1
|
Gheni G, Shinohara M, Masuda-Suzukake M, Shindo A, Watanabe A, Kawai K, Bu G, Tomimoto H, Hasegawa M, Sato N. Cerebral hypoperfusion reduces tau accumulation. Ann Clin Transl Neurol 2025; 12:69-85. [PMID: 39621511 DOI: 10.1002/acn3.52247] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Revised: 10/03/2024] [Accepted: 10/24/2024] [Indexed: 01/22/2025] Open
Abstract
OBJECTIVE Alzheimer's disease (AD) often coexists with cerebrovascular diseases. However, the impact of cerebrovascular diseases such as stroke on AD pathology remains poorly understood. METHODS This study examines the correlation between cerebrovascular diseases and AD pathology. The research was carried out using clinical and neuropathological data collected from the National Alzheimer's Coordinating Center (NACC) database and an animal model in which bilateral common carotid artery stenosis surgery was performed, following the injection of tau seeds into the brains of wild-type mice. RESULTS Analysis of the NACC database suggests that clinical stroke history and lacunar infarcts are associated with lower neurofibrillary tangle pathology. An animal model demonstrates that chronic cerebral hypoperfusion reduces tau pathology, which was observed in not only neurons but also astrocytes, microglia, and oligodendrocytes. Furthermore, we found that astrocytes and microglia were activated in response to tau pathology and chronic cerebral hypoperfusion. Additionally, cerebral hypoperfusion increased a lysosomal enzyme, cathepsin D. INTERPRETATION These data together indicate that cerebral hypoperfusion reduces tau accumulation likely through an increase in microglial phagocytic activity towards tau and an elevation in degradation through cathepsin D. This study contributes to understanding the relationship between tau pathology and cerebrovascular diseases in older people with multimorbidity.
Collapse
Grants
- MEXT26293167 Grants-in-Aid from Japan Promotion of Science; the Japanese Ministry of Education, Culture, Sports, Science, and Technology
- MEXT15K15272 Grants-in-Aid from Japan Promotion of Science; the Japanese Ministry of Education, Culture, Sports, Science, and Technology
- MEXT17H04154 Grants-in-Aid from Japan Promotion of Science; the Japanese Ministry of Education, Culture, Sports, Science, and Technology
- MEXT21H02844 Grants-in-Aid from Japan Promotion of Science; the Japanese Ministry of Education, Culture, Sports, Science, and Technology
- MEXT24K02361 Grants-in-Aid from Japan Promotion of Science; the Japanese Ministry of Education, Culture, Sports, Science, and Technology
- SENSHIN Medical Research Foundation Research Grant
- Takeda Science Foundation Research Encouragement Grant
- Takeda Medical Research Foundation Research
- Novartis Foundation for Gerontological Research Award
- Annual Research Award Grant from the Japanese Society of Anti-aging Medicine
- 28-45 Funding for Longevity Sciences from the National Center for Geriatrics and Gerontology
- 19-9 Funding for Longevity Sciences from the National Center for Geriatrics and Gerontology
- 19-3 Funding for Longevity Sciences from the National Center for Geriatrics and Gerontology
- 21-12 Funding for Longevity Sciences from the National Center for Geriatrics and Gerontology
- 24-16 Funding for Longevity Sciences from the National Center for Geriatrics and Gerontology
- Mitsui Sumitomo Insurance Welfare Foundation
Collapse
Affiliation(s)
- Ghupurjan Gheni
- Department of Aging Neurobiology, Center for Development of Advanced Medicine for Dementia, National Center for Geriatrics and Gerontology, 7-430 Morioka, Obu, Aichi, 474-8511, Japan
| | - Mitsuru Shinohara
- Department of Aging Neurobiology, Center for Development of Advanced Medicine for Dementia, National Center for Geriatrics and Gerontology, 7-430 Morioka, Obu, Aichi, 474-8511, Japan
- Department of Neuroscience, Mayo Clinic, Jacksonville, Florida, 32224, USA
| | - Masami Masuda-Suzukake
- Dementia Research Project, Tokyo Metropolitan Institute of Medical Science, 2-1-6 Kamikitazawa, Setagaya-ku, Tokyo, 156-8506, Japan
| | - Akihiko Shindo
- Department of Neurology, Graduate School of Medicine, Mie University, 1577 Kurima Machiyacho, Tsu City, Mie, 514-8507, Japan
| | - Atsushi Watanabe
- Equipment Management Division, Center for Core Facility Administration, National Center for Geriatrics and Gerontology, 7-430 Morioka, Obu, Aichi, 474-8511, Japan
| | - Kaori Kawai
- Department of Aging Neurobiology, Center for Development of Advanced Medicine for Dementia, National Center for Geriatrics and Gerontology, 7-430 Morioka, Obu, Aichi, 474-8511, Japan
| | - Guojun Bu
- Department of Neuroscience, Mayo Clinic, Jacksonville, Florida, 32224, USA
- Division of Life Science, The Hong Kong University of Science and Technology, Hong Kong, China
| | - Hidekazu Tomimoto
- Department of Neurology, Graduate School of Medicine, Mie University, 1577 Kurima Machiyacho, Tsu City, Mie, 514-8507, Japan
| | - Masato Hasegawa
- Dementia Research Project, Tokyo Metropolitan Institute of Medical Science, 2-1-6 Kamikitazawa, Setagaya-ku, Tokyo, 156-8506, Japan
| | - Naoyuki Sato
- Department of Aging Neurobiology, Center for Development of Advanced Medicine for Dementia, National Center for Geriatrics and Gerontology, 7-430 Morioka, Obu, Aichi, 474-8511, Japan
| |
Collapse
|
2
|
Wang S, Wang Y, Xu FH, Tian X, Fredericks CA, Shen L, Zhao Y. Sex-specific topological structure associated with dementia via latent space estimation. Alzheimers Dement 2024; 20:8387-8401. [PMID: 39530632 DOI: 10.1002/alz.14266] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2024] [Revised: 08/13/2024] [Accepted: 08/26/2024] [Indexed: 11/16/2024]
Abstract
INTRODUCTION We investigate sex-specific topological structures associated with typical Alzheimer's disease (AD) dementia using a novel state-of-the-art latent space estimation technique. METHODS This study applies a probabilistic approach for latent space estimation that extends current multiplex network modeling approaches and captures the higher-order dependence in functional connectomes by preserving transitivity and modularity structures. RESULTS We find sex differences in network topology with females showing more default mode network (DMN)-centered hyperactivity and males showing more limbic system (LS)-centered hyperactivity, while both show DMN-centered hypoactivity. We find that centrality plays an important role in dementia-related dysfunction with stronger association between connectivity changes and regional centrality in females than in males. DISCUSSION The study contributes to the current literature by providing a more comprehensive picture of dementia-related neurodegeneration linking centrality, network segregation, and DMN-centered changes in functional connectomes, and how these components of neurodegeneration differ between the sexes. HIGHLIGHTS We find evidence supporting the active role network topology plays in neurodegeneration with an imbalance between the excitatory and inhibitory mechanisms that can lead to whole-brain destabilization in dementia patients. We find sex-based differences in network topology with females showing more default mode network (DMN)-centered hyperactivity, males showing more limbic system (LS)-centered hyperactivity, while both show DMN-centered hypoactivity. We find that brain region centrality plays an important role in dementia-related dysfunction with a stronger association between connectivity changes and regional centrality in females than in males. Females, compared to males, tend to exhibit stronger dementia-related changes in regions that are the central actors of the brain networks. Taken together, this research uniquely contributes to the current literature by providing a more comprehensive picture of dementia-related neurodegeneration linking centrality, network segregation, and DMN-centered changes in functional connectomes, and how these components of neurodegeneration differ between the sexes.
Collapse
Affiliation(s)
- Selena Wang
- Department of Biostatistics and Health Data Science, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Yiting Wang
- Department of Statistics, University of Virginia, Charlottesville, Virginia, USA
| | - Frederick H Xu
- Department of Bioengineering, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Xinyuan Tian
- Department of Neurology, Yale School of Medicine, Yale University, New Haven, Connecticut, USA
| | - Carolyn A Fredericks
- Department of Biostatistics, Epidemiology and Informatics, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Li Shen
- Department of Biostatistics, Yale School of Public Health, New Haven, Connecticut, USA
| | - Yize Zhao
- Department of Neurology, Yale School of Medicine, Yale University, New Haven, Connecticut, USA
| |
Collapse
|
3
|
Luan Y, Rubinski A, Biel D, Otero Svaldi D, Alonzo Higgins I, Shcherbinin S, Pontecorvo M, Franzmeier N, Ewers M. Tau-network mapping of domain-specific cognitive impairment in Alzheimer's disease. Neuroimage Clin 2024; 44:103699. [PMID: 39509992 PMCID: PMC11574813 DOI: 10.1016/j.nicl.2024.103699] [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: 04/15/2024] [Revised: 10/01/2024] [Accepted: 10/28/2024] [Indexed: 11/15/2024]
Abstract
Fibrillar tau gradually progresses in the brain during the course of Alzheimer's disease (AD). However, the contribution of tau accumulation in a given brain region to decline in different cognitive domains and thus phenotypic heterogeneity in AD remains unclear. Here, we leveraged the functional connectome to link the locality of tau accumulation to domain-specific cognitive impairment. In the current study, we mapped regional tau-PET accumulation onto the normative functional connectome. Subsequently, we cross-validated in two samples of AD-patients the associations between the tau-connectivity profiles and cognitive domains (episodic memory, executive function, or language). Lastly, we tested the effect of local tau-PET accumulation on the domain-specific tau-lesion networks and cognition. We identified cognitive-domain-specific tau-lesion networks, where closer topological proximity of tau-PET locations to a network was predictive of worse impairment in that domain. Higher tau-PET was associated with decreased domain-specific network connectivity, and the decrease in connectivity was associated with lower domain-specific cognition. The tau locations' connectivity profile explained domain-specific cognitive impairment, where disrupted connectivity may underlie the effect of tau on cognitive impairment.
Collapse
Affiliation(s)
- Ying Luan
- Department of Radiology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, China; Institute for Stroke and Dementia Research (ISD), University Hospital, Ludwig Maximilian University (LMU), Munich, Germany
| | - Anna Rubinski
- Institute for Stroke and Dementia Research (ISD), University Hospital, Ludwig Maximilian University (LMU), Munich, Germany
| | - Davina Biel
- Institute for Stroke and Dementia Research (ISD), University Hospital, Ludwig Maximilian University (LMU), Munich, Germany
| | | | | | | | | | - Nicolai Franzmeier
- Institute for Stroke and Dementia Research (ISD), University Hospital, Ludwig Maximilian University (LMU), Munich, Germany; Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Michael Ewers
- Institute for Stroke and Dementia Research (ISD), University Hospital, Ludwig Maximilian University (LMU), Munich, Germany; German Center for Neurodegenerative Diseases (DZNE), Munich, Germany.
| |
Collapse
|
4
|
Yang C, Tian S, Du W, Liu M, Hu R, Gao B, Pan T, Song Q, Liu T, Wang W, Zhang H, Miao Y. Glymphatic function assessment with diffusion tensor imaging along the perivascular space in patients with major depressive disorder and its relation to cerebral white-matter alteration. Quant Imaging Med Surg 2024; 14:6397-6412. [PMID: 39281139 PMCID: PMC11400689 DOI: 10.21037/qims-24-510] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2024] [Accepted: 07/22/2024] [Indexed: 09/18/2024]
Abstract
Background The link between glymphatic system function in the brain and alterations in white-matter microstructure among individuals with major depressive disorder (MDD) remains unclear. This study aimed to examine the assessment of glymphatic system function in patients with MDD using the diffusion tensor imaging along the perivascular space (DTI-ALPS) index and to evaluate its association with cerebral-white-matter abnormalities and neuropsychological scores. Methods From February 2023 to November 2023, this cross-sectional study recruited 35 patients with MDD from the Psychosomatic Diseases Department of the First Affiliated Hospital of Dalian Medical University. In this time period, 23 healthy controls (HCs) were enlisted from the community and matched with the MDD cohort in terms of years of education, gender, and age. All participants underwent magnetic resonance imaging, depression, anxiety, and cognitive assessments. The tract-based spatial statistics (TBSS) analyzed DTI parameters and identified significant clusters. Automated fiber quantification (AFQ) was used to automatically identify fiber bundles with statistical differences. Mann-Whitney tests or two-sample t-tests were used for comparisons. Interobserver consistency of the DTI-ALPS measurements was evaluated using the interclass correlation coefficient (ICC). Partial correlation analyses and linear regression analyses were used to examine relationships. A comparison of the DTI-ALPS index was made between the two groups. Correlations among diffusion characteristics, neuropsychological scores, and the DTI-ALPS index were analyzed. Results Compared to HCs, patients with MDD exhibited a lower DTI-ALPS score (P=0.001). According to using linear regression analysis, the ALPS index was found to be an independent predictor of the Hamilton Depression Rating Scale [B=-25.32; P=0.001; 95% confidence interval (CI): -40.35 to -11.55], Hamilton Anxiety Rating Scale (B=-33.48; P=0.003; 95% CI: -55.38 to -11.24), and Montreal Cognitive Assessment total score (B=8.59; P=0.008; 95% CI: 2.38 to 14.79). According to the TBSS analysis, there were clusters of increased axial diffusivity (AD), mean diffusivity (MD), and radial diffusivity (RD) in patients with MDD as compared to HCs (all P values <0.05). A lower DTI-ALPS score was correlated with higher AD (r=-0.592; P<0.001), MD (cluster 1: r=-0.567, P=0.001; cluster 2: r=-0.581, P<0.001), and RD (r=-0.491; P=0.004) values. AFQ analysis identified the significantly different diffusion indicators in the left cingulum bundle (CB_L), left inferior longitudinal fasciculus (ILF_L), and left uncinate fasciculus (UF_L) between the two groups (all false discovery rate P values <0.05). DTI-ALPS score was negatively correlated with the AD value of CB_L (r=-0.304; P=0.024), ILF_L (r=-0.35; P=0.008), and UF_L (r=-0.354; P=0.008) in AFQ tract-level analysis. In point-wise analysis, the MD value of CB_L at nodes 33 to 36 was negatively correlated with DTI-ALPS score (r ranging from -0.504 to -0.535; P<0.01). Conclusions Our results indicated a decrease in DTI-ALPS index score in patients with MDD. DTI-ALPS score was associated with depression, anxiety, declined cognitive ability, and white-matter microstructural abnormalities and may thus be a promising biomarker for the partial evaluation of glymphatic system function in patients with MDD.
Collapse
Affiliation(s)
- Chun Yang
- Department of Radiology, the First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Shiyun Tian
- Department of Radiology, the First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Wei Du
- Department of Radiology, the First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Meichen Liu
- Department of Neurology, the First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Rui Hu
- Department of Radiology, the First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Bingbing Gao
- Department of Radiology, the First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Tao Pan
- Department of Interventional Therapy, the First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Qingwei Song
- Department of Radiology, the First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Tieli Liu
- School of Medical Imaging, Dalian Medical University, Dalian, China
| | - Weiwei Wang
- Department of Radiology, the First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Huimin Zhang
- Department of Neurology, the First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Yanwei Miao
- Department of Radiology, the First Affiliated Hospital of Dalian Medical University, Dalian, China
| |
Collapse
|
5
|
Barrera-Ocampo A. Monoclonal antibodies and aptamers: The future therapeutics for Alzheimer's disease. Acta Pharm Sin B 2024; 14:2795-2814. [PMID: 39027235 PMCID: PMC11252463 DOI: 10.1016/j.apsb.2024.03.034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Revised: 03/06/2024] [Accepted: 03/14/2024] [Indexed: 07/20/2024] Open
Abstract
Alzheimer's disease (AD) is considered the most common and prevalent form of dementia of adult-onset with characteristic progressive impairment in cognition and memory. The cure for AD has not been found yet and the treatments available until recently were only symptomatic. Regardless of multidisciplinary approaches and efforts made by pharmaceutical companies, it was only in the past two years that new drugs were approved for the treatment of the disease. Amyloid beta (Aβ) immunotherapy is at the core of this therapy, which is one of the most innovative approaches looking to change the course of AD. This technology is based on synthetic peptides or monoclonal antibodies (mAb) to reduce Aβ levels in the brain and slow down the advance of neurodegeneration. Hence, this article reviews the state of the art about AD neuropathogenesis, the traditional pharmacologic treatment, as well as the modern active and passive immunization describing approved drugs, and drug prototypes currently under investigation in different clinical trials. In addition, future perspectives on immunotherapeutic strategies for AD and the rise of the aptamer technology as a non-immunogenic alternative to curb the disease progression are discussed.
Collapse
Affiliation(s)
- Alvaro Barrera-Ocampo
- Facultad de Ingeniería, Diseño y Ciencias Aplicadas, Departamento de Ciencias Farmacéuticas y Químicas, Grupo Natura, Universidad Icesi, Cali 760031, Colombia
| |
Collapse
|
6
|
Pini L, Lombardi G, Sansone G, Gaiola M, Padovan M, Volpin F, Denaro L, Corbetta M, Salvalaggio A. Indirect functional connectivity does not predict overall survival in glioblastoma. Neurobiol Dis 2024; 196:106521. [PMID: 38697575 DOI: 10.1016/j.nbd.2024.106521] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Revised: 04/14/2024] [Accepted: 04/29/2024] [Indexed: 05/05/2024] Open
Abstract
BACKGROUND Lesion network mapping (LNM) is a popular framework to assess clinical syndromes following brain injury. The classical approach involves embedding lesions from patients into a normative functional connectome and using the corresponding functional maps as proxies for disconnections. However, previous studies indicated limited predictive power of this approach in behavioral deficits. We hypothesized similarly low predictiveness for overall survival (OS) in glioblastoma (GBM). METHODS A retrospective dataset of patients with GBM was included (n = 99). Lesion masks were registered in the normative space to compute disconnectivity maps. The brain functional normative connectome consisted in data from 173 healthy subjects obtained from the Human Connectome Project. A modified version of the LNM was then applied to core regions of GBM masks. Linear regression, classification, and principal component (PCA) analyses were conducted to explore the relationship between disconnectivity and OS. OS was considered both as continuous and categorical (low, intermediate, and high survival) variable. RESULTS The results revealed no significant associations between OS and network disconnection strength when analyzed at both voxel-wise and classification levels. Moreover, patients stratified into different OS groups did not exhibit significant differences in network connectivity patterns. The spatial similarity among the first PCA of network maps for each OS group suggested a lack of distinctive network patterns associated with survival duration. CONCLUSIONS Compared with indirect structural measures, functional indirect mapping does not provide significant predictive power for OS in patients with GBM. These findings are consistent with previous research that demonstrated the limitations of indirect functional measures in predicting clinical outcomes, underscoring the need for more comprehensive methodologies and a deeper understanding of the factors influencing clinical outcomes in this challenging disease.
