symptomatic patients aged 48-95 years, including 29 patients with logopenic variant primary progressive aphasia (lvPPA) and 21 with posterior cortical atrophy (PCA). Pittsburgh compound B (PiB)-Aβ and flortaucipir (tau)-PET standardized uptake value ratio (SUVR) images were created. General linear models assessed relationships between demographic/clinical variables (phenotype, age), APOE4, and PET (including global cortical and voxelwise SUVR values) while controlling for disease severity using the Clinical Dementia Rating Sum of Boxes. RESULTS PiB-PET binding showed a widespread cortical distribution with subtle differences across phenotypes and was unrelated to demographic/clinical variables or APOE4. Flortaucipir-PET was commonly elevated in temporoparietal regions, but showed marked phenotype-associated differences, with higher binding observed in occipito-parietal areas for PCA, in left temporal and inferior frontal for lvPPA, and in medial temporal areas for other AD. Cortical flortaucipir-PET binding was higher in younger patients across phenotypes (r = -0.63, 95% confidence interval [CI] -0.72, -0.50), especially in parietal and dorsal prefrontal cortices. The presence of APOE4 was associated with a focal medial temporal flortaucipir-SUVR increase, controlling for all other variables (entorhinal: + 0.310 SUVR, 95% CI 0.091, 0.530). CONCLUSIONS Clinical phenotypes are associated with differential patterns of tau but not amyloid pathology. Older age and APOE4 are not only risk factors for AD but also seem to affect disease expression by promoting a more medial temporal lobe-predominant pattern of tau pathology.
Collapse
Affiliation(s)
- Renaud La Joie
- From the Memory and Aging Center, Department of Neurology, Weill Institute for Neurosciences (R.L.J., A.V.V., O.H.L.-V., L.E., L.I., D.N.S.-M., T.M., Z.A.M., D.C.P., J.P., A.S., M.L.G.-T., H.J.R., B.L.M., G.D.R.), and Department of Radiology and Biomedical Imaging (G.D.R.), University of California, San Francisco; Department of Diagnostic Imaging (O.H.L.-V.), Sheba Medical Center, Tel Hashomer, Ramat Gan, Israel; Molecular Biophysics and Integrated Bioimaging Division (S.L.B., M.J., W.J.J., G.D.R.), Lawrence Berkeley National Laboratory; and Helen Wills Neuroscience Institute (W.J.J., G.D.R.), University of California Berkeley.
| | - Adrienne V Visani
- From the Memory and Aging Center, Department of Neurology, Weill Institute for Neurosciences (R.L.J., A.V.V., O.H.L.-V., L.E., L.I., D.N.S.-M., T.M., Z.A.M., D.C.P., J.P., A.S., M.L.G.-T., H.J.R., B.L.M., G.D.R.), and Department of Radiology and Biomedical Imaging (G.D.R.), University of California, San Francisco; Department of Diagnostic Imaging (O.H.L.-V.), Sheba Medical Center, Tel Hashomer, Ramat Gan, Israel; Molecular Biophysics and Integrated Bioimaging Division (S.L.B., M.J., W.J.J., G.D.R.), Lawrence Berkeley National Laboratory; and Helen Wills Neuroscience Institute (W.J.J., G.D.R.), University of California Berkeley
| | - Orit H Lesman-Segev
- From the Memory and Aging Center, Department of Neurology, Weill Institute for Neurosciences (R.L.J., A.V.V., O.H.L.-V., L.E., L.I., D.N.S.-M., T.M., Z.A.M., D.C.P., J.P., A.S., M.L.G.-T., H.J.R., B.L.M., G.D.R.), and Department of Radiology and Biomedical Imaging (G.D.R.), University of California, San Francisco; Department of Diagnostic Imaging (O.H.L.-V.), Sheba Medical Center, Tel Hashomer, Ramat Gan, Israel; Molecular Biophysics and Integrated Bioimaging Division (S.L.B., M.J., W.J.J., G.D.R.), Lawrence Berkeley National Laboratory; and Helen Wills Neuroscience Institute (W.J.J., G.D.R.), University of California Berkeley
| | - Suzanne L Baker