Collapse
Affiliation(s)
- Lorenzo Pini
- Padova Neuroscience Center, University of Padova, Italy
| | - Giuseppe Lombardi
- Department of Oncology, Oncology 1, Veneto Institute of Oncology IOV-IRCCS, Padova, Italy
| | - Giulio Sansone
- Departments of Neuroscience, University of Padova, Italy
| | - Matteo Gaiola
- Departments of Neuroscience, University of Padova, Italy
| | - Marta Padovan
- Department of Oncology, Oncology 1, Veneto Institute of Oncology IOV-IRCCS, Padova, Italy
| | - Francesco Volpin
- Division of Neurosurgery, Azienda Ospedaliera Università di Padova, Padova, Italy
| | - Luca Denaro
- Departments of Neuroscience, University of Padova, Italy
| | - Maurizio Corbetta
- Padova Neuroscience Center, University of Padova, Italy; Departments of Neuroscience, University of Padova, Italy; Veneto institute of Molecular Medicine (VIMM), Padova, Italy
| | | |
Collapse
|
7
|
Qiu T, Liu Z, Rheault F, Legarreta JH, Valcourt Caron A, St‐Onge F, Strikwerda‐Brown C, Metz A, Dadar M, Soucy J, Pichet Binette A, Spreng RN, Descoteaux M, Villeneuve S. Structural white matter properties and cognitive resilience to tau pathology. Alzheimers Dement 2024; 20:3364-3377. [PMID: 38561254 PMCID: PMC11095478 DOI: 10.1002/alz.13776] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2023] [Revised: 01/11/2024] [Accepted: 02/07/2024] [Indexed: 04/04/2024]
Abstract
INTRODUCTION We assessed whether macro- and/or micro-structural white matter properties are associated with cognitive resilience to Alzheimer's disease pathology years prior to clinical onset. METHODS We examined whether global efficiency, an indicator of communication efficiency in brain networks, and diffusion measurements within the limbic network and default mode network moderate the association between amyloid-β/tau pathology and cognitive decline. We also investigated whether demographic and health/risk factors are associated with white matter properties. RESULTS Higher global efficiency of the limbic network, as well as free-water corrected diffusion measures within the tracts of both networks, attenuated the impact of tau pathology on memory decline. Education, age, sex, white matter hyperintensities, and vascular risk factors were associated with white matter properties of both networks. DISCUSSION White matter can influence cognitive resilience against tau pathology, and promoting education and vascular health may enhance optimal white matter properties. HIGHLIGHTS Aβ and tau were associated with longitudinal memory change over ∼7.5 years. White matter properties attenuated the impact of tau pathology on memory change. Health/risk factors were associated with white matter properties.
Collapse
Affiliation(s)
- Ting Qiu
- Douglas Mental Health University InstituteMontrealCanada
| | - Zhen‐Qi Liu
- Montreal Neurological InstituteDepartment of Neurology and NeurosurgeryMcGill UniversityMontrealCanada
| | - François Rheault
- Medical Imaging and NeuroInformatics LabUniversité de SherbrookeSherbrookeCanada
| | - Jon Haitz Legarreta
- Department of RadiologyBrigham and Women's HospitalMass General Brigham/Harvard Medical SchoolBostonMassachusettsUSA
| | - Alex Valcourt Caron
- Sherbrooke Connectivity Imaging LaboratoryUniversité de SherbrookeSherbrookeCanada
| | | | - Cherie Strikwerda‐Brown
- Douglas Mental Health University InstituteMontrealCanada
- School of Psychological ScienceThe University of Western AustraliaPerthAustralia
| | - Amelie Metz
- Douglas Mental Health University InstituteMontrealCanada
| | - Mahsa Dadar
- Douglas Mental Health University InstituteMontrealCanada
- Department of PsychiatryMcGill UniversityMontrealCanada
| | - Jean‐Paul Soucy
- Montreal Neurological InstituteDepartment of Neurology and NeurosurgeryMcGill UniversityMontrealCanada
| | | | - R. Nathan Spreng
- Douglas Mental Health University InstituteMontrealCanada
- Montreal Neurological InstituteDepartment of Neurology and NeurosurgeryMcGill UniversityMontrealCanada
- Department of PsychiatryMcGill UniversityMontrealCanada
| | - Maxime Descoteaux
- Sherbrooke Connectivity Imaging LaboratoryUniversité de SherbrookeSherbrookeCanada
| | - Sylvia Villeneuve
- Douglas Mental Health University InstituteMontrealCanada
- Department of PsychiatryMcGill UniversityMontrealCanada
| | | |
Collapse
|
8
|
Heo S, Yoon CW, Kim SY, Kim WR, Na DL, Noh Y. Alterations of Structural Network Efficiency in Early-Onset and Late-Onset Alzheimer's Disease. J Clin Neurol 2024; 20:265-275. [PMID: 38330417 PMCID: PMC11076196 DOI: 10.3988/jcn.2023.0092] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Revised: 08/17/2023] [Accepted: 10/05/2023] [Indexed: 02/10/2024] Open
Abstract
BACKGROUND AND PURPOSE Early- and late-onset Alzheimer's disease (EOAD and LOAD, respectively) share the same neuropathological hallmarks of amyloid and neurofibrillary tangles but have distinct cognitive features. We compared structural brain connectivity between the EOAD and LOAD groups using structural network efficiency and evaluated the association of structural network efficiency with the cognitive profile and pathological markers of Alzheimer's disease (AD). METHODS The structural brain connectivity networks of 80 AD patients (47 with EOAD and 33 with LOAD) and 57 healthy controls were reconstructed using diffusion-tensor imaging. Graph-theoretic indices were calculated and intergroup differences were evaluated. Correlations between network parameters and neuropsychological test results were analyzed. The correlations of the amyloid and tau burdens with network parameters were evaluated for the patients and controls. RESULTS Compared with the age-matched control group, the EOAD patients had increased global path length and decreased global efficiency, averaged local efficiency, and averaged clustering coefficient. In contrast, no significant differences were found in the LOAD patients. Locally, the EOAD patients showed decreases in local efficiency and the clustering coefficient over a wide area compared with the control group, whereas LOAD patients showed such decreases only within a limited area. Changes in network parameters were significantly correlated with multiple cognitive domains in EOAD patients, but only with Clinical Dementia Rating Sum-of-Boxes scores in LOAD patients. Finally, the tau burden was correlated with changes in network parameters in AD signature areas in both patient groups, while there was no correlation with the amyloid burden. CONCLUSIONS The impairment of structural network efficiency and its effects on cognition may differ between EOAD and LOAD.
Collapse
Affiliation(s)
- Suyeon Heo
- Gachon University, College of Medicine, Incheon, Korea
| | - Cindy W Yoon
- Department of Neurology, Inha University School of Medicine, Incheon, Korea
| | - Sang-Young Kim
- Neuroscience Research Institute, Gachon University, Incheon, Korea
- MR Clinical Science, Health Systems, Philips Healthcare, Seoul, Korea
| | - Woo-Ram Kim
- Neuroscience Research Institute, Gachon University, Incheon, Korea
| | - Duk L Na
- Department of Neurology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
- Happymind Clinic, Seoul, Korea
| | - Young Noh
- Neuroscience Research Institute, Gachon University, Incheon, Korea
- Department of Neurology, Gil Medical Center, Gachon University College of Medicine, Incheon, Korea.
| |
Collapse
|
9
|
Chen H, Xu J, Li W, Hu Z, Ke Z, Qin R, Xu Y. The characteristic patterns of individual brain susceptibility networks underlie Alzheimer's disease and white matter hyperintensity-related cognitive impairment. Transl Psychiatry 2024; 14:177. [PMID: 38575556 PMCID: PMC10994911 DOI: 10.1038/s41398-024-02861-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/02/2024] [Revised: 03/04/2024] [Accepted: 03/06/2024] [Indexed: 04/06/2024] Open
Abstract
Excessive iron accumulation in the brain cortex increases the risk of cognitive deterioration. However, interregional relationships (defined as susceptibility connectivity) of local brain iron have not been explored, which could provide new insights into the underlying mechanisms of cognitive decline. Seventy-six healthy controls (HC), 58 participants with mild cognitive impairment due to probable Alzheimer's disease (MCI-AD) and 66 participants with white matter hyperintensity (WMH) were included. We proposed a novel approach to construct a brain susceptibility network by using Kullback‒Leibler divergence similarity estimation from quantitative susceptibility mapping and further evaluated its topological organization. Moreover, sparse logistic regression (SLR) was applied to classify MCI-AD from HC and WMH with normal cognition (WMH-NC) from WMH with MCI (WMH-MCI).The altered susceptibility connectivity in the MCI-AD patients indicated that relatively more connectivity was involved in the default mode network (DMN)-related and visual network (VN)-related connectivity, while more altered DMN-related and subcortical network (SN)-related connectivity was found in the WMH-MCI patients. For the HC vs. MCI-AD classification, the features selected by the SLR were primarily distributed throughout the DMN-related and VN-related connectivity (accuracy = 76.12%). For the WMH-NC vs. WMH-MCI classification, the features with high appearance frequency were involved in SN-related and DMN-related connectivity (accuracy = 84.85%). The shared and specific patterns of the susceptibility network identified in both MCI-AD and WMH-MCI may provide a potential diagnostic biomarker for cognitive impairment, which could enhance the understanding of the relationships between brain iron burden and cognitive decline from a network perspective.
Collapse
Affiliation(s)
- Haifeng Chen
- Nanjing Drum Tower Hospital Clinical College of Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, China
- Department of Neurology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
- Jiangsu Key Laboratory of Molecular Medicine, Medical School of Nanjing University, Nanjing, China
- Jiangsu Province Stroke Center for Diagnosis and Therapy, Nanjing, China
- Nanjing Neuropsychiatry Clinic Medical Center, Nanjing, China
| | - Jingxian Xu
- Department of Neurology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Weikai Li
- School of Mathematics and Statistics, Chongqing Jiaotong University, Chongqing, China
- MIIT Key Laboratory of Pattern Analysis and Machine Intelligence, Nanjing, China
| | - Zheqi Hu
- Department of Neurology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Zhihong Ke
- Department of Neurology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Ruomeng Qin
- Department of Neurology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
- Jiangsu Key Laboratory of Molecular Medicine, Medical School of Nanjing University, Nanjing, China
- Jiangsu Province Stroke Center for Diagnosis and Therapy, Nanjing, China
- Nanjing Neuropsychiatry Clinic Medical Center, Nanjing, China
| | - Yun Xu
- Nanjing Drum Tower Hospital Clinical College of Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, China.
- Department of Neurology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China.
- Jiangsu Key Laboratory of Molecular Medicine, Medical School of Nanjing University, Nanjing, China.
- Jiangsu Province Stroke Center for Diagnosis and Therapy, Nanjing, China.
- Nanjing Neuropsychiatry Clinic Medical Center, Nanjing, China.
| |
Collapse
|
10
|
Cabrera-Álvarez J, Stefanovski L, Martin L, Susi G, Maestú F, Ritter P. A Multiscale Closed-Loop Neurotoxicity Model of Alzheimer's Disease Progression Explains Functional Connectivity Alterations. eNeuro 2024; 11:ENEURO.0345-23.2023. [PMID: 38565295 PMCID: PMC11026343 DOI: 10.1523/eneuro.0345-23.2023] [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: 09/08/2023] [Revised: 12/05/2023] [Accepted: 12/22/2023] [Indexed: 04/04/2024] Open
Abstract
The accumulation of amyloid-β (Aβ) and hyperphosphorylated-tau (hp-tau) are two classical histopathological biomarkers in Alzheimer's disease (AD). However, their detailed interactions with the electrophysiological changes at the meso- and macroscale are not yet fully understood. We developed a mechanistic multiscale model of AD progression, linking proteinopathy to its effects on neural activity and vice-versa. We integrated a heterodimer model of prion-like protein propagation and a brain network model of Jansen-Rit neural masses derived from human neuroimaging data whose parameters varied due to neurotoxicity. Results showed that changes in inhibition guided the electrophysiological alterations found in AD, and these changes were mainly attributed to Aβ effects. Additionally, we found a causal disconnection between cellular hyperactivity and interregional hypersynchrony contrary to previous beliefs. Finally, we demonstrated that early Aβ and hp-tau depositions' location determine the spatiotemporal profile of the proteinopathy. The presented model combines the molecular effects of both Aβ and hp-tau together with a mechanistic protein propagation model and network effects within a closed-loop model. This holds the potential to enlighten the interplay between AD mechanisms on various scales, aiming to develop and test novel hypotheses on the contribution of different AD-related variables to the disease evolution.
Collapse
Affiliation(s)
- Jesús Cabrera-Álvarez
- Department of Experimental Psychology, Complutense University of Madrid, Pozuelo de Alarcón 28223, Spain
- Centre for Cognitive and Computational Neuroscience, Complutense University of Madrid, Madrid 28040, Spain
| | - Leon Stefanovski
- Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Berlin 10117, Germany
- Department of Neurology with Experimental Neurology, Brain Simulation Section, Charité - Universitätsmedizin Berlin, Berlin 10117, Germany
| | - Leon Martin
- Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Berlin 10117, Germany
- Department of Neurology with Experimental Neurology, Brain Simulation Section, Charité - Universitätsmedizin Berlin, Berlin 10117, Germany
| | - Gianluca Susi
- Centre for Cognitive and Computational Neuroscience, Complutense University of Madrid, Madrid 28040, Spain
- Department of Structure of Matter, Thermal Physics and Electronics, Complutense University of Madrid, Madrid 28040, Spain
| | - Fernando Maestú
- Department of Experimental Psychology, Complutense University of Madrid, Pozuelo de Alarcón 28223, Spain
- Centre for Cognitive and Computational Neuroscience, Complutense University of Madrid, Madrid 28040, Spain
| | - Petra Ritter
- Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Berlin 10117, Germany
- Department of Neurology with Experimental Neurology, Brain Simulation Section, Charité - Universitätsmedizin Berlin, Berlin 10117, Germany
- Bernstein Center for Computational Neuroscience Berlin, Berlin 10115, Germany
| |
Collapse
|
11
|
Yang DX, Sun Z, Yu MM, Zou QQ, Li PY, Zhang JK, Wu X, Li YH, Wang ML. Associations of MRI-Derived Glymphatic System Impairment With Global White Matter Damage and Cognitive Impairment in Mild Traumatic Brain Injury: A DTI-ALPS Study. J Magn Reson Imaging 2024; 59:639-647. [PMID: 37276070 DOI: 10.1002/jmri.28797] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2023] [Revised: 05/08/2023] [Accepted: 05/09/2023] [Indexed: 06/07/2023] Open
Abstract
BACKGROUND Assessing the glymphatic function using diffusion tensor image analysis along the perivascular space (DTI-ALPS) may be helpful for mild traumatic brain injury (mTBI) management. PURPOSE To assess glymphatic function using DTI-ALPS and its associations with global white matter damage and cognitive impairment in mTBI. STUDY TYPE Prospective. POPULATION Thirty-four controls (44.1% female, mean age 49.2 years) and 58 mTBI subjects (43.1% female, mean age 48.7 years), including uncomplicated mTBI (N = 32) and complicated mTBI (N = 26). FIELD STRENGTH/SEQUENCE 3-T, single-shot echo-planar imaging sequence. ASSESSMENT Magnetic resonance imaging (MRI) was done within 1 month since injury. DTI-ALPS was performed to assess glymphatic function, and peak width of skeletonized mean diffusivity (PSMD) was used to assess global white matter damage. Cognitive tests included Auditory Verbal Learning Test and Digit Span Test (forward and backward). STATISTICAL TESTS Neuroimaging findings comparisons were done between mTBI and control groups. Partial correlation and multivariable linear regression assessed the associations between DTI-ALPS, PSMD, and cognitive impairment. Mediation effects of PSMD on the relationship between DTI-ALPS and cognitive impairment were explored. P-value <0.05 was considered statistically significant, except for cognitive correlational analyses with a Bonferroni-corrected P-value set at 0.05/3 ≈ 0.017. RESULTS mTBI showed lower DTI-ALPS and higher PSMD, especially in complicated mTBI. DTI-ALPS was significantly correlated with verbal memory (r = 0.566), attention abilities (r = 0.792), executive function (r = 0.618), and PSMD (r = -0.533). DTI-ALPS was associated with verbal memory (β = 8.77, 95% confidence interval [CI] 5.00, 12.54), attention abilities (β = 5.67, 95% CI 4.56, 6.97), executive function (β = 2.34, 95% CI 1.49, 3.20), and PSMD (β = -0.79, 95% CI -1.15, -0.43). PSMD mediated 46.29%, 20.46%, and 24.36% of the effects for the relationship between DTI-ALPS and verbal memory, attention abilities, and executive function. DATA CONCLUSION Glymphatic function may be impaired in mTBI reflected by DTI-ALPS. Glymphatic dysfunction may cause cognitive impairment related to global white matter damage after mTBI. LEVEL OF EVIDENCE 2 TECHNICAL EFFICACY: Stage 2.
Collapse
Affiliation(s)
- Dian-Xu Yang
- Department of Neurosurgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zheng Sun
- Department of Radiology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Meng-Meng Yu
- Department of Radiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Medical Imaging, Shanghai, China
| | - Qiao-Qiao Zou
- Department of Radiology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Peng-Yang Li
- Division of Cardiology, Pauley Heart Center, Virginia Commonwealth, University, Richmond, Virginia, USA
| | - Jing-Kun Zhang
- Cardiovascular Research Institute, University of California San Francisco, San Francisco, California, USA
| | - Xue Wu
- Institute for Global Health Sciences, University of California San Francisco, San Francisco, California, USA
| | - Yue-Hua Li
- Department of Radiology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ming-Liang Wang
- Department of Radiology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
12
|
Lizarraga A, Ripp I, Sala A, Shi K, Düring M, Koch K, Yakushev I. Similarity between structural and proxy estimates of brain connectivity. J Cereb Blood Flow Metab 2024; 44:284-295. [PMID: 37773727 PMCID: PMC10993877 DOI: 10.1177/0271678x231204769] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/15/2023] [Revised: 08/01/2023] [Accepted: 08/18/2023] [Indexed: 10/01/2023]
Abstract
Functional magnetic resonance and diffusion weighted imaging have so far made a major contribution to delineation of the brain connectome at the macroscale. While functional connectivity (FC) was shown to be related to structural connectivity (SC) to a certain degree, their spatial overlap is unknown. Even less clear are relations of SC with estimates of connectivity from inter-subject covariance of regional F18-fluorodeoxyglucose uptake (FDGcov) and grey matter volume (GMVcov). Here, we asked to what extent SC underlies three proxy estimates of brain connectivity: FC, FDGcov and GMVcov. Simultaneous PET/MR acquisitions were performed in 56 healthy middle-aged individuals. Similarity between four networks was assessed using Spearman correlation and convergence ratio (CR), a measure of spatial overlap. Spearman correlation coefficient was 0.27 for SC-FC, 0.40 for SC-FDGcov, and 0.15 for SC-GMVcov. Mean CRs were 51% for SC-FC, 48% for SC-FDGcov, and 37% for SC-GMVcov. These results proved to be reproducible and robust against image processing steps. In sum, we found a relevant similarity of SC with FC and FDGcov, while GMVcov consistently showed the weakest similarity. These findings indicate that white matter tracts underlie FDGcov to a similar degree as FC, supporting FDGcov as estimate of functional brain connectivity.