- From the Memory and Aging Center, Department of Neurology, Weill Institute for Neurosciences (R.L.J., A.V.V., O.H.L.-V., L.E., L.I., D.N.S.-M., T.M., Z.A.M., D.C.P., J.P., A.S., M.L.G.-T., H.J.R., B.L.M., G.D.R.), and Department of Radiology and Biomedical Imaging (G.D.R.), University of California, San Francisco; Department of Diagnostic Imaging (O.H.L.-V.), Sheba Medical Center, Tel Hashomer, Ramat Gan, Israel; Molecular Biophysics and Integrated Bioimaging Division (S.L.B., M.J., W.J.J., G.D.R.), Lawrence Berkeley National Laboratory; and Helen Wills Neuroscience Institute (W.J.J., G.D.R.), University of California Berkeley
| | - Lauren Edwards
- From the Memory and Aging Center, Department of Neurology, Weill Institute for Neurosciences (R.L.J., A.V.V., O.H.L.-V., L.E., L.I., D.N.S.-M., T.M., Z.A.M., D.C.P., J.P., A.S., M.L.G.-T., H.J.R., B.L.M., G.D.R.), and Department of Radiology and Biomedical Imaging (G.D.R.), University of California, San Francisco; Department of Diagnostic Imaging (O.H.L.-V.), Sheba Medical Center, Tel Hashomer, Ramat Gan, Israel; Molecular Biophysics and Integrated Bioimaging Division (S.L.B., M.J., W.J.J., G.D.R.), Lawrence Berkeley National Laboratory; and Helen Wills Neuroscience Institute (W.J.J., G.D.R.), University of California Berkeley
| | - Leonardo Iaccarino
- From the Memory and Aging Center, Department of Neurology, Weill Institute for Neurosciences (R.L.J., A.V.V., O.H.L.-V., L.E., L.I., D.N.S.-M., T.M., Z.A.M., D.C.P., J.P., A.S., M.L.G.-T., H.J.R., B.L.M., G.D.R.), and Department of Radiology and Biomedical Imaging (G.D.R.), University of California, San Francisco; Department of Diagnostic Imaging (O.H.L.-V.), Sheba Medical Center, Tel Hashomer, Ramat Gan, Israel; Molecular Biophysics and Integrated Bioimaging Division (S.L.B., M.J., W.J.J., G.D.R.), Lawrence Berkeley National Laboratory; and Helen Wills Neuroscience Institute (W.J.J., G.D.R.), University of California Berkeley
| | - David N Soleimani-Meigooni
- From the Memory and Aging Center, Department of Neurology, Weill Institute for Neurosciences (R.L.J., A.V.V., O.H.L.-V., L.E., L.I., D.N.S.-M., T.M., Z.A.M., D.C.P., J.P., A.S., M.L.G.-T., H.J.R., B.L.M., G.D.R.), and Department of Radiology and Biomedical Imaging (G.D.R.), University of California, San Francisco; Department of Diagnostic Imaging (O.H.L.-V.), Sheba Medical Center, Tel Hashomer, Ramat Gan, Israel; Molecular Biophysics and Integrated Bioimaging Division (S.L.B., M.J., W.J.J., G.D.R.), Lawrence Berkeley National Laboratory; and Helen Wills Neuroscience Institute (W.J.J., G.D.R.), University of California Berkeley
| | - Taylor Mellinger
- From the Memory and Aging Center, Department of Neurology, Weill Institute for Neurosciences (R.L.J., A.V.V., O.H.L.-V., L.E., L.I., D.N.S.-M., T.M., Z.A.M., D.C.P., J.P., A.S., M.L.G.-T., H.J.R., B.L.M., G.D.R.), and Department of Radiology and Biomedical Imaging (G.D.R.), University of California, San Francisco; Department of Diagnostic Imaging (O.H.L.-V.), Sheba Medical Center, Tel Hashomer, Ramat Gan, Israel; Molecular Biophysics and Integrated Bioimaging Division (S.L.B., M.J., W.J.J., G.D.R.), Lawrence Berkeley National Laboratory; and Helen Wills Neuroscience Institute (W.J.J., G.D.R.), University of California Berkeley
| | - Mustafa Janabi
- From the Memory and Aging Center, Department of Neurology, Weill Institute for Neurosciences (R.L.J., A.V.V., O.H.L.-V., L.E., L.I., D.N.S.-M., T.M., Z.A.M., D.C.P., J.P., A.S., M.L.G.-T., H.J.R., B.L.M., G.D.R.), and Department of Radiology and Biomedical Imaging (G.D.R.), University of California, San Francisco; Department of Diagnostic Imaging (O.H.L.-V.), Sheba Medical Center, Tel Hashomer, Ramat Gan, Israel; Molecular Biophysics and Integrated Bioimaging Division (S.L.B., M.J., W.J.J., G.D.R.), Lawrence Berkeley National Laboratory; and Helen Wills Neuroscience Institute (W.J.J., G.D.R.), University of California Berkeley
| | - Zachary A Miller