Collapse
Affiliation(s)
- Aldana Lizarraga
- Department of Nuclear Medicine, School of Medicine, Klinikum Rechts der Isar, Technical University of Munich, Munich, Germany
| | - Isabelle Ripp
- Department of Nuclear Medicine, School of Medicine, Klinikum Rechts der Isar, Technical University of Munich, Munich, Germany
| | - Arianna Sala
- Department of Nuclear Medicine, School of Medicine, Klinikum Rechts der Isar, Technical University of Munich, Munich, Germany
- Coma Science Group, GIGA Consciousness, University of Liege; Centre du Cerveau2, University Hospital of Liege, Avenue de L'Hôpital 1, Liege, Belgium
| | - Kuangyu Shi
- Department of Nuclear Medicine, University Hospital Bern, Bern, Switzerland
| | - Marco Düring
- Medical Image Analysis Center (MIAC AG) and Qbig, Department of Biomedical Engineering, University of Basel, Basel, Switzerland
| | - Kathrin Koch
- Department of Neuroradiology, School of Medicine, Klinikum Rechts der Isar, Technical University of Munich, Munich, Germany
| | - Igor Yakushev
- Department of Nuclear Medicine, School of Medicine, Klinikum Rechts der Isar, Technical University of Munich, Munich, Germany
| |
Collapse
|
13
|
Zhang L, Qu J, Ma H, Chen T, Liu T, Zhu D. Exploring Alzheimer's disease: a comprehensive brain connectome-based survey. PSYCHORADIOLOGY 2024; 4:kkad033. [PMID: 38333558 PMCID: PMC10848159 DOI: 10.1093/psyrad/kkad033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/28/2023] [Revised: 12/21/2023] [Accepted: 01/03/2024] [Indexed: 02/10/2024]
Abstract
Dementia is an escalating global health challenge, with Alzheimer's disease (AD) at its forefront. Substantial evidence highlights the accumulation of AD-related pathological proteins in specific brain regions and their subsequent dissemination throughout the broader area along the brain network, leading to disruptions in both individual brain regions and their interconnections. Although a comprehensive understanding of the neurodegeneration-brain network link is lacking, it is undeniable that brain networks play a pivotal role in the development and progression of AD. To thoroughly elucidate the intricate network of elements and connections constituting the human brain, the concept of the brain connectome was introduced. Research based on the connectome holds immense potential for revealing the mechanisms underlying disease development, and it has become a prominent topic that has attracted the attention of numerous researchers. In this review, we aim to systematically summarize studies on brain networks within the context of AD, critically analyze the strengths and weaknesses of existing methodologies, and offer novel perspectives and insights, intending to serve as inspiration for future research.
Collapse
Affiliation(s)
- Lu Zhang
- Department of Computer Science and Engineering, The University of Texas at Arlington, Arlington, TX 76019, USA
| | - Junqi Qu
- Department of Computer Science and Engineering, The University of Texas at Arlington, Arlington, TX 76019, USA
| | - Haotian Ma
- Department of Computer Science and Engineering, The University of Texas at Arlington, Arlington, TX 76019, USA
| | - Tong Chen
- Department of Computer Science and Engineering, The University of Texas at Arlington, Arlington, TX 76019, USA
| | - Tianming Liu
- Department of Computer Science, The University of Georgia, Athens, GA 30602, USA
| | - Dajiang Zhu
- Department of Computer Science and Engineering, The University of Texas at Arlington, Arlington, TX 76019, USA
| |
Collapse
|
14
|
Mattsson-Carlgren N, Collij LE, Stomrud E, Pichet Binette A, Ossenkoppele R, Smith R, Karlsson L, Lantero-Rodriguez J, Snellman A, Strandberg O, Palmqvist S, Ashton NJ, Blennow K, Janelidze S, Hansson O. Plasma Biomarker Strategy for Selecting Patients With Alzheimer Disease for Antiamyloid Immunotherapies. JAMA Neurol 2024; 81:69-78. [PMID: 38048096 PMCID: PMC10696515 DOI: 10.1001/jamaneurol.2023.4596] [Citation(s) in RCA: 23] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Accepted: 10/09/2023] [Indexed: 12/05/2023]
Abstract
Importance Antiamyloid immunotherapies against Alzheimer disease (AD) are emerging. Scalable, cost-effective tools will be needed to identify amyloid β (Aβ)-positive patients without an advanced stage of tau pathology who are most likely to benefit from these therapies. Blood-based biomarkers might reduce the need to use cerebrospinal fluid (CSF) or positron emission tomography (PET) for this. Objective To evaluate plasma biomarkers for identifying Aβ positivity and stage of tau accumulation. Design, Setting, and Participants The cohort study (BioFINDER-2) was a prospective memory-clinic and population-based study. Participants with cognitive concerns were recruited from 2017 to 2022 and divided into a training set (80% of the data) and test set (20%). Exposure Baseline values for plasma phosphorylated tau 181 (p-tau181), p-tau217, p-tau231, N-terminal tau, glial fibrillary acidic protein, and neurofilament light chain. Main Outcomes and Measures Performance to classify participants by Aβ status (defined by Aβ-PET or CSF Aβ42/40) and tau status (tau PET). Number of hypothetically saved PET scans in a plasma biomarker-guided workflow. Results Of a total 912 participants, there were 499 males (54.7%) and 413 females (45.3%), and the mean (SD) age was 71.1 (8.49) years. Among the biomarkers, plasma p-tau217 was most strongly associated with Aβ positivity (test-set area under the receiver operating characteristic curve [AUC] = 0.94; 95% CI, 0.90-0.97). A 2-cut-point procedure was evaluated, where only participants with ambiguous plasma p-tau217 values (17.1% of the participants in the test set) underwent CSF or PET to assign definitive Aβ status. This procedure had an overall sensitivity of 0.94 (95% CI, 0.90-0.98) and a specificity of 0.86 (95% CI, 0.77-0.95). Next, plasma biomarkers were used to differentiate low-intermediate vs high tau-PET load among Aβ-positive participants. Plasma p-tau217 again performed best, with the test AUC = 0.92 (95% CI, 0.86-0.97), without significant improvement when adding any of the other plasma biomarkers. At a false-negative rate less than 10%, the use of plasma p-tau217 could avoid 56.9% of tau-PET scans needed to identify high tau PET among Aβ-positive participants. The results were validated in an independent cohort (n = 118). Conclusions and Relevance This study found that algorithms using plasma p-tau217 can accurately identify most Aβ-positive individuals, including those likely to have a high tau load who would require confirmatory tau-PET imaging. Plasma p-tau217 measurements may substantially reduce the number of invasive and costly confirmatory tests required to identify individuals who would likely benefit from antiamyloid therapies.
Collapse
Affiliation(s)
- Niklas Mattsson-Carlgren
- Clinical Memory Research Unit, Clinical Sciences Malmö, Lund University, Lund, Sweden
- Department of Neurology, Skåne University Hospital, Lund University, Lund, Sweden
- Wallenberg Center for Molecular Medicine, Lund University, Lund, Sweden
| | - Lyduine E. Collij
- Clinical Memory Research Unit, Clinical Sciences Malmö, Lund University, Lund, Sweden
- Radiology and Nuclear Medicine, Amsterdam UMC location VUmc, Amsterdam, the Netherlands
- Amsterdam Neuroscience, Brain Imaging, Amsterdam, the Netherlands
| | - Erik Stomrud
- Clinical Memory Research Unit, Clinical Sciences Malmö, Lund University, Lund, Sweden
- Memory Clinic, Skåne University Hospital, Lund University, Lund, Sweden
| | - Alexa Pichet Binette
- Clinical Memory Research Unit, Clinical Sciences Malmö, Lund University, Lund, Sweden
| | - Rik Ossenkoppele
- Clinical Memory Research Unit, Clinical Sciences Malmö, Lund University, Lund, Sweden
- Alzheimer Center Amsterdam, Neurology, Amsterdam UMC location VUmc, Amsterdam, the Netherlands
- Amsterdam Neuroscience, Neurodegeneration, Amsterdam, the Netherlands
| | - Ruben Smith
- Clinical Memory Research Unit, Clinical Sciences Malmö, Lund University, Lund, Sweden
- Department of Neurology, Skåne University Hospital, Lund University, Lund, Sweden
| | - Linda Karlsson
- Clinical Memory Research Unit, Clinical Sciences Malmö, Lund University, Lund, Sweden
| | - Juan Lantero-Rodriguez
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Mölndal, Sweden
| | - Anniina Snellman
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Mölndal, Sweden
- Turku PET Centre, University of Turku, Turku University Hospital, Turku, Finland
| | - Olof Strandberg
- Clinical Memory Research Unit, Clinical Sciences Malmö, Lund University, Lund, Sweden
| | - Sebastian Palmqvist
- Clinical Memory Research Unit, Clinical Sciences Malmö, Lund University, Lund, Sweden
- Memory Clinic, Skåne University Hospital, Lund University, Lund, Sweden
| | - Nicholas J. Ashton
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Mölndal, Sweden
- King’s College London, Institute of Psychiatry, Psychology and Neuroscience, Maurice Wohl Institute Clinical Neuroscience Institute, London, United Kingdom
- NIHR Biomedical Research Centre for Mental Health and Biomedical Research Unit for Dementia at South London and Maudsley NHS Foundation, London, United Kingdom
- Centre for Age-Related Medicine, Stavanger University Hospital, Stavanger, Norway
| | - Kaj Blennow
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
| | - Shorena Janelidze
- Clinical Memory Research Unit, Clinical Sciences Malmö, Lund University, Lund, Sweden
| | - Oskar Hansson
- Clinical Memory Research Unit, Clinical Sciences Malmö, Lund University, Lund, Sweden
- Memory Clinic, Skåne University Hospital, Lund University, Lund, Sweden
| |
Collapse
|
15
|
Horvat A, Vlašić I, Štefulj J, Oršolić N, Jazvinšćak Jembrek M. Flavonols as a Potential Pharmacological Intervention for Alleviating Cognitive Decline in Diabetes: Evidence from Preclinical Studies. Life (Basel) 2023; 13:2291. [PMID: 38137892 PMCID: PMC10744738 DOI: 10.3390/life13122291] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2023] [Revised: 11/15/2023] [Accepted: 11/24/2023] [Indexed: 12/24/2023] Open
Abstract
Diabetes mellitus is a complex metabolic disease associated with reduced synaptic plasticity, atrophy of the hippocampus, and cognitive decline. Cognitive impairment results from several pathological mechanisms, including increased levels of advanced glycation end products (AGEs) and their receptors, prolonged oxidative stress and impaired activity of endogenous mechanisms of antioxidant defense, neuroinflammation driven by the nuclear factor kappa-light-chain enhancer of activated B cells (NF-κB), decreased expression of brain-derived neurotrophic factor (BDNF), and disturbance of signaling pathways involved in neuronal survival and cognitive functioning. There is increasing evidence that dietary interventions can reduce the risk of various diabetic complications. In this context, flavonols, a highly abundant class of flavonoids in the human diet, are appreciated as a potential pharmacological intervention against cognitive decline in diabetes. In preclinical studies, flavonols have shown neuroprotective, antioxidative, anti-inflammatory, and memory-enhancing properties based on their ability to regulate glucose levels, attenuate oxidative stress and inflammation, promote the expression of neurotrophic factors, and regulate signaling pathways. The present review gives an overview of the molecular mechanisms involved in diabetes-induced cognitive dysfunctions and the results of preclinical studies showing that flavonols have the ability to alleviate cognitive impairment. Although the results from animal studies are promising, clinical and epidemiological studies are still needed to advance our knowledge on the potential of flavonols to improve cognitive decline in diabetic patients.
Collapse
Affiliation(s)
- Anđela Horvat
- Division of Molecular Medicine, Ruđer Bošković Institute, Bijenička 54, 10000 Zagreb, Croatia
| | - Ignacija Vlašić
- Division of Molecular Medicine, Ruđer Bošković Institute, Bijenička 54, 10000 Zagreb, Croatia
| | - Jasminka Štefulj
- Division of Molecular Biology, Ruđer Bošković Institute, Bijenička 54, 10000 Zagreb, Croatia
- Department of Psychology, Catholic University of Croatia, Ilica 242, 10000 Zagreb, Croatia
| | - Nada Oršolić
- Division of Animal Physiology, Faculty of Science, University of Zagreb, Rooseveltov trg 6, 10000 Zagreb, Croatia
| | - Maja Jazvinšćak Jembrek
- Division of Molecular Medicine, Ruđer Bošković Institute, Bijenička 54, 10000 Zagreb, Croatia
- Department of Psychology, Catholic University of Croatia, Ilica 242, 10000 Zagreb, Croatia
| |
Collapse
|
16
|
Burnham SC, Iaccarino L, Pontecorvo MJ, Fleisher AS, Lu M, Collins EC, Devous MD. A review of the flortaucipir literature for positron emission tomography imaging of tau neurofibrillary tangles. Brain Commun 2023; 6:fcad305. [PMID: 38187878 PMCID: PMC10768888 DOI: 10.1093/braincomms/fcad305] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2023] [Revised: 09/13/2023] [Accepted: 11/14/2023] [Indexed: 01/09/2024] Open
Abstract
Alzheimer's disease is defined by the presence of β-amyloid plaques and neurofibrillary tau tangles potentially preceding clinical symptoms by many years. Previously only detectable post-mortem, these pathological hallmarks are now identifiable using biomarkers, permitting an in vivo definitive diagnosis of Alzheimer's disease. 18F-flortaucipir (previously known as 18F-T807; 18F-AV-1451) was the first tau positron emission tomography tracer to be introduced and is the only Food and Drug Administration-approved tau positron emission tomography tracer (Tauvid™). It has been widely adopted and validated in a number of independent research and clinical settings. In this review, we present an overview of the published literature on flortaucipir for positron emission tomography imaging of neurofibrillary tau tangles. We considered all accessible peer-reviewed literature pertaining to flortaucipir through 30 April 2022. We found 474 relevant peer-reviewed publications, which were organized into the following categories based on their primary focus: typical Alzheimer's disease, mild cognitive impairment and pre-symptomatic populations; atypical Alzheimer's disease; non-Alzheimer's disease neurodegenerative conditions; head-to-head comparisons with other Tau positron emission tomography tracers; and technical considerations. The available flortaucipir literature provides substantial evidence for the use of this positron emission tomography tracer in assessing neurofibrillary tau tangles in Alzheimer's disease and limited support for its use in other neurodegenerative disorders. Visual interpretation and quantitation approaches, although heterogeneous, mostly converge and demonstrate the high diagnostic and prognostic value of flortaucipir in Alzheimer's disease.
Collapse
Affiliation(s)
| | | | | | | | - Ming Lu
- Avid, Eli Lilly and Company, Philadelphia, PA 19104, USA
| | | | | |
Collapse
|
17
|
Vogel JW, Corriveau-Lecavalier N, Franzmeier N, Pereira JB, Brown JA, Maass A, Botha H, Seeley WW, Bassett DS, Jones DT, Ewers M. Connectome-based modelling of neurodegenerative diseases: towards precision medicine and mechanistic insight. Nat Rev Neurosci 2023; 24:620-639. [PMID: 37620599 DOI: 10.1038/s41583-023-00731-8] [Citation(s) in RCA: 35] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/26/2023] [Indexed: 08/26/2023]
Abstract
Neurodegenerative diseases are the most common cause of dementia. Although their underlying molecular pathologies have been identified, there is substantial heterogeneity in the patterns of progressive brain alterations across and within these diseases. Recent advances in neuroimaging methods have revealed that pathological proteins accumulate along specific macroscale brain networks, implicating the network architecture of the brain in the system-level pathophysiology of neurodegenerative diseases. However, the extent to which 'network-based neurodegeneration' applies across the wide range of neurodegenerative disorders remains unclear. Here, we discuss the state-of-the-art of neuroimaging-based connectomics for the mapping and prediction of neurodegenerative processes. We review findings supporting brain networks as passive conduits through which pathological proteins spread. As an alternative view, we also discuss complementary work suggesting that network alterations actively modulate the spreading of pathological proteins between connected brain regions. We conclude this Perspective by proposing an integrative framework in which connectome-based models can be advanced along three dimensions of innovation: incorporating parameters that modulate propagation behaviour on the basis of measurable biological features; building patient-tailored models that use individual-level information and allowing model parameters to interact dynamically over time. We discuss promises and pitfalls of these strategies for improving disease insights and moving towards precision medicine.
Collapse
Affiliation(s)
- Jacob W Vogel
- Department of Clinical Sciences, SciLifeLab, Lund University, Lund, Sweden.
| | - Nick Corriveau-Lecavalier
- Department of Neurology, Mayo Clinic, Rochester, MN, USA
- Department of Psychiatry and Psychology, Mayo Clinic, Rochester, MN, USA
| | - Nicolai Franzmeier
- Institute for Stroke and Dementia Research (ISD), University Hospital, LMU Munich, Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Acadamy, University of Gothenburg, Mölndal and Gothenburg, Sweden
| | - Joana B Pereira
- Clinical Memory Research Unit, Department of Clinical Sciences, Lund University, Malmö, Sweden
- Neuro Division, Department of Clinical Neurosciences, Karolinska Institute, Stockholm, Sweden
| | - Jesse A Brown
- Memory and Aging Center, Department of Neurology, University of California, San Francisco, CA, USA
| | - Anne Maass
- German Center for Neurodegenerative Diseases (DZNE), Magdeburg, Germany
| | - Hugo Botha
- Department of Neurology, Mayo Clinic, Rochester, MN, USA
| | - William W Seeley
- Memory and Aging Center, Department of Neurology, University of California, San Francisco, CA, USA
- Department of Pathology, University of California, San Francisco, CA, USA
| | - Dani S Bassett
- Departments of Bioengineering, Electrical and Systems Engineering, Physics and Astronomy, Neurology and Psychiatry, University of Pennsylvania, Philadelphia, PA, USA
- Santa Fe Institute, Santa Fe, NM, USA
| | - David T Jones
- Department of Neurology, Mayo Clinic, Rochester, MN, USA
- Department of Radiology, Mayo Clinic, Rochester, MN, USA
| | - Michael Ewers
- Institute for Stroke and Dementia Research (ISD), University Hospital, LMU Munich, Munich, Germany.
| |
Collapse
|
18
|
Han F, Lee J, Chen X, Ziontz J, Ward T, Landau SM, Baker SL, Harrison TM, Jagust WJ. Global brain activity and its coupling with cerebrospinal fluid flow is related to tau pathology. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.09.12.557492. [PMID: 37745434 PMCID: PMC10515801 DOI: 10.1101/2023.09.12.557492] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/26/2023]
Abstract
Amyloid-β (Aβ) and tau deposition constitute Alzheimer's disease (AD) neuropathology. Cortical tau deposits first in the entorhinal cortex and hippocampus and then propagates to neocortex in an Aβ-dependent manner. Tau also tends to accumulate earlier in higher-order association cortex than in lower-order primary sensory-motor cortex. While previous research has examined the production and spread of tau, little attention has been paid to its clearance. Low-frequency (<0.1 Hz) global brain activity during the resting state is coupled with cerebrospinal fluid (CSF) flow and potentially reflects glymphatic clearance. Here we report that tau deposition in subjects with evaluated Aβ, accompanied by cortical thinning and cognitive decline, is strongly associated with decreased coupling between CSF flow and global brain activity. Substantial modulation of global brain activity is also manifested as propagating waves of brain activation between higher- and lower-order regions, resembling tau spreading. Together, the findings suggest an important role of resting-state global brain activity in AD tau pathology.