- From the Memory and Aging Center, Department of Neurology, Weill Institute for Neurosciences (R.L.J., A.V.V., O.H.L.-V., L.E., L.I., D.N.S.-M., T.M., Z.A.M., D.C.P., J.P., A.S., M.L.G.-T., H.J.R., B.L.M., G.D.R.), and Department of Radiology and Biomedical Imaging (G.D.R.), University of California, San Francisco; Department of Diagnostic Imaging (O.H.L.-V.), Sheba Medical Center, Tel Hashomer, Ramat Gan, Israel; Molecular Biophysics and Integrated Bioimaging Division (S.L.B., M.J., W.J.J., G.D.R.), Lawrence Berkeley National Laboratory; and Helen Wills Neuroscience Institute (W.J.J., G.D.R.), University of California Berkeley
| | - David C Perry
- From the Memory and Aging Center, Department of Neurology, Weill Institute for Neurosciences (R.L.J., A.V.V., O.H.L.-V., L.E., L.I., D.N.S.-M., T.M., Z.A.M., D.C.P., J.P., A.S., M.L.G.-T., H.J.R., B.L.M., G.D.R.), and Department of Radiology and Biomedical Imaging (G.D.R.), University of California, San Francisco; Department of Diagnostic Imaging (O.H.L.-V.), Sheba Medical Center, Tel Hashomer, Ramat Gan, Israel; Molecular Biophysics and Integrated Bioimaging Division (S.L.B., M.J., W.J.J., G.D.R.), Lawrence Berkeley National Laboratory; and Helen Wills Neuroscience Institute (W.J.J., G.D.R.), University of California Berkeley
| | - Julie Pham
- From the Memory and Aging Center, Department of Neurology, Weill Institute for Neurosciences (R.L.J., A.V.V., O.H.L.-V., L.E., L.I., D.N.S.-M., T.M., Z.A.M., D.C.P., J.P., A.S., M.L.G.-T., H.J.R., B.L.M., G.D.R.), and Department of Radiology and Biomedical Imaging (G.D.R.), University of California, San Francisco; Department of Diagnostic Imaging (O.H.L.-V.), Sheba Medical Center, Tel Hashomer, Ramat Gan, Israel; Molecular Biophysics and Integrated Bioimaging Division (S.L.B., M.J., W.J.J., G.D.R.), Lawrence Berkeley National Laboratory; and Helen Wills Neuroscience Institute (W.J.J., G.D.R.), University of California Berkeley
| | - Amelia Strom
- From the Memory and Aging Center, Department of Neurology, Weill Institute for Neurosciences (R.L.J., A.V.V., O.H.L.-V., L.E., L.I., D.N.S.-M., T.M., Z.A.M., D.C.P., J.P., A.S., M.L.G.-T., H.J.R., B.L.M., G.D.R.), and Department of Radiology and Biomedical Imaging (G.D.R.), University of California, San Francisco; Department of Diagnostic Imaging (O.H.L.-V.), Sheba Medical Center, Tel Hashomer, Ramat Gan, Israel; Molecular Biophysics and Integrated Bioimaging Division (S.L.B., M.J., W.J.J., G.D.R.), Lawrence Berkeley National Laboratory; and Helen Wills Neuroscience Institute (W.J.J., G.D.R.), University of California Berkeley
| | - Maria Luisa Gorno-Tempini
- From the Memory and Aging Center, Department of Neurology, Weill Institute for Neurosciences (R.L.J., A.V.V., O.H.L.-V., L.E., L.I., D.N.S.-M., T.M., Z.A.M., D.C.P., J.P., A.S., M.L.G.-T., H.J.R., B.L.M., G.D.R.), and Department of Radiology and Biomedical Imaging (G.D.R.), University of California, San Francisco; Department of Diagnostic Imaging (O.H.L.-V.), Sheba Medical Center, Tel Hashomer, Ramat Gan, Israel; Molecular Biophysics and Integrated Bioimaging Division (S.L.B., M.J., W.J.J., G.D.R.), Lawrence Berkeley National Laboratory; and Helen Wills Neuroscience Institute (W.J.J., G.D.R.), University of California Berkeley
| | - Howard J Rosen
- From the Memory and Aging Center, Department of Neurology, Weill Institute for Neurosciences (R.L.J., A.V.V., O.H.L.-V., L.E., L.I., D.N.S.-M., T.M., Z.A.M., D.C.P., J.P., A.S., M.L.G.-T., H.J.R., B.L.M., G.D.R.), and Department of Radiology and Biomedical Imaging (G.D.R.), University of California, San Francisco; Department of Diagnostic Imaging (O.H.L.-V.), Sheba Medical Center, Tel Hashomer, Ramat Gan, Israel; Molecular Biophysics and Integrated Bioimaging Division (S.L.B., M.J., W.J.J., G.D.R.), Lawrence Berkeley National Laboratory; and Helen Wills Neuroscience Institute (W.J.J., G.D.R.), University of California Berkeley
| | - Bruce L Miller