Collapse
Affiliation(s)
- Feng Han
- Helen Wills Neuroscience Institute, University of California, Berkeley, Berkeley, CA, USA
| | - JiaQie Lee
- Helen Wills Neuroscience Institute, University of California, Berkeley, Berkeley, CA, USA
| | - Xi Chen
- Helen Wills Neuroscience Institute, University of California, Berkeley, Berkeley, CA, USA
- Lawrence Berkeley National Laboratory, Berkeley, CA, USA
| | - Jacob Ziontz
- Helen Wills Neuroscience Institute, University of California, Berkeley, Berkeley, CA, USA
| | - Tyler Ward
- Helen Wills Neuroscience Institute, University of California, Berkeley, Berkeley, CA, USA
| | - Susan M Landau
- Helen Wills Neuroscience Institute, University of California, Berkeley, Berkeley, CA, USA
| | | | - Theresa M Harrison
- Helen Wills Neuroscience Institute, University of California, Berkeley, Berkeley, CA, USA
| | - William J Jagust
- Helen Wills Neuroscience Institute, University of California, Berkeley, Berkeley, CA, USA
- Lawrence Berkeley National Laboratory, Berkeley, CA, USA
| |
Collapse
|
19
|
Mijalkov M, Veréb D, Canal-Garcia A, Hinault T, Volpe G, Pereira JB. Nonlinear changes in delayed functional network topology in Alzheimer's disease: relationship with amyloid and tau pathology. Alzheimers Res Ther 2023; 15:112. [PMID: 37328909 DOI: 10.1186/s13195-023-01252-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Accepted: 05/31/2023] [Indexed: 06/18/2023]
Abstract
BACKGROUND Alzheimer's disease is a neurodegenerative disorder associated with the abnormal deposition of pathological processes, such as amyloid-ß and tau, which produces nonlinear changes in the functional connectivity patterns between different brain regions across the Alzheimer's disease continuum. However, the mechanisms underlying these nonlinear changes remain largely unknown. Here, we address this question using a novel method based on temporal or delayed correlations and calculate new whole-brain functional networks to tackle these mechanisms. METHODS To assess our method, we evaluated 166 individuals from the ADNI database, including amyloid-beta negative and positive cognitively normal subjects, patients with mild cognitive impairment, and patients with Alzheimer's disease dementia. We used the clustering coefficient and the global efficiency to measure the functional network topology and assessed their relationship with amyloid and tau pathology measured by positron emission tomography, as well as cognitive performance using tests measuring memory, executive function, attention, and global cognition. RESULTS Our study found nonlinear changes in the global efficiency, but not in the clustering coefficient, showing that the nonlinear changes in functional connectivity are due to an altered ability of brain regions to communicate with each other through direct paths. These changes in global efficiency were most prominent in early disease stages. However, later stages of Alzheimer's disease were associated with widespread network disruptions characterized by changes in both network measures. The temporal delays required for the detection of these changes varied across the Alzheimer's disease continuum, with shorter delays necessary to detect changes in early stages and longer delays necessary to detect changes in late stages. Both global efficiency and clustering coefficient showed quadratic associations with pathological amyloid and tau burden as well as cognitive decline. CONCLUSIONS This study suggests that global efficiency is a more sensitive indicator of network changes in Alzheimer's disease when compared to clustering coefficient. Both network properties were associated with pathology and cognitive performance, demonstrating their relevance in clinical settings. Our findings provide an insight into the mechanisms underlying nonlinear changes in functional network organization in Alzheimer's disease, suggesting that it is the lack of direct connections that drives these functional changes.
Collapse
Affiliation(s)
- Mite Mijalkov
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden.
| | - Dániel Veréb
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Anna Canal-Garcia
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Thomas Hinault
- Normandie Univ, Unicaen, PSL, Université Paris, EPHE, Inserm, U1077, CHU de Caen, Centre Cyceron, 14000, Caen, France
| | - Giovanni Volpe
- Department of Physics, Goteborg University, Goteborg, Sweden
| | - Joana B Pereira
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden.
| |
Collapse
|
20
|
Tian J, Raghavan S, Reid RI, Przybelski SA, Lesnick TG, Gebre RK, Graff-Radford J, Schwarz CG, Lowe VJ, Kantarci K, Knopman DS, Petersen RC, Jack CR, Vemuri P. White Matter Degeneration Pathways Associated With Tau Deposition in Alzheimer Disease. Neurology 2023; 100:e2269-e2278. [PMID: 37068958 PMCID: PMC10259272 DOI: 10.1212/wnl.0000000000207250] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Accepted: 02/16/2023] [Indexed: 04/19/2023] Open
Abstract
BACKGROUND AND OBJECTIVES The dynamics of white matter (WM) changes are understudied in Alzheimer disease (AD). Our goal was to study the association between flortaucipir PET and WM health using neurite orientation dispersion and density imaging (NODDI) and evaluate its association with cognitive performance. Specifically, we focused on NODDI's Neurite Density Index (NDI), which aids in capturing axonal degeneration in WM and has greater specificity than single-shell diffusion MRI methods. METHOD We estimated regional flortaucipir PET standard uptake value ratios (SUVRs) from 3 regions corresponding to Braak stage I, III/IV, and V/VI to capture the spatial distribution pattern of the 3R/4R tau in AD. Then, we evaluated the associations between these measurements and NDIs in 29 candidate WM tracts using Pearson correlation and multiple regression models. RESULTS Based on 223 participants who were amyloid positive (mean age of 78 years and 57.0% male, 119 cognitively unimpaired, 56 mild cognitive impairment, and 48 dementia), the results showed that WM tracts NDI decreased with increasing regional Braak tau SUVRs. Of all the significant WM tracts, the uncinate fasciculus (r = -0.274 for Braak I, -0.311 for Braak III/IV, and -0.292 for Braak V/VI, p < 0.05) and cingulum adjoining hippocampus (r = -0.274, -0.288, -0.233, p < 0.05), both tracts anatomically connected to areas of early tau deposition, were consistently found to be within the top 5 distinguishing WM tracts associated with flortaucipir SUVRs. The increase in tau deposition measurable outside the medial temporal lobes in Braak III-VI was associated with a decrease in NDI in the middle and inferior temporal WM tracts. For cognitive performance, WM NDI had similar coefficients of determination (r 2 = 31%) as regional Braak flortaucipir SUVRs (29%), and together WM NDI and regional Braak flortaucipir SUVRs explained 46% of the variance in cognitive performance. DISCUSSION We found spatially dependent WM degeneration associated with regional flortaucipir SUVRs in Braak stages, suggesting a spatial pattern in WM damage. NDI, a specific marker of axonal density, provides complementary information about disease staging and progression in addition to tau deposition. Measurements of WM changes are important for the mechanistic understanding of multifactorial pathways through which AD causes cognitive dysfunction.
Collapse
Affiliation(s)
- Jianqiao Tian
- From the Department of Radiology (J.T., S.R., R.K.G., C.G.S., V.J.L., K.K., C.R.J., P.V.), Mayo Clinic; Mayo Clinic Graduate School of Biomedical Sciences (J.T.); and Department of Information Technology (R.I.R.), Department of Quantitative Health Sciences (S.A.P., T.G.L.), and Department of Neurology (J.G.-R., D.S.K., R.C.P.), Mayo Clinic, Rochester, MN
| | - Sheelakumari Raghavan
- From the Department of Radiology (J.T., S.R., R.K.G., C.G.S., V.J.L., K.K., C.R.J., P.V.), Mayo Clinic; Mayo Clinic Graduate School of Biomedical Sciences (J.T.); and Department of Information Technology (R.I.R.), Department of Quantitative Health Sciences (S.A.P., T.G.L.), and Department of Neurology (J.G.-R., D.S.K., R.C.P.), Mayo Clinic, Rochester, MN
| | - Robert I Reid
- From the Department of Radiology (J.T., S.R., R.K.G., C.G.S., V.J.L., K.K., C.R.J., P.V.), Mayo Clinic; Mayo Clinic Graduate School of Biomedical Sciences (J.T.); and Department of Information Technology (R.I.R.), Department of Quantitative Health Sciences (S.A.P., T.G.L.), and Department of Neurology (J.G.-R., D.S.K., R.C.P.), Mayo Clinic, Rochester, MN
| | - Scott A Przybelski
- From the Department of Radiology (J.T., S.R., R.K.G., C.G.S., V.J.L., K.K., C.R.J., P.V.), Mayo Clinic; Mayo Clinic Graduate School of Biomedical Sciences (J.T.); and Department of Information Technology (R.I.R.), Department of Quantitative Health Sciences (S.A.P., T.G.L.), and Department of Neurology (J.G.-R., D.S.K., R.C.P.), Mayo Clinic, Rochester, MN
| | - Timothy G Lesnick
- From the Department of Radiology (J.T., S.R., R.K.G., C.G.S., V.J.L., K.K., C.R.J., P.V.), Mayo Clinic; Mayo Clinic Graduate School of Biomedical Sciences (J.T.); and Department of Information Technology (R.I.R.), Department of Quantitative Health Sciences (S.A.P., T.G.L.), and Department of Neurology (J.G.-R., D.S.K., R.C.P.), Mayo Clinic, Rochester, MN
| | - Robel K Gebre
- From the Department of Radiology (J.T., S.R., R.K.G., C.G.S., V.J.L., K.K., C.R.J., P.V.), Mayo Clinic; Mayo Clinic Graduate School of Biomedical Sciences (J.T.); and Department of Information Technology (R.I.R.), Department of Quantitative Health Sciences (S.A.P., T.G.L.), and Department of Neurology (J.G.-R., D.S.K., R.C.P.), Mayo Clinic, Rochester, MN
| | - Jonathan Graff-Radford
- From the Department of Radiology (J.T., S.R., R.K.G., C.G.S., V.J.L., K.K., C.R.J., P.V.), Mayo Clinic; Mayo Clinic Graduate School of Biomedical Sciences (J.T.); and Department of Information Technology (R.I.R.), Department of Quantitative Health Sciences (S.A.P., T.G.L.), and Department of Neurology (J.G.-R., D.S.K., R.C.P.), Mayo Clinic, Rochester, MN
| | - Christopher G Schwarz
- From the Department of Radiology (J.T., S.R., R.K.G., C.G.S., V.J.L., K.K., C.R.J., P.V.), Mayo Clinic; Mayo Clinic Graduate School of Biomedical Sciences (J.T.); and Department of Information Technology (R.I.R.), Department of Quantitative Health Sciences (S.A.P., T.G.L.), and Department of Neurology (J.G.-R., D.S.K., R.C.P.), Mayo Clinic, Rochester, MN
| | - Val J Lowe
- From the Department of Radiology (J.T., S.R., R.K.G., C.G.S., V.J.L., K.K., C.R.J., P.V.), Mayo Clinic; Mayo Clinic Graduate School of Biomedical Sciences (J.T.); and Department of Information Technology (R.I.R.), Department of Quantitative Health Sciences (S.A.P., T.G.L.), and Department of Neurology (J.G.-R., D.S.K., R.C.P.), Mayo Clinic, Rochester, MN
| | - Kejal Kantarci
- From the Department of Radiology (J.T., S.R., R.K.G., C.G.S., V.J.L., K.K., C.R.J., P.V.), Mayo Clinic; Mayo Clinic Graduate School of Biomedical Sciences (J.T.); and Department of Information Technology (R.I.R.), Department of Quantitative Health Sciences (S.A.P., T.G.L.), and Department of Neurology (J.G.-R., D.S.K., R.C.P.), Mayo Clinic, Rochester, MN
| | - David S Knopman
- From the Department of Radiology (J.T., S.R., R.K.G., C.G.S., V.J.L., K.K., C.R.J., P.V.), Mayo Clinic; Mayo Clinic Graduate School of Biomedical Sciences (J.T.); and Department of Information Technology (R.I.R.), Department of Quantitative Health Sciences (S.A.P., T.G.L.), and Department of Neurology (J.G.-R., D.S.K., R.C.P.), Mayo Clinic, Rochester, MN
| | - Ronald C Petersen
- From the Department of Radiology (J.T., S.R., R.K.G., C.G.S., V.J.L., K.K., C.R.J., P.V.), Mayo Clinic; Mayo Clinic Graduate School of Biomedical Sciences (J.T.); and Department of Information Technology (R.I.R.), Department of Quantitative Health Sciences (S.A.P., T.G.L.), and Department of Neurology (J.G.-R., D.S.K., R.C.P.), Mayo Clinic, Rochester, MN
| | - Clifford R Jack
- From the Department of Radiology (J.T., S.R., R.K.G., C.G.S., V.J.L., K.K., C.R.J., P.V.), Mayo Clinic; Mayo Clinic Graduate School of Biomedical Sciences (J.T.); and Department of Information Technology (R.I.R.), Department of Quantitative Health Sciences (S.A.P., T.G.L.), and Department of Neurology (J.G.-R., D.S.K., R.C.P.), Mayo Clinic, Rochester, MN
| | - Prashanthi Vemuri
- From the Department of Radiology (J.T., S.R., R.K.G., C.G.S., V.J.L., K.K., C.R.J., P.V.), Mayo Clinic; Mayo Clinic Graduate School of Biomedical Sciences (J.T.); and Department of Information Technology (R.I.R.), Department of Quantitative Health Sciences (S.A.P., T.G.L.), and Department of Neurology (J.G.-R., D.S.K., R.C.P.), Mayo Clinic, Rochester, MN.
| |
Collapse
|
21
|
Veréb D, Mijalkov M, Chang YW, Canal-Garcia A, Gomez-Ruis E, Maass A, Villeneuve S, Volpe G, Pereira JB. Functional gradients of the medial parietal cortex in a healthy cohort with family history of sporadic Alzheimer's disease. Alzheimers Res Ther 2023; 15:82. [PMID: 37076873 PMCID: PMC10114342 DOI: 10.1186/s13195-023-01228-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2022] [Accepted: 04/05/2023] [Indexed: 04/21/2023]
Abstract
BACKGROUND The medial parietal cortex is an early site of pathological protein deposition in Alzheimer's disease (AD). Previous studies have identified different subregions within this area; however, these subregions are often heterogeneous and disregard individual differences or subtle pathological alterations in the underlying functional architecture. To address this limitation, here we measured the continuous connectivity gradients of the medial parietal cortex and assessed their relationship with cerebrospinal fluid (CSF) biomarkers, ApoE ε4 carriership and memory in asymptomatic individuals at risk to develop AD. METHODS Two hundred sixty-three cognitively normal participants with a family history of sporadic AD who underwent resting-state and task-based functional MRI using encoding and retrieval tasks were included from the PREVENT-AD cohort. A novel method for characterizing spatially continuous patterns of functional connectivity was applied to estimate functional gradients in the medial parietal cortex during the resting-state and task-based conditions. This resulted in a set of nine parameters that described the appearance of the gradient across different spatial directions. We performed correlation analyses to assess whether these parameters were associated with CSF biomarkers of phosphorylated tau181 (p-tau), total tau (t-tau), and amyloid-ß1-42 (Aß). Then, we compared the spatial parameters between ApoE ε4 carriers and noncarriers, and evaluated the relationship between these parameters and memory. RESULTS Alterations involving the superior part of the medial parietal cortex, which was connected to regions of the default mode network, were associated with higher p-tau, t-tau levels as well as lower Aß/p-tau levels during the resting-state condition (p < 0.01). Similar alterations were found in ApoE ε4 carriers compared to non-carriers (p < 0.003). In contrast, lower immediate memory scores were associated with changes in the middle part of the medial parietal cortex, which was connected to inferior temporal and posterior parietal regions, during the encoding task (p = 0.001). No results were found when using conventional connectivity measures. CONCLUSIONS Functional alterations in the medial parietal gradients are associated with CSF AD biomarkers, ApoE ε4 carriership, and lower memory in an asymptomatic cohort with a family history of sporadic AD, suggesting that functional gradients are sensitive to subtle changes associated with early AD stages.
Collapse
Affiliation(s)
- Dániel Veréb
- Department of Neurobiology, Care Sciences and Society, Division of Clinical Geriatrics, Karolinska Institutet, Stockholm, Sweden.
| | - Mite Mijalkov
- Department of Neurobiology, Care Sciences and Society, Division of Clinical Geriatrics, Karolinska Institutet, Stockholm, Sweden
| | - Yu-Wei Chang
- Department of Physics, Goteborg University, Goteborg, Sweden
| | - Anna Canal-Garcia
- Department of Neurobiology, Care Sciences and Society, Division of Clinical Geriatrics, Karolinska Institutet, Stockholm, Sweden
| | | | - Anne Maass
- German Center for Neurodegenerative Diseases (DZNE), 39120, Magdeburg, Germany
| | - Sylvia Villeneuve
- Douglas Mental Health University Institute, McGill University, Montreal, QC, Canada
- McConnell Brain Imaging Center, Montreal Neurological Institute, McGill University, Montreal, QC, Canada
| | - Giovanni Volpe
- Department of Physics, Goteborg University, Goteborg, Sweden
| | - Joana B Pereira
- Department of Neurobiology, Care Sciences and Society, Division of Clinical Geriatrics, Karolinska Institutet, Stockholm, Sweden.
- Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, Lund, Sweden.
| |
Collapse
|
22
|
Pini L, Salvalaggio A, Wennberg AM, Dimakou A, Matteoli M, Corbetta M. The pollutome-connectome axis: a putative mechanism to explain pollution effects on neurodegeneration. Ageing Res Rev 2023; 86:101867. [PMID: 36720351 DOI: 10.1016/j.arr.2023.101867] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Revised: 01/17/2023] [Accepted: 01/26/2023] [Indexed: 01/29/2023]
Abstract
The study of pollutant effects is extremely important to address the epochal challenges we are facing, where world populations are increasingly moving from rural to urban centers, revolutionizing our world into an urban world. These transformations will exacerbate pollution, thus highlighting the necessity to unravel its effect on human health. Epidemiological studies have reported that pollution increases the risk of neurological diseases, with growing evidence on the risk of neurodegenerative disorders. Air pollution and water pollutants are the main chemicals driving this risk. These chemicals can promote inflammation, acting in synergy with genotype vulnerability. However, the biological underpinnings of this association are unknown. In this review, we focus on the link between pollution and brain network connectivity at the macro-scale level. We provide an updated overview of epidemiological findings and studies investigating brain network changes associated with pollution exposure, and discuss the mechanistic insights of pollution-induced brain changes through neural networks. We explain, in detail, the pollutome-connectome axis that might provide the functional substrate for pollution-induced processes leading to cognitive impairment and neurodegeneration. We describe this model within the framework of two pollutants, air pollution, a widely recognized threat, and polyfluoroalkyl substances, a large class of synthetic chemicals which are currently emerging as new neurotoxic source.