- From the Memory and Aging Center, Department of Neurology, Weill Institute for Neurosciences (R.L.J., A.V.V., O.H.L.-V., L.E., L.I., D.N.S.-M., T.M., Z.A.M., D.C.P., J.P., A.S., M.L.G.-T., H.J.R., B.L.M., G.D.R.), and Department of Radiology and Biomedical Imaging (G.D.R.), University of California, San Francisco; Department of Diagnostic Imaging (O.H.L.-V.), Sheba Medical Center, Tel Hashomer, Ramat Gan, Israel; Molecular Biophysics and Integrated Bioimaging Division (S.L.B., M.J., W.J.J., G.D.R.), Lawrence Berkeley National Laboratory; and Helen Wills Neuroscience Institute (W.J.J., G.D.R.), University of California Berkeley
| | - William J Jagust
- From the Memory and Aging Center, Department of Neurology, Weill Institute for Neurosciences (R.L.J., A.V.V., O.H.L.-V., L.E., L.I., D.N.S.-M., T.M., Z.A.M., D.C.P., J.P., A.S., M.L.G.-T., H.J.R., B.L.M., G.D.R.), and Department of Radiology and Biomedical Imaging (G.D.R.), University of California, San Francisco; Department of Diagnostic Imaging (O.H.L.-V.), Sheba Medical Center, Tel Hashomer, Ramat Gan, Israel; Molecular Biophysics and Integrated Bioimaging Division (S.L.B., M.J., W.J.J., G.D.R.), Lawrence Berkeley National Laboratory; and Helen Wills Neuroscience Institute (W.J.J., G.D.R.), University of California Berkeley
| | - Gil D Rabinovici
- From the Memory and Aging Center, Department of Neurology, Weill Institute for Neurosciences (R.L.J., A.V.V., O.H.L.-V., L.E., L.I., D.N.S.-M., T.M., Z.A.M., D.C.P., J.P., A.S., M.L.G.-T., H.J.R., B.L.M., G.D.R.), and Department of Radiology and Biomedical Imaging (G.D.R.), University of California, San Francisco; Department of Diagnostic Imaging (O.H.L.-V.), Sheba Medical Center, Tel Hashomer, Ramat Gan, Israel; Molecular Biophysics and Integrated Bioimaging Division (S.L.B., M.J., W.J.J., G.D.R.), Lawrence Berkeley National Laboratory; and Helen Wills Neuroscience Institute (W.J.J., G.D.R.), University of California Berkeley
| |
Collapse
|
44
|
Emrani S, Arain HA, DeMarshall C, Nuriel T. APOE4 is associated with cognitive and pathological heterogeneity in patients with Alzheimer's disease: a systematic review. Alzheimers Res Ther 2020; 12:141. [PMID: 33148345 PMCID: PMC7643479 DOI: 10.1186/s13195-020-00712-4] [Citation(s) in RCA: 98] [Impact Index Per Article: 19.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2020] [Accepted: 10/22/2020] [Indexed: 02/06/2023]
Abstract
Possession of the ε4 allele of apolipoprotein E (APOE) is the primary genetic risk factor for the sporadic form of Alzheimer's disease (AD). While researchers have extensively characterized the impact that APOE ε4 (APOE4) has on the susceptibility of AD, far fewer studies have investigated the phenotypic differences of patients with AD who are APOE4 carriers vs. those who are non-carriers. In order to understand these differences, we performed a qualitative systematic literature review of the reported cognitive and pathological differences between APOE4-positive (APOE4+) vs. APOE4-negative (APOE4-) AD patients. The studies performed on this topic to date suggest that APOE4 is not only an important mediator of AD susceptibility, but that it likely confers specific phenotypic heterogeneity in AD presentation, as well. Specifically, APOE4+ AD patients appear to possess more tau accumulation and brain atrophy in the medial temporal lobe, resulting in greater memory impairment, compared to APOE4- AD patients. On the other hand, APOE4- AD patients appear to possess more tau accumulation and brain atrophy in the frontal and parietal lobes, resulting in greater impairment in executive function, visuospatial abilities, and language, compared to APOE4+ AD patients. Although more work is necessary to validate and interrogate these findings, these initial observations of pathological and cognitive heterogeneity between APOE4+ vs. APOE4- AD patients suggest that there is a fundamental divergence in AD manifestation related to APOE genotype, which may have important implications in regard to the therapeutic treatment of these two patient populations.