Collapse
Affiliation(s)
- Lorenzo Pini
- Department of Neuroscience and Padova Neuroscience Center, University of Padova, Italy; Venetian Institute of Molecular Medicine, VIMM, Padova, Italy.
| | | | - Alexandra M Wennberg
- Unit of Epidemiology, Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Anastasia Dimakou
- Department of Neuroscience and Padova Neuroscience Center, University of Padova, Italy
| | - Michela Matteoli
- Neuro Center, IRCCS Humanitas Research Hospital, via Manzoni 56, 20089 Rozzano, Milano, Italy; CNR Institute of Neuroscience, Milano, Italy
| | - Maurizio Corbetta
- Department of Neuroscience and Padova Neuroscience Center, University of Padova, Italy; Venetian Institute of Molecular Medicine, VIMM, Padova, Italy
| |
Collapse
|
23
|
Sala A, Lizarraga A, Caminiti SP, Calhoun VD, Eickhoff SB, Habeck C, Jamadar SD, Perani D, Pereira JB, Veronese M, Yakushev I. Brain connectomics: time for a molecular imaging perspective? Trends Cogn Sci 2023; 27:353-366. [PMID: 36621368 PMCID: PMC10432882 DOI: 10.1016/j.tics.2022.11.015] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Revised: 11/19/2022] [Accepted: 11/30/2022] [Indexed: 01/09/2023]
Abstract
In the past two decades brain connectomics has evolved into a major concept in neuroscience. However, the current perspective on brain connectivity and how it underpins brain function relies mainly on the hemodynamic signal of functional magnetic resonance imaging (MRI). Molecular imaging provides unique information inaccessible to MRI-based and electrophysiological techniques. Thus, positron emission tomography (PET) has been successfully applied to measure neural activity, neurotransmission, and proteinopathies in normal and pathological cognition. Here, we position molecular imaging within the brain connectivity framework from the perspective of timeliness, validity, reproducibility, and resolution. We encourage the neuroscientific community to take an integrative approach whereby MRI-based, electrophysiological techniques, and molecular imaging contribute to our understanding of the brain connectome.
Collapse
Affiliation(s)
- Arianna Sala
- Department of Nuclear Medicine, Klinikum Rechts der Isar, Technical University of Munich, School of Medicine, 81675 Munich, Germany; Coma Science Group, GIGA-Consciousness, University of Liege, 4000 Liege, Belgium; Centre du Cerveau(2), University Hospital of Liege, 4000 Liege, Belgium
| | - Aldana Lizarraga
- Department of Nuclear Medicine, Klinikum Rechts der Isar, Technical University of Munich, School of Medicine, 81675 Munich, Germany
| | - Silvia Paola Caminiti
- Vita-Salute San Raffaele University, 20132 Milan, Italy; In Vivo Human Molecular and Structural Neuroimaging Unit, Division of Neuroscience, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Vince D Calhoun
- Tri-Institutional Center for Translational Research in Neuroimaging and Data Science (TReNDS), Georgia State University, Georgia Institute of Technology, and Emory University, Atlanta, GA 30303, USA
| | - Simon B Eickhoff
- Institute of Neuroscience and Medicine, Brain, and Behaviour (INM-7), Research Centre Jülich, 52428 Jülich, Germany; Institute of Systems Neuroscience, Medical Faculty, Heinrich-Heine-University Düsseldorf, 40225 Düsseldorf, Germany
| | - Christian Habeck
- Cognitive Neuroscience Division, Department of Neurology, Columbia University, New York, NY 10032, USA
| | - Sharna D Jamadar
- Turner Institute for Brain and Mental Health, Monash University, 3800 Melbourne, Australia; Monash Biomedical Imaging, Monash University, 3800 Melbourne, Australia
| | - Daniela Perani
- Vita-Salute San Raffaele University, 20132 Milan, Italy; In Vivo Human Molecular and Structural Neuroimaging Unit, Division of Neuroscience, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) San Raffaele Scientific Institute, 20132 Milan, Italy; Nuclear Medicine Unit, San Raffaele Hospital, 20132 Milan, Italy
| | - Joana B Pereira
- Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, 14152 Stockholm, Sweden; Memory Research Unit, Department of Clinical Sciences, Malmö Lund University, 20502 Lund, Sweden
| | - Mattia Veronese
- Department of Neuroimaging, King's College London, London SE5 8AF, UK; Department of Information Engineering, University of Padua, 35131 Padua, Italy
| | - Igor Yakushev
- Department of Nuclear Medicine, Klinikum Rechts der Isar, Technical University of Munich, School of Medicine, 81675 Munich, Germany.
| |
Collapse
|
24
|
Lipoprotein Metabolism, Protein Aggregation, and Alzheimer's Disease: A Literature Review. Int J Mol Sci 2023; 24:ijms24032944. [PMID: 36769268 PMCID: PMC9918279 DOI: 10.3390/ijms24032944] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2022] [Revised: 01/28/2023] [Accepted: 01/30/2023] [Indexed: 02/05/2023] Open
Abstract
Alzheimer's disease (AD) is the most common form of dementia. The physiopathology of AD is well described by the presence of two neuropathological features: amyloid plaques and tau neurofibrillary tangles. In the last decade, neuroinflammation and cellular stress have gained importance as key factors in the development and pathology of AD. Chronic cellular stress occurs in degenerating neurons. Stress Granules (SGs) are nonmembranous organelles formed as a response to stress, with a protective role; however, SGs have been noted to turn into pathological and neurotoxic features when stress is chronic, and they are related to an increased tau aggregation. On the other hand, correct lipid metabolism is essential to good function of the brain; apolipoproteins are highly associated with risk of AD, and impaired cholesterol efflux and lipid transport are associated with an increased risk of AD. In this review, we provide an insight into the relationship between cellular stress, SGs, protein aggregation, and lipid metabolism in AD.
Collapse
|
25
|
Green ZD, Vidoni ED, Swerdlow RH, Burns JM, Morris JK, Honea RA. Increased Functional Connectivity of the Precuneus in Individuals with a Family History of Alzheimer's Disease. J Alzheimers Dis 2023; 91:559-571. [PMID: 36463439 PMCID: PMC9912732 DOI: 10.3233/jad-210326] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
BACKGROUND First-degree relatives of individuals with late-onset Alzheimer's disease (AD) have increased risk for AD, with children of affected parents at an especially high risk. OBJECTIVE We aimed to investigate default mode network connectivity, medial temporal cortex volume, and cognition in cognitively healthy (CH) individuals with (FH+) and without (FH-) a family history of AD, alongside amnestic mild cognitive impairment (aMCI) and AD individuals, to determine the context and directionality of dysfunction in at-risk individuals. Our primary hypothesis was that there would be a linear decline (CH FH- > CH FH+ > aMCI > AD) within the risk groups on all measures of AD risk. METHODS We used MRI and fMRI to study cognitively healthy individuals (n = 28) with and without AD family history (FH+ and FH-, respectively), those with aMCI (n = 31) and early-stage AD (n = 25). We tested connectivity within the default mode network, as well as measures of volume and thickness within the medial temporal cortex and selected seed regions. RESULTS As expected, we identified decreased medial temporal cortex volumes in the aMCI and AD groups compared to cognitively healthy groups. We also observed patterns of connectivity across risk groups that suggest a nonlinear relationship of change, such that the FH+ group showed increased connectivity compared to the FH- and AD groups (CH FH+ > CH FH- > aMCI > AD). This pattern emerged primarily in connectivity between the precuneus and frontal regions. CONCLUSION These results add to a growing literature that suggests compensatory brain function in otherwise cognitively healthy individuals with a family history of AD.
Collapse
Affiliation(s)
- Zachary D. Green
- University of Kansas Alzheimer’s Disease Research Center, University of Kansas School of Medicine, Kansas City, KS, USA,
Department of Neurology, University of Kansas School of Medicine, Kansas City, KS, USA
| | - Eric D. Vidoni
- University of Kansas Alzheimer’s Disease Research Center, University of Kansas School of Medicine, Kansas City, KS, USA,
Department of Neurology, University of Kansas School of Medicine, Kansas City, KS, USA
| | - Russell H. Swerdlow
- University of Kansas Alzheimer’s Disease Research Center, University of Kansas School of Medicine, Kansas City, KS, USA,
Department of Neurology, University of Kansas School of Medicine, Kansas City, KS, USA
| | - Jeffrey M. Burns
- University of Kansas Alzheimer’s Disease Research Center, University of Kansas School of Medicine, Kansas City, KS, USA,
Department of Neurology, University of Kansas School of Medicine, Kansas City, KS, USA
| | - Jill K. Morris
- University of Kansas Alzheimer’s Disease Research Center, University of Kansas School of Medicine, Kansas City, KS, USA,
Department of Neurology, University of Kansas School of Medicine, Kansas City, KS, USA
| | - Robyn A. Honea
- University of Kansas Alzheimer’s Disease Research Center, University of Kansas School of Medicine, Kansas City, KS, USA,
Department of Neurology, University of Kansas School of Medicine, Kansas City, KS, USA,Correspondence to: Robyn A. Honea, University of Kansas School of Medicine, Department of Neurology, University of Kansas Alzheimer’s Disease Research Center, 4350 Shawnee Mission Parkway, Fairway, KS, 66205, USA. Tel.: +1 913 588 5514; E-mail:
| |
Collapse
|
26
|
Wang WE, Chen R, Mayrand RP, Adjouadi M, Fang R, DeKosky ST, Duara R, Coombes SA, Vaillancourt DE. Association of Longitudinal Cognitive Decline with Diffusion MRI in Gray Matter, Amyloid, and Tau Deposition. Neurobiol Aging 2022; 121:166-178. [DOI: 10.1016/j.neurobiolaging.2022.10.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Revised: 10/21/2022] [Accepted: 10/25/2022] [Indexed: 11/06/2022]
|
27
|
Xiao D, Wang K, Theriault L, Charbel E. White matter integrity and key structures affected in Alzheimer's disease characterized by diffusion tensor imaging. Eur J Neurosci 2022; 56:5319-5331. [PMID: 36048971 DOI: 10.1111/ejn.15815] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Revised: 08/13/2022] [Accepted: 08/22/2022] [Indexed: 12/14/2022]
Abstract
White matter (WM) degeneration is suggested to predict the early signs of Alzheimer's disease (AD). The exact structural regions of brain circuitry involved are not known. This study aims to examine the associations between WM tract integrity, represented by the diffusion tensor imaging (DTI) measures, and AD diagnosis and to denote the key substrates in predicting AD. It included DTI measures of mean diffusivity (MD), fractional anisotropy, radial diffusivity and axial diffusivity of 18 main WM tracts in 84 non-Hispanic white participants from the Alzheimer's Disease Neuroimaging Initiative dataset. The multivariable general linear model was used to examine the association of AD diagnosis with each DTI measure adjusting for age, gender and education. The corpus callosum, fornix, cingulum hippocampus, uncinate fasciculus, sagittal striatum, left posterior thalamic radiation and fornix-stria terminalis showed significant increases in MD, radial and axial diffusivity, whereas the splenium of corpus callosum and the fornix showed significant decreases in fractional anisotropy among AD patients. Variable cluster analysis identified that hippocampus volume, mini-mental state examination (MMSE), cingulate gyrus/hippocampus, inferior fronto-occipital fasciculus and uncinate fasciculus are highly correlated in one cluster with MD measures. In conclusion, there were significant differences in DTI measures between the brain WM of AD patients and controls. Age is the risk factor associated with AD, not gender or education. Right cingulum gyrus and right uncinate fasciculus are particularly affected, correlating well with a cognitive test MMSE and MD measures for dementia in AD patients and could be a region of focus for AD staging.
Collapse
Affiliation(s)
- Danqing Xiao
- Department of STEM, School of Arts and Sciences, Regis College, Weston, Massachusetts, USA.,Neuroimaging Center, McLean Hospital, Belmont, Massachusetts, USA
| | - Kesheng Wang
- Department of Family and Community Health, School of Nursing, Health Sciences Center, West Virginia University, Morgantown, West Virginia, USA
| | - Luke Theriault
- Department of STEM, School of Arts and Sciences, Regis College, Weston, Massachusetts, USA.,School of Medicine, St. George's University, Saint George's, Grenada
| | - Elhelou Charbel
- Department of STEM, School of Arts and Sciences, Regis College, Weston, Massachusetts, USA
| | | |
Collapse
|
28
|
Zhao H, Wen W, Cheng J, Jiang J, Kochan N, Niu H, Brodaty H, Sachdev P, Liu T. An accelerated degeneration of white matter microstructure and networks in the nondemented old-old. Cereb Cortex 2022; 33:4688-4698. [PMID: 36178117 DOI: 10.1093/cercor/bhac372] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Revised: 08/26/2022] [Accepted: 08/27/2022] [Indexed: 11/12/2022] Open
Abstract
The nondemented old-old over the age of 80 comprise a rapidly increasing population group; they can be regarded as exemplars of successful aging. However, our current understanding of successful aging in advanced age and its neural underpinnings is limited. In this study, we measured the microstructural and network-based topological properties of brain white matter using diffusion-weighted imaging scans of 419 community-dwelling nondemented older participants. The participants were further divided into 230 young-old (between 72 and 79, mean = 76.25 ± 2.00) and 219 old-old (between 80 and 92, mean = 83.98 ± 2.97). Results showed that white matter connectivity in microstructure and brain networks significantly declined with increased age and that the declined rates were faster in the old-old compared with young-old. Mediation models indicated that cognitive decline was in part through the age effect on the white matter connectivity in the old-old but not in the young-old. Machine learning predictive models further supported the crucial role of declines in white matter connectivity as a neural substrate of cognitive aging in the nondemented older population. Our findings shed new light on white matter connectivity in the nondemented aging brains and may contribute to uncovering the neural substrates of successful brain aging.
Collapse
Affiliation(s)
- Haichao Zhao
- Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing, China
| | - Wei Wen
- Centre for Healthy Brain Ageing, School of Psychiatry (CHeBA), University of New South Wales, Sydney, NSW, Australia.,Neuropsychiatric Institute, Prince of Wales Hospital, Sydney, NSW, Australia
| | - Jian Cheng
- School of Computer Science and Engineering, Beihang University, Beijing, China
| | - Jiyang Jiang
- Centre for Healthy Brain Ageing, School of Psychiatry (CHeBA), University of New South Wales, Sydney, NSW, Australia
| | - Nicole Kochan
- Centre for Healthy Brain Ageing, School of Psychiatry (CHeBA), University of New South Wales, Sydney, NSW, Australia.,Neuropsychiatric Institute, Prince of Wales Hospital, Sydney, NSW, Australia
| | - Haijun Niu
- Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing, China
| | - Henry Brodaty
- Centre for Healthy Brain Ageing, School of Psychiatry (CHeBA), University of New South Wales, Sydney, NSW, Australia.,Neuropsychiatric Institute, Prince of Wales Hospital, Sydney, NSW, Australia
| | - Perminder Sachdev
- Centre for Healthy Brain Ageing, School of Psychiatry (CHeBA), University of New South Wales, Sydney, NSW, Australia.,Neuropsychiatric Institute, Prince of Wales Hospital, Sydney, NSW, Australia
| | - Tao Liu
- Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing, China
| |
Collapse
|
29
|
Rauchmann B, Brendel M, Franzmeier N, Trappmann L, Zaganjori M, Ersoezlue E, Morenas‐Rodriguez E, Guersel S, Burow L, Kurz C, Haeckert J, Tatò M, Utecht J, Papazov B, Pogarell O, Janowitz D, Buerger K, Ewers M, Palleis C, Weidinger E, Biechele G, Schuster S, Finze A, Eckenweber F, Rupprecht R, Rominger A, Goldhardt O, Grimmer T, Keeser D, Stoecklein S, Dietrich O, Bartenstein P, Levin J, Höglinger G, Perneczky R. Microglial activation and connectivity in Alzheimer's disease and aging. Ann Neurol 2022; 92:768-781. [DOI: 10.1002/ana.26465] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Revised: 07/27/2022] [Accepted: 07/28/2022] [Indexed: 11/06/2022]
Affiliation(s)
- Boris‐Stephan Rauchmann
- Department of Psychiatry and Psychotherapy University Hospital, LMU Munich Munich Germany
- German Center for Neurodegenerative Diseases (DZNE) Munich Munich Germany
- Sheffield Institute for Translational Neuroscience (SITraN) University of Sheffield Sheffield UK
- Department of Neuroradiology University Hospital LMU Munich Germany
| | - Matthias Brendel
- Department of Nuclear Medicine University Hospital, LMU Munich Munich Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich Germany
| | - Nicolai Franzmeier
- Institute for Stroke and Dementia Research, University Hospital, LMU Munich Munich Germany
| | - Lena Trappmann
- Department of Psychiatry and Psychotherapy University Hospital, LMU Munich Munich Germany
| | - Mirlind Zaganjori
- Department of Psychiatry and Psychotherapy University Hospital, LMU Munich Munich Germany
| | - Ersin Ersoezlue
- Department of Psychiatry and Psychotherapy University Hospital, LMU Munich Munich Germany
| | - Estrella Morenas‐Rodriguez
- German Center for Neurodegenerative Diseases (DZNE) Munich Munich Germany
- Chair of Metabolic Biochemistry, Biomedical Center (BMC), Faculty of Medicine, LMU Munich Munich Germany
| | - Selim Guersel
- Department of Psychiatry and Psychotherapy University Hospital, LMU Munich Munich Germany
- German Center for Neurodegenerative Diseases (DZNE) Munich Munich Germany
| | - Lena Burow
- Department of Psychiatry and Psychotherapy University Hospital, LMU Munich Munich Germany
| | - Carolin Kurz
- Department of Psychiatry and Psychotherapy University Hospital, LMU Munich Munich Germany
| | - Jan Haeckert
- Department of Psychiatry and Psychotherapy University Hospital, LMU Munich Munich Germany
- Department of Psychiatry, Psychotherapy and Psychosomatics University of Augsburg, Bezirkskrankenhaus Augsburg Augsburg Germany
| | - Maia Tatò
- Department of Psychiatry and Psychotherapy University Hospital, LMU Munich Munich Germany
| | - Julia Utecht
- Department of Psychiatry and Psychotherapy University Hospital, LMU Munich Munich Germany
| | - Boris Papazov
- Department of Radiology University Hospital, LMU Munich Munich Germany
| | - Oliver Pogarell
- Department of Psychiatry and Psychotherapy University Hospital, LMU Munich Munich Germany
| | - Daniel Janowitz
- Institute for Stroke and Dementia Research, University Hospital, LMU Munich Munich Germany
| | - Katharina Buerger
- Institute for Stroke and Dementia Research, University Hospital, LMU Munich Munich Germany
- German Center for Neurodegenerative Diseases (DZNE) Munich Munich Germany
| | - Michael Ewers
- Institute for Stroke and Dementia Research, University Hospital, LMU Munich Munich Germany
| | - Carla Palleis
- German Center for Neurodegenerative Diseases (DZNE) Munich Munich Germany
- Department of Neurology University Hospital, LMU Munich Munich Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich Germany
| | - Endy Weidinger
- Department of Neurology University Hospital, LMU Munich Munich Germany
| | - Gloria Biechele
- Department of Nuclear Medicine University Hospital, LMU Munich Munich Germany
| | - Sebastian Schuster
- Department of Nuclear Medicine University Hospital, LMU Munich Munich Germany
| | - Anika Finze
- Department of Nuclear Medicine University Hospital, LMU Munich Munich Germany
| | - Florian Eckenweber
- Department of Nuclear Medicine University Hospital, LMU Munich Munich Germany
| | - Rainer Rupprecht
- Department of Psychiatry and Psychotherapy University of Regensburg Regensburg Germany
| | - Axel Rominger
- Department of Nuclear Medicine University Hospital, LMU Munich Munich Germany
- Department of Nuclear Medicine University of Bern, Inselspital Bern Switzerland
| | - Oliver Goldhardt
- Department of Psychiatry and Psychotherapy, Klinikum rechts der Isar Technical University Munich Munich Germany
| | - Timo Grimmer
- Department of Psychiatry and Psychotherapy, Klinikum rechts der Isar Technical University Munich Munich Germany
| | - Daniel Keeser
- Department of Radiology University Hospital, LMU Munich Munich Germany
- Department of Psychiatry and Psychotherapy University Hospital, LMU Munich Munich Germany
- Department of Neuroradiology University Hospital LMU Munich Germany
| | - Sophia Stoecklein
- Department of Radiology University Hospital, LMU Munich Munich Germany
| | - Olaf Dietrich
- Department of Radiology University Hospital, LMU Munich Munich Germany
| | - Peter Bartenstein
- Department of Nuclear Medicine University Hospital, LMU Munich Munich Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich Germany
| | - Johannes Levin
- German Center for Neurodegenerative Diseases (DZNE) Munich Munich Germany
- Department of Neurology University Hospital, LMU Munich Munich Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich Germany
| | - Günter Höglinger
- German Center for Neurodegenerative Diseases (DZNE) Munich Munich Germany
- Department of Neurology Hannover Medical School Hannover Germany
| | - Robert Perneczky
- Department of Psychiatry and Psychotherapy University Hospital, LMU Munich Munich Germany
- German Center for Neurodegenerative Diseases (DZNE) Munich Munich Germany
- Ageing Epidemiology (AGE) Research Unit, School of Public Health Imperial College London London UK
- Munich Cluster for Systems Neurology (SyNergy), Munich Germany
- Sheffield Institute for Translational Neuroscience (SITraN) University of Sheffield Sheffield UK
| |
Collapse
|
30
|
Kim HJ, Jo S, Lee S, Oh M, Lee JH. Crossed Hemispheric Accumulation of β-Amyloid and Tau Protein in a Patient With Typical Alzheimer Disease. Alzheimer Dis Assoc Disord 2022; 36:263-265. [PMID: 34132670 DOI: 10.1097/wad.0000000000000460] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Accepted: 04/20/2021] [Indexed: 11/26/2022]
Abstract
Amyloid (Aβ) and tau proteins are pathologic hallmarks of Alzheimer disease (AD). It is well known that there is spatial disparity between Aβ and tau protein deposition but, crossed hemispheric accumulation of these 2 proteins has not been reported. Here we report the case of a 76-year-old woman with typical AD who underwent amyloid positron emission tomography (PET) ([ 18 F]-florbetaben) and tau PET scans ([ 18 F]PI-2620), revealing crossed accumulation of Aβ and tau in the cerebral hemisphere. A neuropsychological assessment showed impairment in memory with spared activities of daily living. In the PET analysis, amyloid deposition was observed only in the left side of the cerebral hemisphere and tau only in the right side. Neuroimaging follow-up indicated that the spatial pattern of these protein accumulations had not changed. This case suggests the possibility of independent Aβ and tau pathogenic pathways in AD.
Collapse
Affiliation(s)
| | | | | | - Minyoung Oh
- Nuclear Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | | |
Collapse
|
31
|
Wendimu MY, Hooks SB. Microglia Phenotypes in Aging and Neurodegenerative Diseases. Cells 2022; 11:2091. [PMID: 35805174 PMCID: PMC9266143 DOI: 10.3390/cells11132091] [Citation(s) in RCA: 126] [Impact Index Per Article: 42.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2022] [Revised: 06/24/2022] [Accepted: 06/29/2022] [Indexed: 02/08/2023] Open
Abstract
Neuroinflammation is a hallmark of many neurodegenerative diseases (NDs) and plays a fundamental role in mediating the onset and progression of disease. Microglia, which function as first-line immune guardians of the central nervous system (CNS), are the central drivers of neuroinflammation. Numerous human postmortem studies and in vivo imaging analyses have shown chronically activated microglia in patients with various acute and chronic neuropathological diseases. While microglial activation is a common feature of many NDs, the exact role of microglia in various pathological states is complex and often contradictory. However, there is a consensus that microglia play a biphasic role in pathological conditions, with detrimental and protective phenotypes, and the overall response of microglia and the activation of different phenotypes depends on the nature and duration of the inflammatory insult, as well as the stage of disease development. This review provides a comprehensive overview of current research on the various microglia phenotypes and inflammatory responses in health, aging, and NDs, with a special emphasis on the heterogeneous phenotypic response of microglia in acute and chronic diseases such as hemorrhagic stroke (HS), Alzheimer's disease (AD), and Parkinson's disease (PD). The primary focus is translational research in preclinical animal models and bulk/single-cell transcriptome studies in human postmortem samples. Additionally, this review covers key microglial receptors and signaling pathways that are potential therapeutic targets to regulate microglial inflammatory responses during aging and in NDs. Additionally, age-, sex-, and species-specific microglial differences will be briefly reviewed.
Collapse
Affiliation(s)
| | - Shelley B. Hooks
- Hooks Lab, Department of Pharmaceutical and Biomedical Sciences, College of Pharmacy, University of Georgia, Athens, GA 30602, USA;
| |
Collapse
|
32
|
Manukyan AL. Noise as a cause of neurodegenerative disorders: molecular and cellular mechanisms. Neurol Sci 2022; 43:2983-2993. [PMID: 35166975 DOI: 10.1007/s10072-022-05948-6] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Accepted: 02/08/2022] [Indexed: 12/21/2022]
Abstract
Noise as an environmental stressor becomes of increasing importance in our industrialized world, and especially traffic noise from the environment represents a potential novel neurodegenerative risk factor, as well as for hearing loss. A significant number of studies have been suggested that the overproduction of reactive oxygen species (ROS) has a complex role in stimulation of pathologic events. Experimental studies upon molecular pathways of traffic noise exposure proposed that it increased the level of stress hormones and mediated the inflammatory and oxidative stress (OS) pathways resulting in endothelial and neuronal dysfunction. Studies have shown that neurons are especially sensitive to OS due to high polyunsaturated fatty acids content in membranes, high oxygen uptake, and weak antioxidant defense. However, OS induces the necrotic and apoptotic cell deaths in the cochlea. Chronic noise is one of the many overall reasons of obtained sensorineural hearing loss which destroys cognitive functions in human and animals, as well as suppresses neurogenesis in the hippocampus. Nevertheless, behavioral disorders caused by noise are mainly accompanied with oxidative stress, but the clear molecular mechanism of neurodegeneration due to disruption of the pro- and antioxidant systems is still not fully understood. This paper aims to highlight the down-stream pathophysiology of noise-induced mental disorders, including hearing loss, annoyance, anxiety, depression, memory loss, and Alzheimer's disease, describing the underlying mechanisms of induction of inflammation and oxidative stress.
Collapse
Affiliation(s)
- Ashkhen L Manukyan
- Department of Medical Chemistry, Yerevan State Medical University after M. Heratsi, Koryun 2, 0025, Yerevan, Armenia.
| |
Collapse
|
33
|
Giliberto L. Editorial: Degenerative and cognitive diseases. Curr Opin Neurol 2022; 35:208-211. [PMID: 35232933 DOI: 10.1097/wco.0000000000001037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Affiliation(s)
- Luca Giliberto
- Litwin-Zucker Center for the Study of Alzheimer's Diseases and Memory Disorders, Feinstein Institutes for Medical Research and Institute for Neurology and Neurosurgery, Northwell Health System, Manhasset, New York, USA
| |
Collapse
|
34
|
Karvelas N, Bennett S, Politis G, Kouris NI, Kole C. Advances in stem cell therapy in Alzheimer's disease: a comprehensive clinical trial review. Stem Cell Investig 2022; 9:2. [PMID: 35280344 PMCID: PMC8898169 DOI: 10.21037/sci-2021-063] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2021] [Accepted: 01/27/2022] [Indexed: 07/30/2023]
Abstract
Alzheimer's disease (AD) is the most common type of dementia responsible for more than 121,499 deaths from AD in 2019 making AD the sixth-leading cause in the United States. AD is a progressive neurodegenerative disorder characterized by decline of memory, behavioral impairments that affects a person's ability to function independently ultimately leading to death. The current pressing need for a treatment for (AD) and advances in the field of cell therapy, has rendered stem cell therapeutics a promising field of research. Despite advancements in stem cell technology, confirmed by encouraging pre-clinical utilization of stem cells in AD animal models, the number of clinical trials evaluating the efficacy of stem cell therapy is limited, with the results of many ongoing clinical trials on cell therapy for AD still pending. Mesenchymal stem cells (MSCs) have been the main focus in these studies, reporting encouraging results concerning safety profile, however their efficacy remains unproven. In the current article we review the latest advances regarding different sources of stem cell therapy and present a comprehensive list of every available clinical trial in national and international registries. Finally, we discuss drawbacks arising from AD pathology and technical limitations that hinder the transition of stem cell technology from bench to bedside. Our findings emphasize the need to increase clinical trials towards uncovering the mode of action and the underlying therapeutic mechanisms of transplanted cells as well as the molecular mechanisms controlling regeneration and neuronal microenvironment.
Collapse
Affiliation(s)
- Nikolaos Karvelas
- Faculty of Medicine, National and Kapodistrian University of Athens, Athina, Greece
| | | | - Georgios Politis
- Faculty of Medicine, National and Kapodistrian University of Athens, Athina, Greece
| | | | - Christo Kole
- Faculty of Medicine, National and Kapodistrian University of Athens, Athina, Greece
| |
Collapse
|
35
|
Li W, Yang D, Yan C, Chen M, Li Q, Zhu W, Wu G. Characterizing Network Selectiveness to the Dynamic Spreading of Neuropathological Events in Alzheimer's Disease. J Alzheimers Dis 2022; 86:1805-1816. [PMID: 35253761 PMCID: PMC9482760 DOI: 10.3233/jad-215596] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
BACKGROUND Mounting evidence shows that the neuropathological burdens manifest preference in affecting brain regions during the dynamic progression of Alzheimer's disease (AD). Since the distinct brain regions are physically wired by white matter fibers, it is reasonable to hypothesize the differential spreading pattern of neuropathological burdens may underlie the wiring topology, which can be characterized using neuroimaging and network science technologies. OBJECTIVE To study the dynamic spreading patterns of neuropathological events in AD. METHODS We first examine whether hub nodes with high connectivity in the brain network (assemble of white matter wirings) are susceptible to a higher level of pathological burdens than other regions that are less involved in the process of information exchange in the network. Moreover, we propose a novel linear mixed-effect model to characterize the multi-factorial spreading process of neuropathological burdens from hub nodes to non-hub nodes, where age, sex, and APOE4 indicators are considered as confounders. We apply our statistical model to the longitudinal neuroimaging data of amyloid-PET and tau-PET, respectively. RESULTS Our meta-data analysis results show that 1) AD differentially affects hub nodes with a significantly higher level of pathology, and 2) the longitudinal increase of neuropathological burdens on non-hub nodes is strongly correlated with the connectome distance to hub nodes rather than the spatial proximity. CONCLUSION The spreading pathway of AD neuropathological burdens might start from hub regions and propagate through the white matter fibers in a prion-like manner.
Collapse
Affiliation(s)
- Wenchao Li
- Intelligent Information Processing Laboratory, Hangzhou Dianzi University, Hangzhou, China
| | - Defu Yang
- Intelligent Information Processing Laboratory, Hangzhou Dianzi University, Hangzhou, China
- Research Center for Healthcare Data Science, Zhejiang Lab, Hangzhou, Zhejiang, China
| | - Chenggang Yan
- Intelligent Information Processing Laboratory, Hangzhou Dianzi University, Hangzhou, China
| | - Minghan Chen
- Department of Computer Science, Wake Forest University, Winston-Salem, NC, USA
| | - Quefeng Li
- Department of Biostatistics, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Wentao Zhu
- Research Center for Healthcare Data Science, Zhejiang Lab, Hangzhou, Zhejiang, China
| | - Guorong Wu
- Department of Psychiatry, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Department of Computer Science, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| |
Collapse
|
36
|
Borcuk C, Héraud C, Herbeaux K, Diringer M, Panzer É, Scuto J, Hashimoto S, Saido TC, Saito T, Goutagny R, Battaglia D, Mathis C. Early memory deficits and extensive brain network disorganization in the AppNL-F/MAPT double knock-in mouse model of familial Alzheimer's disease. AGING BRAIN 2022; 2:100042. [PMID: 36908877 PMCID: PMC9997176 DOI: 10.1016/j.nbas.2022.100042] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2022] [Accepted: 05/18/2022] [Indexed: 10/18/2022] Open
Abstract
A critical challenge in current research on Alzheimer's disease (AD) is to clarify the relationship between network dysfunction and the emergence of subtle memory deficits in itspreclinical stage. The AppNL-F/MAPT double knock-in (dKI) model with humanized β-amyloid peptide (Aβ) and tau was used to investigate both memory and network dysfunctions at an early stage. Young male dKI mice (2 to 6 months) were tested in three tasks taxing different aspects of recognition memory affected in preclinical AD. An early deficit first appeared in the object-place association task at the age of 4 months, when increased levels of β-CTF and Aβ were detected in both the hippocampus and the medial temporal cortex, and tau pathology was found only in the medial temporal cortex. Object-place task-dependent c-Fos activation was then analyzed in 22 subregions across the medial prefrontal cortex, claustrum, retrosplenial cortex, and medial temporal lobe. Increased c-Fos activation was detected in the entorhinal cortex and the claustrum of dKI mice. During recall, network efficiency was reduced across cingulate regions with a major disruption of information flow through the retrosplenial cortex. Our findings suggest that early perirhinal-entorhinal pathology is associated with abnormal activity which may spread to downstream regions such as the claustrum, the medial prefrontal cortex and ultimately the key retrosplenial hub which relays information from frontal to temporal lobes. The similarity between our findings and those reported in preclinical stages of AD suggests that the AppNL-F/MAPT dKI model has a high potential for providing key insights into preclinical AD.
Collapse
Key Words
- AD, Alzheimer’s disease
- ADAD, autosomal dominant Alzheimer’s disease
- Associative memory
- CLA, claustrum
- Claustrum
- DMN, default mode network
- EI, exploration index
- FC, functional connectivity
- Functional connectivity
- MI, Memory index
- MTC, medial temporal cortex
- MTL, medial temporal lobe
- Medial temporal cortex
- NOR, novel object recognition
- OL, Object location
- OP, object-place
- PS, Pattern Separation
- Preclinical Alzheimer disease
- Retrosplenial cortex
- aMCI, amnestic mild cognitive impairment
- amyloid beta, Aβ
- dKI, AppNL-F/MAPT double knock-in
- ptau Thr 181, Thr181phosphorylated tau protein
Collapse
Affiliation(s)
- Christopher Borcuk
- Université de Strasbourg, CNRS, Laboratoire de Neurosciences Cognitives et Adaptatives (LNCA) UMR 7364, F-67000 Strasbourg, France
| | - Céline Héraud
- Université de Strasbourg, CNRS, Laboratoire de Neurosciences Cognitives et Adaptatives (LNCA) UMR 7364, F-67000 Strasbourg, France
| | - Karine Herbeaux
- Université de Strasbourg, CNRS, Laboratoire de Neurosciences Cognitives et Adaptatives (LNCA) UMR 7364, F-67000 Strasbourg, France
| | - Margot Diringer
- Université de Strasbourg, CNRS, Laboratoire de Neurosciences Cognitives et Adaptatives (LNCA) UMR 7364, F-67000 Strasbourg, France
| | - Élodie Panzer
- Université de Strasbourg, CNRS, Laboratoire de Neurosciences Cognitives et Adaptatives (LNCA) UMR 7364, F-67000 Strasbourg, France
| | - Jil Scuto
- Université de Strasbourg, CNRS, Laboratoire de Neurosciences Cognitives et Adaptatives (LNCA) UMR 7364, F-67000 Strasbourg, France
| | - Shoko Hashimoto
- Laboratory for Proteolytic Neuroscience, RIKEN Center for Brain Science, 2-1 Hirosawa, Wako-city, Saitama 351-0198, Japan
| | - Takaomi C Saido
- Laboratory for Proteolytic Neuroscience, RIKEN Center for Brain Science, 2-1 Hirosawa, Wako-city, Saitama 351-0198, Japan
| | - Takashi Saito
- Laboratory for Proteolytic Neuroscience, RIKEN Center for Brain Science, 2-1 Hirosawa, Wako-city, Saitama 351-0198, Japan
| | - Romain Goutagny
- Université de Strasbourg, CNRS, Laboratoire de Neurosciences Cognitives et Adaptatives (LNCA) UMR 7364, F-67000 Strasbourg, France
| | - Demian Battaglia
- Université de Strasbourg, CNRS, Laboratoire de Neurosciences Cognitives et Adaptatives (LNCA) UMR 7364, F-67000 Strasbourg, France.,University of Strasbourg Institute for Advanced Studies (USIAS), F-67000 Strasbourg, France.,Université d'Aix-Marseille, Inserm, Institut de Neurosciences des Systèmes (INS) UMR_S 1106, F-13005 Marseille, France
| | - Chantal Mathis
- Université de Strasbourg, CNRS, Laboratoire de Neurosciences Cognitives et Adaptatives (LNCA) UMR 7364, F-67000 Strasbourg, France
| |
Collapse
|
37
|
Schumacher J, Gunter JL, Przybelski SA, Jones DT, Graff-Radford J, Savica R, Schwarz CG, Senjem ML, Jack CR, Lowe VJ, Knopman DS, Fields JA, Kremers WK, Petersen RC, Graff-Radford NR, Ferman TJ, Boeve BF, Thomas AJ, Taylor JP, Kantarci K. Dementia with Lewy bodies: association of Alzheimer pathology with functional connectivity networks. Brain 2021; 144:3212-3225. [PMID: 34114602 PMCID: PMC8634124 DOI: 10.1093/brain/awab218] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Revised: 03/19/2021] [Accepted: 04/22/2021] [Indexed: 11/22/2022] Open
Abstract
Dementia with Lewy bodies (DLB) is neuropathologically defined by the presence of α-synuclein aggregates, but many DLB cases show concurrent Alzheimer's disease pathology in the form of amyloid-β plaques and tau neurofibrillary tangles. The first objective of this study was to investigate the effect of Alzheimer's disease co-pathology on functional network changes within the default mode network (DMN) in DLB. Second, we studied how the distribution of tau pathology measured with PET relates to functional connectivity in DLB. Twenty-seven DLB, 26 Alzheimer's disease and 99 cognitively unimpaired participants (balanced on age and sex to the DLB group) underwent tau-PET with AV-1451 (flortaucipir), amyloid-β-PET with Pittsburgh compound-B (PiB) and resting-state functional MRI scans. The resing-state functional MRI data were used to assess functional connectivity within the posterior DMN. This was then correlated with overall cortical flortaucipir PET and PiB PET standardized uptake value ratio (SUVr). The strength of interregional functional connectivity was assessed using the Schaefer atlas. Tau-PET covariance was measured as the correlation in flortaucipir SUVr between any two regions across participants. The association between region-to-region functional connectivity and tau-PET covariance was assessed using linear regression. Additionally, we identified the region with highest and the region with lowest tau SUVrs (tau hot- and cold spots) and tested whether tau SUVr in all other brain regions was associated with the strength of functional connectivity to these tau hot and cold spots. A reduction in posterior DMN connectivity correlated with overall higher cortical tau- (r = -0.39, P = 0.04) and amyloid-PET uptake (r = -0.41, P = 0.03) in the DLB group, i.e. patients with DLB who have more concurrent Alzheimer's disease pathology showed a more severe loss of DMN connectivity. Higher functional connectivity between regions was associated with higher tau covariance in cognitively unimpaired, Alzheimer's disease and DLB. Furthermore, higher functional connectivity of a target region to the tau hotspot (i.e. inferior/medial temporal cortex) was related to higher flortaucipir SUVrs in the target region, whereas higher functional connectivity to the tau cold spot (i.e. sensory-motor cortex) was related to lower flortaucipir SUVr in the target region. Our findings suggest that a higher burden of Alzheimer's disease co-pathology in patients with DLB is associated with more Alzheimer's disease-like changes in functional connectivity. Furthermore, we found an association between the brain's functional network architecture and the distribution of tau pathology that has recently been described in Alzheimer's disease. We show that this relationship also exists in patients with DLB, indicating that similar mechanisms of connectivity-dependent occurrence of tau pathology might be at work in both diseases.