Collapse
Affiliation(s)
- Sheina Emrani
- Department of Psychology, Rowan University, 201 Mullica Hill Road, Glassboro, NJ, 08028, USA
| | - Hirra A Arain
- Department of Pathology and Cell Biology, Columbia University, 630 West 168th Street, New York, NY, 10032, USA
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University, 630 West 168th Street, New York, NY, 10032, USA
| | - Cassandra DeMarshall
- Department of Geriatrics and Gerontology, Rowan University School of Osteopathic Medicine, One Medical Center Drive, Stratford, NJ, 08084, USA
| | - Tal Nuriel
- Department of Pathology and Cell Biology, Columbia University, 630 West 168th Street, New York, NY, 10032, USA.
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University, 630 West 168th Street, New York, NY, 10032, USA.
| |
Collapse
|
45
|
Baek MS, Cho H, Lee HS, Lee JH, Ryu YH, Lyoo CH. Effect of APOE ε4 genotype on amyloid-β and tau accumulation in Alzheimer's disease. ALZHEIMERS RESEARCH & THERAPY 2020; 12:140. [PMID: 33129364 PMCID: PMC7603688 DOI: 10.1186/s13195-020-00710-6] [Citation(s) in RCA: 72] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/07/2020] [Accepted: 10/20/2020] [Indexed: 02/12/2023]
Abstract
Background To assess the effects of apolipoprotein E (ApoE) ε4 genotype on amyloid-β (Aβ) and tau burden and their longitudinal changes in Alzheimer’s disease (AD) spectrum. Methods Among 272 individuals who underwent PET scans (18F-florbetaben for Aβ and 18F-flortaucipir for tau) and ApoE genotyping, 187 individuals completed 2-year follow-up PET scans. After correcting for the partial volume effect, we compared the standardized uptake value ratio (SUVR) for Aβ and tau burden between the ε4+ and ε4− groups. By using a linear mixed-effect model, we measured changes in SUVR in the ApoE ε4+ and ε4− groups. Results The ε4+ group showed greater baseline Aβ burden in the diffuse cortical regions and greater tau burden in the lateral, and medial temporal, cingulate, and insula cortices. Tau accumulation rate was higher in the parietal, occipital, lateral, and medial temporal cortices in the ε4+ group. In Aβ+ individuals, baseline tau burden was greater in the medial temporal cortex, while Aβ burden was conversely greater in the ε4− group. Tau accumulation rate was higher in the ε4+ group in a small region in the lateral temporal cortex. The effect of ApoE ε4 on enhanced tau accumulation persisted even after adjusting for the global cortical Aβ burden. Conclusions Progressive tau accumulation may be more prominent in ε4 carriers, particularly in the medial and lateral temporal cortices. ApoE ε4 allele has differential effects on the Aβ burden depending on the existing amyloidosis and may enhance vulnerability to progressive tau accumulation in the AD spectrum independent of Aβ. Supplementary information Supplementary information accompanies this paper at 10.1186/s13195-020-00710-6.
Collapse
Affiliation(s)
- Min Seok Baek
- Department of Neurology, Gangnam Severance Hospital, Yonsei University College of Medicine, 20 Eonjuro 63-gil, Gangnam-gu, Seoul, South Korea
| | - Hanna Cho
- Department of Neurology, Gangnam Severance Hospital, Yonsei University College of Medicine, 20 Eonjuro 63-gil, Gangnam-gu, Seoul, South Korea
| | - Hye Sun Lee
- Biostatistics Collaboration Unit, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, South Korea
| | - Jae Hoon Lee
- Department of Nuclear Medicine, Gangnam Severance Hospital, Yonsei University College of Medicine, 20 Eonjuro 63-gil, Gangnam-gu, Seoul, South Korea
| | - Young Hoon Ryu
- Department of Nuclear Medicine, Gangnam Severance Hospital, Yonsei University College of Medicine, 20 Eonjuro 63-gil, Gangnam-gu, Seoul, South Korea
| | - Chul Hyoung Lyoo
- Department of Neurology, Gangnam Severance Hospital, Yonsei University College of Medicine, 20 Eonjuro 63-gil, Gangnam-gu, Seoul, South Korea.
| |
Collapse
|
46
|
Rowley PA, Samsonov AA, Betthauser TJ, Pirasteh A, Johnson SC, Eisenmenger LB. Amyloid and Tau PET Imaging of Alzheimer Disease and Other Neurodegenerative Conditions. Semin Ultrasound CT MR 2020; 41:572-583. [PMID: 33308496 DOI: 10.1053/j.sult.2020.08.011] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Although diagnosing the syndrome of dementia is largely a clinical endeavor, neuroimaging plays an increasingly important role in accurately determining the underlying etiology, which extends beyond its traditional role in excluding other causes of altered cognition. New neuroimaging methods not only facilitate the diagnosis of the most common neurodegenerative conditions (particularly Alzheimer Disease [AD]) after symptom onset, but also show diagnostic promise even in the very early or presymptomatic phases of disease. Positron emission tomography (PET) is increasingly recognized as a key clinical tool for differentiating normal age-related changes in brain metabolism (using 18F-fluorodeoxyglucose [FDG]) from those seen in the earliest stages of specific forms of dementia. However, FDG PET only demonstrates nonspecific changes in altered parenchymal glucose uptake and not the specific etiologic proteinopathy causing the abnormal glucose uptake. A growing class of radiotracers targeting specific protein aggregates for amyloid-β (Aβ) and tau are changing the way AD is diagnosed, as these radiotracers directly label the underlying disease pathology. As these pathology-specific radiotracers are currently making their way to the clinic, it is important for the clinical neuroradiologist to understand the underlying patterns of Aβ and tau deposition in the context of AD (across its clinical continuum) and in other causes of dementia, as well as understand the implications of current research.
Collapse
Affiliation(s)
- Paul A Rowley
- Department of Radiology, University of Wisconsin, Madison, WI
| | | | | | - Ali Pirasteh
- Department of Radiology, University of Wisconsin, Madison, WI
| | | | | |
Collapse
|
47
|
Jellinger KA. Neuropathological assessment of the Alzheimer spectrum. J Neural Transm (Vienna) 2020; 127:1229-1256. [PMID: 32740684 DOI: 10.1007/s00702-020-02232-9] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2020] [Accepted: 07/14/2020] [Indexed: 12/12/2022]
Abstract
Alzheimer disease (AD), the most common form of dementia globally, classically defined a clinicopathological entity, is a heterogenous disorder with various pathobiological subtypes, currently referred to as Alzheimer continuum. Its morphological hallmarks are extracellular parenchymal β-amyloid (amyloid plaques) and intraneuronal (tau aggregates forming neurofibrillary tangles) lesions accompanied by synaptic loss and vascular amyloid deposits, that are essential for the pathological diagnosis of AD. In addition to "classical" AD, several subtypes with characteristic regional patterns of tau pathology have been described that show distinct clinical features, differences in age, sex distribution, biomarker levels, and patterns of key network destructions responsible for cognitive decline. AD is a mixed proteinopathy (amyloid and tau), frequently associated with other age-related co-pathologies, such as cerebrovascular lesions, Lewy and TDP-43 pathologies, hippocampal sclerosis, or argyrophilic grain disease. These and other co-pathologies essentially influence the clinical picture of AD and may accelerate disease progression. The purpose of this review is to provide a critical overview of AD pathology, its defining pathological substrates, and the heterogeneity among the Alzheimer spectrum entities that may provide a broader diagnostic coverage of this devastating disorder as a basis for implementing precision medicine approaches and for ultimate development of successful disease-modifying drugs for AD.
Collapse
Affiliation(s)
- Kurt A Jellinger
- Institute of Clinical Neurobiology, Alberichgasse 5/13, 1150, Vienna, Austria.