Collapse
Affiliation(s)
- Julia Schumacher
- Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, UK
- Department of Radiology, Mayo Clinic, Rochester, MN, USA
| | - Jeffrey L Gunter
- Department of Information Technology, Mayo Clinic, Rochester, MN, USA
| | - Scott A Przybelski
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN, USA
| | - David T Jones
- Department of Neurology, Mayo Clinic, Rochester, MN, USA
| | | | - Rodolfo Savica
- Department of Neurology, Mayo Clinic, Rochester, MN, USA
| | | | - Matthew L Senjem
- Department of Information Technology, Mayo Clinic, Rochester, MN, USA
| | | | - Val J Lowe
- Department of Radiology, Mayo Clinic, Rochester, MN, USA
| | | | - Julie A Fields
- Department of Psychiatry and Psychology, Mayo Clinic, Rochester, MN, USA
| | - Walter K Kremers
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN, USA
| | | | | | - Tanis J Ferman
- Department of Psychiatry and Psychology, Mayo Clinic, Jacksonville, FL, USA
| | | | - Alan J Thomas
- Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, UK
| | - John-Paul Taylor
- Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, UK
| | - Kejal Kantarci
- Department of Radiology, Mayo Clinic, Rochester, MN, USA
| |
Collapse
|
38
|
Hampel H, Hardy J, Blennow K, Chen C, Perry G, Kim SH, Villemagne VL, Aisen P, Vendruscolo M, Iwatsubo T, Masters CL, Cho M, Lannfelt L, Cummings JL, Vergallo A. The Amyloid-β Pathway in Alzheimer's Disease. Mol Psychiatry 2021; 26:5481-5503. [PMID: 34456336 PMCID: PMC8758495 DOI: 10.1038/s41380-021-01249-0] [Citation(s) in RCA: 693] [Impact Index Per Article: 173.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/19/2021] [Revised: 07/19/2021] [Accepted: 07/28/2021] [Indexed: 02/06/2023]
Abstract
Breakthroughs in molecular medicine have positioned the amyloid-β (Aβ) pathway at the center of Alzheimer's disease (AD) pathophysiology. While the detailed molecular mechanisms of the pathway and the spatial-temporal dynamics leading to synaptic failure, neurodegeneration, and clinical onset are still under intense investigation, the established biochemical alterations of the Aβ cycle remain the core biological hallmark of AD and are promising targets for the development of disease-modifying therapies. Here, we systematically review and update the vast state-of-the-art literature of Aβ science with evidence from basic research studies to human genetic and multi-modal biomarker investigations, which supports a crucial role of Aβ pathway dyshomeostasis in AD pathophysiological dynamics. We discuss the evidence highlighting a differentiated interaction of distinct Aβ species with other AD-related biological mechanisms, such as tau-mediated, neuroimmune and inflammatory changes, as well as a neurochemical imbalance. Through the lens of the latest development of multimodal in vivo biomarkers of AD, this cross-disciplinary review examines the compelling hypothesis- and data-driven rationale for Aβ-targeting therapeutic strategies in development for the early treatment of AD.
Collapse
Affiliation(s)
- Harald Hampel
- Eisai Inc., Neurology Business Group, Woodcliff Lake, NJ, USA.
| | - John Hardy
- UK Dementia Research Institute at UCL and Department of Neurodegenerative Disease, UCL Institute of Neurology, University College London, London, UK
| | - Kaj Blennow
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
- Institute of Neuroscience and Physiology, Department of Psychiatry and Neurochemistry, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
| | - Christopher Chen
- Memory Aging and Cognition Centre, Departments of Pharmacology and Psychological Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - George Perry
- Department of Biology and Neurosciences Institute, University of Texas at San Antonio (UTSA), San Antonio, TX, USA
| | - Seung Hyun Kim
- Department of Neurology, College of Medicine, Hanyang University, Seoul, Republic of Korea; Cell Therapy Center, Hanyang University Hospital, Seoul, Republic of Korea
| | - Victor L Villemagne
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, USA
- Department of Medicine, The University of Melbourne, Melbourne, VIC, Australia
| | - Paul Aisen
- USC Alzheimer's Therapeutic Research Institute, San Diego, CA, USA
| | - Michele Vendruscolo
- Centre for Misfolding Diseases, Department of Chemistry, University of Cambridge, Cambridge, UK
| | - Takeshi Iwatsubo
- Department of Neuropathology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Colin L Masters
- Laureate Professor of Dementia Research, Florey Institute and The University of Melbourne, Parkville, VIC, Australia
| | - Min Cho
- Eisai Inc., Neurology Business Group, Woodcliff Lake, NJ, USA
| | - Lars Lannfelt
- Uppsala University, Department of of Public Health/Geriatrics, Uppsala, Sweden
- BioArctic AB, Stockholm, Sweden
| | - Jeffrey L Cummings
- Chambers-Grundy Center for Transformative Neuroscience, Department of Brain Health, School of Integrated Health Sciences, University of Nevada Las Vegas (UNLV), Las Vegas, NV, USA
| | - Andrea Vergallo
- Eisai Inc., Neurology Business Group, Woodcliff Lake, NJ, USA.
| |
Collapse
|
39
|
Yu M, Sporns O, Saykin AJ. The human connectome in Alzheimer disease - relationship to biomarkers and genetics. Nat Rev Neurol 2021; 17:545-563. [PMID: 34285392 PMCID: PMC8403643 DOI: 10.1038/s41582-021-00529-1] [Citation(s) in RCA: 112] [Impact Index Per Article: 28.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/10/2021] [Indexed: 02/06/2023]
Abstract
The pathology of Alzheimer disease (AD) damages structural and functional brain networks, resulting in cognitive impairment. The results of recent connectomics studies have now linked changes in structural and functional network organization in AD to the patterns of amyloid-β and tau accumulation and spread, providing insights into the neurobiological mechanisms of the disease. In addition, the detection of gene-related connectome changes might aid in the early diagnosis of AD and facilitate the development of personalized therapeutic strategies that are effective at earlier stages of the disease spectrum. In this article, we review studies of the associations between connectome changes and amyloid-β and tau pathologies as well as molecular genetics in different subtypes and stages of AD. We also highlight the utility of connectome-derived computational models for replicating empirical findings and for tracking and predicting the progression of biomarker-indicated AD pathophysiology.
Collapse
Affiliation(s)
- Meichen Yu
- Indiana Alzheimer's Disease Research Center, Indiana University School of Medicine, Indianapolis, IN, USA
- Department of Radiology and Imaging Sciences, Indiana University School of Medicine, Indianapolis, IN, USA
- Indiana University Network Science Institute, Bloomington, IN, USA
| | - Olaf Sporns
- Indiana Alzheimer's Disease Research Center, Indiana University School of Medicine, Indianapolis, IN, USA
- Department of Radiology and Imaging Sciences, Indiana University School of Medicine, Indianapolis, IN, USA
- Indiana University Network Science Institute, Bloomington, IN, USA
- Department of Psychological and Brain Sciences, Indiana University, Bloomington, IN, USA
| | - Andrew J Saykin
- Indiana Alzheimer's Disease Research Center, Indiana University School of Medicine, Indianapolis, IN, USA.
- Department of Radiology and Imaging Sciences, Indiana University School of Medicine, Indianapolis, IN, USA.
- Indiana University Network Science Institute, Bloomington, IN, USA.
| |
Collapse
|
40
|
Quattrini G, Marizzoni M, Pizzini FB, Galazzo IB, Aiello M, Didic M, Soricelli A, Albani D, Romano M, Blin O, Forloni G, Golay X, Jovicich J, Nathan PJ, Richardson JC, Salvatore M, Frisoni GB, Pievani M. Convergent and Discriminant Validity of Default Mode Network and Limbic Network Perfusion in Amnestic Mild Cognitive Impairment Patients. J Alzheimers Dis 2021; 82:1797-1808. [PMID: 34219733 DOI: 10.3233/jad-210531] [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] [Indexed: 11/15/2022]
Abstract
BACKGROUND Previous studies reported default mode network (DMN) and limbic network (LIN) brain perfusion deficits in patients with amnestic mild cognitive impairment (aMCI), frequently a prodromal stage of Alzheimer's disease (AD). However, the validity of these measures as AD markers has not yet been tested using MRI arterial spin labeling (ASL). OBJECTIVE To investigate the convergent and discriminant validity of DMN and LIN perfusion in aMCI. METHODS We collected core AD markers (amyloid-β 42 [Aβ42], phosphorylated tau 181 levels in cerebrospinal fluid [CSF]), neurodegenerative (hippocampal volumes and CSF total tau), vascular (white matter hyperintensities), genetic (apolipoprotein E [APOE] status), and cognitive features (memory functioning on Paired Associate Learning test [PAL]) in 14 aMCI patients. Cerebral blood flow (CBF) was extracted from DMN and LIN using ASL and correlated with AD features to assess convergent validity. Discriminant validity was assessed carrying out the same analysis with AD-unrelated features, i.e., somatomotor and visual networks' perfusion, cerebellar volume, and processing speed. RESULTS Perfusion was reduced in the DMN (F = 5.486, p = 0.039) and LIN (F = 12.678, p = 0.004) in APOE ɛ4 carriers compared to non-carriers. LIN perfusion correlated with CSF Aβ42 levels (r = 0.678, p = 0.022) and memory impairment (PAL, number of errors, r = -0.779, p = 0.002). No significant correlation was detected with tau, neurodegeneration, and vascular features, nor with AD-unrelated features. CONCLUSION Our results support the validity of DMN and LIN ASL perfusion as AD markers in aMCI, indicating a significant correlation between CBF and amyloidosis, APOE ɛ4, and memory impairment.
Collapse
Affiliation(s)
- Giulia Quattrini
- Laboratory of Alzheimer's Neuroimaging and Epidemiology (LANE), IRCCS Istituto Centro San Giovanni di Dio Fatebenefratelli, Brescia, Italy.,Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Moira Marizzoni
- Laboratory of Alzheimer's Neuroimaging and Epidemiology (LANE), IRCCS Istituto Centro San Giovanni di Dio Fatebenefratelli, Brescia, Italy.,Laboratory of Biological Psychiatry, IRCCS Istituto Centro San Giovanni di Dio Fatebenefratelli, Brescia, Italy
| | - Francesca B Pizzini
- Radiology, Department of Diagnostic and Public Health, University of Verona, Verona, Italy
| | | | | | - Mira Didic
- Aix-Marseille Univ, INSERM, INS, Instit Neurosci des Syst, Marseille, France.,APHM, Timone, Service de Neurologie et Neuropsychologie, Hôpital Timone Adultes, Marseille, France
| | - Andrea Soricelli
- IRCCS SDN, Napoli, Italy.,Department of Sport Sciences, University of Naples Parthenope, Naples, Italy
| | - Diego Albani
- Neuroscience Department, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milano, Italy
| | - Melissa Romano
- Laboratory of Alzheimer's Neuroimaging and Epidemiology (LANE), IRCCS Istituto Centro San Giovanni di Dio Fatebenefratelli, Brescia, Italy
| | - Olivier Blin
- Aix-Marseille Univ, INSERM, INS, Instit Neurosci des Syst, DHUNE, Ap-Hm, Marseille, France
| | - Gianluigi Forloni
- Neuroscience Department, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milano, Italy
| | - Xavier Golay
- Department of Brain Repair and Rehabilitation, UCL Queen Square Institute of Neurology, London, United Kingdom
| | - Jorge Jovicich
- Center for Mind/Brain Sciences - CIMeC, University of Trento, Rovereto, Italy
| | - Pradeep J Nathan
- Department of Psychiatry, University of Cambridge, Cambridge, UK
| | - Jill C Richardson
- Neurosciences Therapeutic Area, GlaxoSmithKline R&D, Gunnels Wood Road, Stevenage, United Kingdom
| | | | - Giovanni B Frisoni
- Memory Clinic and LANVIE-Laboratory of Neuroimaging of Aging, University Hospitals and University of Geneva, Geneva, Switzerland
| | - Michela Pievani
- Laboratory of Alzheimer's Neuroimaging and Epidemiology (LANE), IRCCS Istituto Centro San Giovanni di Dio Fatebenefratelli, Brescia, Italy
| | | |
Collapse
|
41
|
Abstract
PURPOSE OF REVIEW The purpose of this review was to discuss the contribution of the most recent neuroimaging studies to our understanding of the mechanisms underlying Alzheimer's disease. RECENT FINDINGS Studies have applied cross-sectional and longitudinal positron emission tomography (PET), structural and resting-state functional magnetic resonance imaging to primarily investigate (1) how Alzheimer's disease pathological hallmarks like tau and amyloid-beta build up and spread across the brain at different disease stage and in different disease phenotypes and (2) how the spreading of these proteins is related to atrophy, to neuronal network disruption and to neuroinflammation. SUMMARY The findings of these studies offer insight on the mechanisms that drive the pathological and clinical progression of Alzheimer's disease, highlighting their multifactorial nature, which is a crucial aspect for the development of disease-modifying therapeutics and can be captured with multimodal imaging approaches.
Collapse
|
42
|
Carlson ML, Toueg TN, Khalighi MM, Castillo J, Shen B, Azevedo EC, DiGiacomo P, Mouchawar N, Chau G, Zaharchuk G, James ML, Mormino EC, Zeineh MM. Hippocampal subfield imaging and fractional anisotropy show parallel changes in Alzheimer's disease tau progression using simultaneous tau-PET/MRI at 3T. ALZHEIMER'S & DEMENTIA (AMSTERDAM, NETHERLANDS) 2021; 13:e12218. [PMID: 34337132 PMCID: PMC8319659 DOI: 10.1002/dad2.12218] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/11/2021] [Revised: 06/01/2021] [Accepted: 06/04/2021] [Indexed: 01/03/2023]
Abstract
INTRODUCTION Alzheimer's disease (AD) is the most common form of dementia, characterized primarily by abnormal aggregation of two proteins, tau and amyloid beta. We assessed tau pathology and white matter connectivity changes in subfields of the hippocampus simultaneously in vivo in AD. METHODS Twenty-four subjects were scanned using simultaneous time-of-flight 18F-PI-2620 tau positron emission tomography/3-Tesla magnetic resonance imaging and automated segmentation. RESULTS We observed extensive tau elevation in the entorhinal/perirhinal regions, intermediate tau elevation in cornu ammonis 1/subiculum, and an absence of tau elevation in the dentate gyrus, relative to controls. Diffusion tensor imaging showed parahippocampal gyral fractional anisotropy was lower in AD and mild cognitive impairment compared to controls and strongly correlated with early tau accumulation in the entorhinal and perirhinal cortices. DISCUSSION This study demonstrates the potential for quantifiable patterns of 18F-PI2620 binding in hippocampus subfields, accompanied by diffusion and volume metrics, to be valuable markers of AD.