| |
Collapse
|
48
|
Rodriguez-Vieitez E, Nielsen HM. Associations Between APOE Variants, Tau and α-Synuclein. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1184:177-186. [PMID: 32096038 DOI: 10.1007/978-981-32-9358-8_15] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
Neurodegenerative diseases are characterized by the aggregation and deposition of misfolded proteins in the brain, most prominently amyloid-β (Aβ), tau and α-synuclein (α-syn), and are thus referred to as proteinopathies. While tau is a hallmark of Alzheimer's disease (AD) and other non-AD tauopathies, and α-synuclein is the pathological feature of the spectrum of synucleinopathies including Parkinson's disease (PD), Parkinson's disease with dementia (PDD) and dementia with Lewy bodies (DLB), the presence of co-pathologies is very frequent in all these diseases. Positive and synergistic associations between the different types of protein deposits have been reported, leading to worse prognosis and cognitive decline. A large variation in phenotypic clinical presentation of these diseases, largely due to the frequent presence of co-pathologies, makes differential diagnosis challenging. The observed clinico-pathological overlaps suggest common underlying mechanisms, in part due to shared genetic risk factors. The ε4 allele of the apolipoprotein (APOE) gene is one of the major genetic risk factors for the sporadic forms of proteinopathies, but the biological mechanisms linking APOE, tau and α-syn are not fully understood. This chapter describes current experimental evidence on the relationships between APOE variants, tau and α-syn, from clinical studies on fluid biomarkers and positron emission tomography (PET) imaging, and from basic experimental studies in cellular/molecular biology and animal models. The chapter focuses on recent advances and identifies knowledge gaps. In particular, no PET tracer for assessment of brain α-syn deposits is yet available, although it is subject of intense research and development, therefore experimental evidence on in vivo α-syn levels is based on measures in the cerebrospinal fluid (CSF) and plasma. Moreover, tau PET imaging studies comparing the patterns of tracer retention in synucleinopathies versus in other proteinopathies are scarce and much is still unknown regarding the relationships between APOE variants and fluid and/or imaging biomarkers of tau and α-syn. Further research incorporating multimodal imaging, fluid biomarkers and genetic factors will help elucidate the biological mechanisms underlying these proteinopathies, and contribute to differential diagnosis and patient stratification for clinical trials.
Collapse
Affiliation(s)
- Elena Rodriguez-Vieitez
- Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden
| | - Henrietta M Nielsen
- Department of Biochemistry and Biophysics, Stockholm University, Stockholm, Sweden.
| |
Collapse
|
49
|
Koller EJ, Gonzalez De La Cruz E, Weinrich M, Williams T, Cruz PE, Ryu D, Golde TE, Sullivan PM, Lewis J, Borchelt DR, Chakrabarty P. Intracerebral Expression of AAV-APOE4 Is Not Sufficient to Alter Tau Burden in Two Distinct Models of Tauopathy. Mol Neurobiol 2020; 57:1986-2001. [PMID: 31903524 PMCID: PMC7125036 DOI: 10.1007/s12035-019-01859-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2019] [Accepted: 12/15/2019] [Indexed: 12/14/2022]
Abstract
Apolipoprotein E4 (APOE4) is the major genetic risk factor for sporadic Alzheimer's disease (AD), which is characterized by amyloid β (Aβ) plaques and tau tangles. Though the role of APOE4 in Aβ pathogenesis has been mechanistically defined in rodent models, much less is known regarding the relationship of APOE4 to tau pathogenesis. Recent studies have indicated a possible correlation between APOE isoform-dependent alterations in tau pathology and neurodegeneration. To explore whether neuronal expression of APOE4 triggers tauopathy, here we delivered adeno-associated viruses (AAV) expressing human APOE4 in two different models of tauopathy-rTg4510 and PS19 lines. Intracerebroventricular delivery of AAV-APOE4 in neonatal rTg4510 and PS19 mice resulted in increased APOE4 protein in neurons but did not result in altered phosphorylated tau burden, pretangle tau pathology, or silver-positive tangle pathology. Biochemical analysis of synaptic proteins did not reveal substantial alterations. Our results indicate that over-expression of APOE4 in neurons, using an AAV-mediated approache, is not sufficient to accelerate or otherwise alter the inherent tau pathology that occurs in mice overexpressing mutant human tau.
Collapse
Affiliation(s)
- Emily J Koller
- Department of Neuroscience, University of Florida, Gainesville, FL, 32610, USA
- Center for Translational Research in Neurodegenerative Disease, University of Florida, Gainesville, FL, 32610, USA
| | - Elsa Gonzalez De La Cruz
- Center for Translational Research in Neurodegenerative Disease, University of Florida, Gainesville, FL, 32610, USA
| | - Mary Weinrich
- Center for Translational Research in Neurodegenerative Disease, University of Florida, Gainesville, FL, 32610, USA
| | - Tosha Williams
- Department of Neuroscience, University of Florida, Gainesville, FL, 32610, USA
- Center for Translational Research in Neurodegenerative Disease, University of Florida, Gainesville, FL, 32610, USA
| | - Pedro E Cruz
- Center for Translational Research in Neurodegenerative Disease, University of Florida, Gainesville, FL, 32610, USA
| | - Daniel Ryu
- Center for Translational Research in Neurodegenerative Disease, University of Florida, Gainesville, FL, 32610, USA
| | - Todd E Golde
- Department of Neuroscience, University of Florida, Gainesville, FL, 32610, USA
- Center for Translational Research in Neurodegenerative Disease, University of Florida, Gainesville, FL, 32610, USA
- McKnight Brain Institute, University of Florida, Gainesville, FL, 32610, USA
| | - Patrick M Sullivan
- Department of Medicine, Duke University, Durham VA Geriatric Research, Education and Clinical Center, Durham, NC, 27710, USA
| | - Jada Lewis
- Department of Neuroscience, University of Florida, Gainesville, FL, 32610, USA
- Center for Translational Research in Neurodegenerative Disease, University of Florida, Gainesville, FL, 32610, USA
- McKnight Brain Institute, University of Florida, Gainesville, FL, 32610, USA
| | - David R Borchelt
- Department of Neuroscience, University of Florida, Gainesville, FL, 32610, USA
- Center for Translational Research in Neurodegenerative Disease, University of Florida, Gainesville, FL, 32610, USA
- McKnight Brain Institute, University of Florida, Gainesville, FL, 32610, USA
| | - Paramita Chakrabarty
- Department of Neuroscience, University of Florida, Gainesville, FL, 32610, USA.
- Center for Translational Research in Neurodegenerative Disease, University of Florida, Gainesville, FL, 32610, USA.
- McKnight Brain Institute, University of Florida, Gainesville, FL, 32610, USA.
| |
Collapse
|
50
|
Ferreira D, Nordberg A, Westman E. Biological subtypes of Alzheimer disease: A systematic review and meta-analysis. Neurology 2020; 94:436-448. [PMID: 32047067 PMCID: PMC7238917 DOI: 10.1212/wnl.0000000000009058] [Citation(s) in RCA: 250] [Impact Index Per Article: 50.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2019] [Accepted: 12/17/2019] [Indexed: 12/14/2022] Open
Abstract
OBJECTIVE To test the hypothesis that distinct subtypes of Alzheimer disease (AD) exist and underlie the heterogeneity within AD, we conducted a systematic review and meta-analysis on AD subtype studies based on postmortem and neuroimaging data. METHODS EMBASE, PubMed, and Web of Science databases were consulted until July 2019. RESULTS Neuropathology and neuroimaging studies have consistently identified 3 subtypes of AD based on the distribution of tau-related pathology and regional brain atrophy: typical, limbic-predominant, and hippocampal-sparing AD. A fourth subtype, minimal atrophy AD, has been identified in several neuroimaging studies. Typical AD displays tau-related pathology and atrophy both in hippocampus and association cortex and has a pooled frequency of 55%. Limbic-predominant, hippocampal-sparing, and minimal atrophy AD had a pooled frequency of 21%, 17%, and 15%, respectively. Between-subtype differences were found in age at onset, age at assessment, sex distribution, years of education, global cognitive status, disease duration, APOE ε4 genotype, and CSF biomarker levels. CONCLUSION We identified 2 core dimensions of heterogeneity: typicality and severity. We propose that these 2 dimensions determine individuals' belonging to one of the AD subtypes based on the combination of protective factors, risk factors, and concomitant non-AD brain pathologies. This model is envisioned to aid with framing hypotheses, study design, interpretation of results, and understanding mechanisms in future subtype studies. Our model can be used along the A/T/N classification scheme for AD biomarkers. Unraveling the heterogeneity within AD is critical for implementing precision medicine approaches and for ultimately developing successful disease-modifying drugs for AD.
Collapse
Affiliation(s)
- Daniel Ferreira
- From the Division of Clinical Geriatrics (D.F., A.N., E.W.), Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Huddinge, Sweden; Theme Aging (A.N.), Karolinska University Hospital, Huddinge, Sweden; and Department of Neuroimaging (E.W.), Centre for Neuroimaging Sciences, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, United Kingdom.
| | - Agneta Nordberg
- From the Division of Clinical Geriatrics (D.F., A.N., E.W.), Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Huddinge, Sweden; Theme Aging (A.N.), Karolinska University Hospital, Huddinge, Sweden; and Department of Neuroimaging (E.W.), Centre for Neuroimaging Sciences, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, United Kingdom
| | - Eric Westman
- From the Division of Clinical Geriatrics (D.F., A.N., E.W.), Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Huddinge, Sweden; Theme Aging (A.N.), Karolinska University Hospital, Huddinge, Sweden; and Department of Neuroimaging (E.W.), Centre for Neuroimaging Sciences, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, United Kingdom
| |
Collapse
|