Collapse
Affiliation(s)
| | - Tyler N. Toueg
- Department of NeurologyStanford UniversityStanfordCaliforniaUSA
| | | | - Jessa Castillo
- Department of RadiologyStanford UniversityStanfordCaliforniaUSA
| | - Bin Shen
- Department of RadiologyStanford UniversityStanfordCaliforniaUSA
| | | | - Phillip DiGiacomo
- Department of BioengineeringStanford UniversityStanfordCaliforniaUSA
| | | | - Gustavo Chau
- Department of BioengineeringStanford UniversityStanfordCaliforniaUSA
| | - Greg Zaharchuk
- Department of RadiologyStanford UniversityStanfordCaliforniaUSA
| | - Michelle L. James
- Department of NeurologyStanford UniversityStanfordCaliforniaUSA
- Department of RadiologyStanford UniversityStanfordCaliforniaUSA
| | | | | |
Collapse
|
43
|
Dautricourt S, de Flores R, Landeau B, Poisnel G, Vanhoutte M, Delcroix N, Eustache F, Vivien D, de la Sayette V, Chételat G. Longitudinal Changes in Hippocampal Network Connectivity in Alzheimer's Disease. Ann Neurol 2021; 90:391-406. [PMID: 34279043 PMCID: PMC9291910 DOI: 10.1002/ana.26168] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2021] [Revised: 07/13/2021] [Accepted: 07/15/2021] [Indexed: 11/05/2022]
Abstract
Objective The hippocampus is connected to 2 distinct cortical brain networks, the posterior–medial and the anterior–temporal networks, involving different medial temporal lobe (MTL) subregions. The aim of this study was to assess the functional alterations of these 2 networks, their changes over time, and links to cognition in Alzheimer's disease. Methods We assessed MTL connectivity in 53 amyloid‐β–positive patients with mild cognitive impairment and AD dementia and 68 healthy elderly controls, using resting‐state functional magnetic resonance imaging, cross‐sectionally and longitudinally. First, we compared the functional connectivity of the posterior–medial and anterior–temporal networks within the control group to highlight their specificities. Second, we compared the connectivity of these networks between groups, and between baseline and 18‐month follow‐up in patients. Third, we assessed the association in the connectivity changes between the 2 networks, and with cognitive performance. Results We found decreased connectivity in patients specifically between the hippocampus and the posterior–medial network, together with increased connectivity between several MTL subregions and the anterior–temporal network. Moreover, changes in the posterior–medial and anterior–temporal networks were interrelated such that decreased MTL–posterior–medial connectivity was associated with increased MTL–anterior–temporal connectivity. Finally, both MTL–posterior–medial decrease and MTL–anterior–temporal increase predicted cognitive decline. Interpretation Our findings demonstrate that longitudinal connectivity changes in the posterior–medial and anterior–temporal hippocampal networks are linked together and that they both contribute to cognitive decline in Alzheimer's disease. These results shed light on the critical role of the posterior–medial and anterior–temporal networks in Alzheimer's disease pathophysiology and clinical symptoms. ANN NEUROL 2021;90:391–406
Collapse
Affiliation(s)
- Sophie Dautricourt
- Normandie Univ, UNICAEN, INSERM, PhIND.,Neurology Department, Caen-Normandie University Hospital, Caen, France
| | | | | | | | | | - Nicolas Delcroix
- CNRS, Unité Mixte de Service-3408, GIP CYCERON, Bd Henri Becquerel, BP5229, 14074 Caen cedex, France
| | - Francis Eustache
- Normandie Univ, UNICAEN, PSL Université, EPHE, INSERM, U1077, CHU de Caen, GIP Cyceron, Neuropsychologie et Imagerie de la Mémoire Humaine, Caen, France
| | - Denis Vivien
- Normandie Univ, UNICAEN, INSERM, PhIND.,Department of Clinical Research, Caen-Normandie University Hospital, Caen, France
| | - Vincent de la Sayette
- Neurology Department, Caen-Normandie University Hospital, Caen, France.,Normandie Univ, UNICAEN, PSL Université, EPHE, INSERM, U1077, CHU de Caen, GIP Cyceron, Neuropsychologie et Imagerie de la Mémoire Humaine, Caen, France
| | | |
Collapse
|
44
|
Schäfer A, Peirlinck M, Linka K, Kuhl E. Bayesian Physics-Based Modeling of Tau Propagation in Alzheimer's Disease. Front Physiol 2021; 12:702975. [PMID: 34335308 PMCID: PMC8322942 DOI: 10.3389/fphys.2021.702975] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Accepted: 06/22/2021] [Indexed: 11/24/2022] Open
Abstract
Amyloid-β and hyperphosphorylated tau protein are known drivers of neuropathology in Alzheimer's disease. Tau in particular spreads in the brains of patients following a spatiotemporal pattern that is highly sterotypical and correlated with subsequent neurodegeneration. Novel medical imaging techniques can now visualize the distribution of tau in the brain in vivo, allowing for new insights to the dynamics of this biomarker. Here we personalize a network diffusion model with global spreading and local production terms to longitudinal tau positron emission tomography data of 76 subjects from the Alzheimer's Disease Neuroimaging Initiative. We use Bayesian inference with a hierarchical prior structure to infer means and credible intervals for our model parameters on group and subject levels. Our results show that the group average protein production rate for amyloid positive subjects is significantly higher with 0.019±0.27/yr, than that for amyloid negative subjects with -0.143±0.21/yr (p = 0.0075). These results support the hypothesis that amyloid pathology drives tau pathology. The calibrated model could serve as a valuable clinical tool to identify optimal time points for follow-up scans and predict the timeline of disease progression.
Collapse
Affiliation(s)
- Amelie Schäfer
- Department of Mechanical Engineering, Stanford University, Stanford, CA, United States
| | - Mathias Peirlinck
- Department of Mechanical Engineering, Stanford University, Stanford, CA, United States
| | - Kevin Linka
- Institute of Continuum and Materials Mechanics, Hamburg University of Technology, Hamburg, Germany
| | - Ellen Kuhl
- Department of Mechanical Engineering, Stanford University, Stanford, CA, United States
| | | |
Collapse
|
45
|
Merenstein JL, Corrada MM, Kawas CH, Bennett IJ. Age affects white matter microstructure and episodic memory across the older adult lifespan. Neurobiol Aging 2021; 106:282-291. [PMID: 34332220 DOI: 10.1016/j.neurobiolaging.2021.06.021] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2020] [Revised: 06/25/2021] [Accepted: 06/28/2021] [Indexed: 02/07/2023]
Abstract
Diffusion imaging studies have observed age-related degradation of white matter that contributes to cognitive deficits separately in younger-old (ages 65-89) and oldest-old (ages 90+) adults. But it remains unclear whether these age effects are magnified in advanced age groups, which may reflect disease-related pathology. Here, we tested whether age-related differences in white matter microstructure followed linear or nonlinear patterns across the entire older adult lifespan (65-98 years), these patterns were influenced by oldest-old adults at increased risk of dementia (cognitive impairment no dementia, CIND), and they explained age effects on episodic memory. Results revealed nonlinear microstructure declines across fiber classes (medial temporal, callosal, association, projection and/or thalamic) that were largest for medial temporal fibers. These patterns remained after excluding oldest-old participants with CIND, indicating that aging of white matter microstructure cannot solely be explained by pathology associated with early cognitive impairment. Moreover, finding that the effect of age on episodic memory was mediated by medial temporal fiber microstructure suggests it is essential for facilitating memory-related neural signals across the older adult lifespan.
Collapse
Affiliation(s)
| | - María M Corrada
- Institute for Memory Impairments and Neurological Disorders, University of California, Irvine, CA, USA; Department of Neurology, University of California, Irvine, CA, USA; Department of Epidemiology, University of California, Irvine, CA, USA
| | - Claudia H Kawas
- Institute for Memory Impairments and Neurological Disorders, University of California, Irvine, CA, USA; Department of Neurology, University of California, Irvine, CA, USA; Department of Neurobiology and Behavior, University of California, Irvine, CA, USA
| | - Ilana J Bennett
- Department of Psychology, University of California, Riverside, CA, USA
| |
Collapse
|
46
|
Wales RM, Leung HC. The Effects of Amyloid and Tau on Functional Network Connectivity in Older Populations. Brain Connect 2021; 11:599-612. [PMID: 33813858 DOI: 10.1089/brain.2020.0902] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Background: Neuroimaging studies suggest that aged brains show altered connectivity within and across functional networks. Similar changes in functional network integrity are also linked to the accumulation of pathological proteins in the brain, such as amyloid-beta plaques and neurofibrillary tau tangles seen in Alzheimer's disease. However, less is known about the specific impacts of amyloid and tau on functional network connectivity in cognitively normal older adults who harbor these proteins. Methods: We briefly summarize recent neuroimaging studies of aging and then thoroughly review positron emission tomography and functional magnetic resonance imaging studies measuring the relationship between amyloid-tau pathology and functional connectivity in cognitively normal older individuals. Results: The literature overall suggests that amyloid-positive older individuals show minor cognitive dysfunction and aberrant default mode network connectivity compared with amyloid-negative individuals. Tau, however, is more closely associated with network hypoconnectivity and poorer cognition. Those with substantial amyloid and tau experience even greater cognitive decline compared with those with primarily amyloid or tau, suggesting a potential interaction. Multimodal neuroimaging studies suggest that older adults with pathological protein deposits show amyloid-related hyperconnectivity and tau-related hypoconnectivity in multiple functional networks, including the default mode and frontoparietal networks. Discussion: We propose an updated model considering the effects of amyloid and tau on functional connectivity in older individuals. Large, longitudinal neuroimaging studies with multiple levels of analysis are required to obtain a deeper understanding of the dynamic relationship between pathological protein accumulation and functional connectivity changes, as amyloid- and tau-induced connectivity alterations may have critical and time-varying effects on neurodegeneration and cognitive decline. Impact statement Amyloid and tau accumulation have been linked with altered functional connectivity in cognitively normal older adults. This review synthesized recent functional imaging literatures in a discussion of how amyloid and tau can interactively affect functional connectivity in nonlinear ways, which can explain previous conflicting findings. Changes in connectivity strength may depend on the accumulation of both amyloid and tau, and their integrative effects seem to have critical consequences on cognition. Elucidating the effects of these pathological proteins on brain functioning is paramount to understand the etiology of Alzheimer's disease and the aging process overall.
Collapse
Affiliation(s)
- Ryan Michael Wales
- Integrative Neuroscience Program, Department of Psychology, Stony Brook University, Stony Brook, New York, USA
| | - Hoi-Chung Leung
- Integrative Neuroscience Program, Department of Psychology, Stony Brook University, Stony Brook, New York, USA
| |
Collapse
|
47
|
Pichet Binette A, Theaud G, Rheault F, Roy M, Collins DL, Levin J, Mori H, Lee JH, Farlow MR, Schofield P, Chhatwal JP, Masters CL, Benzinger T, Morris J, Bateman R, Breitner JC, Poirier J, Gonneaud J, Descoteaux M, Villeneuve S. Bundle-specific associations between white matter microstructure and Aβ and tau pathology in preclinical Alzheimer's disease. eLife 2021; 10:62929. [PMID: 33983116 PMCID: PMC8169107 DOI: 10.7554/elife.62929] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2020] [Accepted: 05/12/2021] [Indexed: 12/12/2022] Open
Abstract
Beta-amyloid (Aβ) and tau proteins, the pathological hallmarks of Alzheimer's disease (AD), are believed to spread through connected regions of the brain. Combining diffusion imaging and positron emission tomography, we investigated associations between white matter microstructure specifically in bundles connecting regions where Aβ or tau accumulates and pathology. We focused on free-water-corrected diffusion measures in the anterior cingulum, posterior cingulum, and uncinate fasciculus in cognitively normal older adults at risk of sporadic AD and presymptomatic mutation carriers of autosomal dominant AD. In Aβ-positive or tau-positive groups, lower tissue fractional anisotropy and higher mean diffusivity related to greater Aβ and tau burden in both cohorts. Associations were found in the posterior cingulum and uncinate fasciculus in preclinical sporadic AD, and in the anterior and posterior cingulum in presymptomatic mutation carriers. These results suggest that microstructural alterations accompany pathological accumulation as early as the preclinical stage of both sporadic and autosomal dominant AD.
Collapse
Affiliation(s)
- Alexa Pichet Binette
- Department of Psychiatry, Faculty of Medicine, McGill University, Montreal, Canada.,Douglas Mental Health University Institute, Montreal, Canada
| | - Guillaume Theaud
- Sherbrooke Connectivity Imaging Laboratory (SCIL), Université de Sherbrooke, Sherbrooke, Canada
| | - François Rheault
- Electrical Engineering, Vanderbilt University, Nashville, United States
| | - Maggie Roy
- Sherbrooke Connectivity Imaging Laboratory (SCIL), Université de Sherbrooke, Sherbrooke, Canada
| | - D Louis Collins
- McConnell Brain Imaging Centre, Montreal Neurological Institute, Montreal, Canada
| | - Johannes Levin
- Department of Neurology, Ludwig-Maximilians-Universität München, Munich, Germany.,German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
| | - Hiroshi Mori
- Department of Clinical Neuroscience, Osaka City University Medical School, Osaka, Japan
| | - Jae Hong Lee
- Department of Neurology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | | | - Peter Schofield
- Neuroscience Research Australia, Sydney, Australia.,School of Medical Sciences, UNSW Sydney, Sydney, Australia
| | - Jasmeer P Chhatwal
- Harvard Medical School, Massachusetts General Hospital, Boston, United States
| | - Colin L Masters
- The Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, Australia
| | - Tammie Benzinger
- Knight Alzheimer Disease Research Center, Washington University School of Medicine, St. Louis, United States.,Department of Neurology, Washington University School of Medicine, St. Louis, United States
| | - John Morris
- Knight Alzheimer Disease Research Center, Washington University School of Medicine, St. Louis, United States.,Department of Neurology, Washington University School of Medicine, St. Louis, United States
| | - Randall Bateman
- Knight Alzheimer Disease Research Center, Washington University School of Medicine, St. Louis, United States.,Department of Neurology, Washington University School of Medicine, St. Louis, United States
| | - John Cs Breitner
- Department of Psychiatry, Faculty of Medicine, McGill University, Montreal, Canada.,Douglas Mental Health University Institute, Montreal, Canada
| | - Judes Poirier
- Department of Psychiatry, Faculty of Medicine, McGill University, Montreal, Canada.,Douglas Mental Health University Institute, Montreal, Canada
| | - Julie Gonneaud
- Douglas Mental Health University Institute, Montreal, Canada.,Normandie Univ, UNICAEN, INSERM, U1237, Institut Blood and Brain @ Caen-Normandie, Cyceron, Caen, France
| | - Maxime Descoteaux
- Sherbrooke Connectivity Imaging Laboratory (SCIL), Université de Sherbrooke, Sherbrooke, Canada
| | - Sylvia Villeneuve
- Department of Psychiatry, Faculty of Medicine, McGill University, Montreal, Canada.,Douglas Mental Health University Institute, Montreal, Canada.,McConnell Brain Imaging Centre, Montreal Neurological Institute, Montreal, Canada
| | | | | |
Collapse
|
48
|
Nanjundaiah S, Chidambaram H, Chandrashekar M, Chinnathambi S. Role of Microglia in Regulating Cholesterol and Tau Pathology in Alzheimer's Disease. Cell Mol Neurobiol 2021; 41:651-668. [PMID: 32468440 DOI: 10.1007/s10571-020-00883-6] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2019] [Accepted: 05/19/2020] [Indexed: 01/21/2023]
Abstract
Cholesterol, a principal constituent of the cell membrane, plays a crucial role in the brain by regulating the synaptic transmission, neuronal signaling, as well as neurodegenerative diseases. Defects in the cholesterol trafficking are associated with enhanced generation of hyperphosphorylated Tau and Amyloid-β protein. Tau, a major microtubule-associated protein in the brain, is the key regulator of the mature neuron. Abnormally hyperphosphorylated Tau hampers the major functions related to microtubule assembly by promoting neurofibrillary tangles of paired helical filaments, twisted ribbons, and straight filaments. The observed pathological changes due to impaired cholesterol and Tau protein accumulation cause Alzheimer's disease. Thus, in order to regulate the pathogenesis of Alzheimer's disease, regulation of cholesterol metabolism, as well as Tau phosphorylation, is essential. The current review provides an overview of (1) cholesterol synthesis in the brain, neurons, astrocytes, and microglia; (2) the mechanism involved in modulating cholesterol concentration between the astrocytes and brain; (3) major mechanisms involved in the hyperphosphorylation of Tau and amyloid-β protein; and (4) microglial involvement in its regulation. Thus, the answering key questions will provide an in-depth information on microglia involvement in managing the pathogenesis of cholesterol-modulated hyperphosphorylated Tau protein.
Collapse
Affiliation(s)
- Shwetha Nanjundaiah
- Neurobiology Group, Division of Biochemical Sciences, CSIR-National Chemical Laboratory (CSIR-NCL), Dr. Homi Bhabha Road, Pune, 411008, India
| | - Hariharakrishnan Chidambaram
- Neurobiology Group, Division of Biochemical Sciences, CSIR-National Chemical Laboratory (CSIR-NCL), Dr. Homi Bhabha Road, Pune, 411008, India
- Academy of Scientific and Innovative Research (AcSIR), New Delhi, 110025, India
| | - Madhura Chandrashekar
- School of Biomedical Engineering and Sciences, MIT University, Loni Kalbhor, Pune, 412201, India
| | - Subashchandrabose Chinnathambi
- Neurobiology Group, Division of Biochemical Sciences, CSIR-National Chemical Laboratory (CSIR-NCL), Dr. Homi Bhabha Road, Pune, 411008, India.
- Academy of Scientific and Innovative Research (AcSIR), New Delhi, 110025, India.
| |
Collapse
|
49
|
Ni R, Röjdner J, Voytenko L, Dyrks T, Thiele A, Marutle A, Nordberg A. In vitro Characterization of the Regional Binding Distribution of Amyloid PET Tracer Florbetaben and the Glia Tracers Deprenyl and PK11195 in Autopsy Alzheimer's Brain Tissue. J Alzheimers Dis 2021; 80:1723-1737. [PMID: 33749648 PMCID: PMC8150513 DOI: 10.3233/jad-201344] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
BACKGROUND Emerging evidence indicates a central role of gliosis in Alzheimer's disease (AD) pathophysiology. However, the regional distribution and interaction of astrogliosis and microgliosis in association with amyloid-β (Aβ) still remain uncertain. OBJECTIVE Here we studied the pathological profiles in autopsy AD brain by using specific imaging tracers. METHODS Autopsy brain tissues of AD (n = 15, age 70.4±8.5 years) and control cases (n = 12, age 76.6±10.9) were examined with homogenate binding assays, autoradiography for Aβ plaques (3H-florbetaben/3H-PIB), astrogliosis (3H-L-deprenyl), and microgliosis (3H-PK11195/3H-FEMPA), as well as immunoassays. RESULTS In vitro saturation analysis revealed high-affinity binding sites of 3H-florbetaben, 3H-L-deprenyl, and 3H-PK11195/3H-FEMPA in the frontal cortex of AD cases. In vitro3H-florbetaben binding increased across cortical and subcortical regions of AD compared to control with the highest binding in the frontal and parietal cortices. The in vitro3H-L-deprenyl binding showed highest binding in the hippocampus (dentate gyrus) followed by cortical and subcortical regions of AD while the GFAP expression was upregulated only in the hippocampus compared to control. The in vitro3H-PK11195 binding was solely increased in the parietal cortex and the hippocampus of AD compared to control. The 3H-florbetaben binding positively correlated with the 3H-L-deprenyl binding in the hippocampus and parietal cortex of AD and controls. Similarly, a positive correlation was observed between 3H-florbetaben binding and GFAP expression in hippocampus of AD and control. CONCLUSION The use of multi-imaging tracers revealed different regional pattern of changes in autopsy AD brain with respect to amyloid plaque pathology versus astrogliosis and microgliosis.
Collapse
Affiliation(s)
- Ruiqing Ni
- Division of Clinical Geriatrics, Center for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden
| | - Jennie Röjdner
- Division of Clinical Geriatrics, Center for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden
| | - Larysa Voytenko
- Division of Clinical Geriatrics, Center for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden
| | | | | | - Amelia Marutle
- Division of Clinical Geriatrics, Center for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden
| | - Agneta Nordberg
- Division of Clinical Geriatrics, Center for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden.,Theme Aging, The Aging Brain Unit, Karolinska University Hospital, Stockholm, Sweden
| |
Collapse
|
50
|
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
|