1
|
Fan W, Yang S, Wei Y, Tian M, Liu Q, Li X, Ding J, Li X, Mao M, Han X, Du Y, Qiu C, Dong Y, Wang Y. Characterization of brain morphology associated with metabolic dysfunction-associated steatotic liver disease in the UK Biobank. Diabetes Obes Metab 2025; 27:3419-3430. [PMID: 40171859 DOI: 10.1111/dom.16362] [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: 10/09/2024] [Revised: 03/08/2025] [Accepted: 03/13/2025] [Indexed: 04/04/2025]
Abstract
BACKGROUND Emerging evidence has linked metabolic dysfunction-associated steatotic liver disease (MASLD) with accelerated cognitive decline and dementia. We aimed to investigate the associations of MASLD with volumes of total brain tissue and subcortical grey matter, and white matter microstructures in the UK Biobank. METHODS This cross-sectional study included 29,195 individuals (aged 45-82 years) from the UK Biobank who undertook a magnetic resonance imaging (MRI) sub-study between 2014 and 2022. The brain MRI covers three modalities (T1, T2 FLAIR, and diffusion). Volumes of grey matter, subcortical grey matter structures, and regional cortex were derived from T1-weighted images. Fractional anisotropy (FA) and mean diffusivity (MD) were derived from diffusion tensor imaging (DTI) to assess global and tract-specific microstructure. MASLD was defined as the MRI-derived proton density fat fraction (MRI-PDFF) ≥5% and the presence of at least one cardiometabolic criterion. Data were analysed using multiple linear regression models. RESULTS MASLD was significantly associated with smaller volumes of total grey matter and subcortical grey matter (p < 0.05) and reduced Alzheimer's disease (AD)-signature cortical thickness (multivariable-adjusted β = -0.04; 95% confidence interval [CI]: -0.07, -0.01). Having MASLD was associated with higher total white matter hyperintensity (WMH) volume (multivariable-adjusted β = 0.12; 95% CI: 0.10, 0.15). For white matter microstructure, MASLD was associated with increased global FA (multivariable-adjusted β = 0.05; 95% CI: 0.03, 0.08) and reduced global MD (multivariable-adjusted β = -0.04; 95% CI: -0.07, -0.01). CONCLUSIONS Brain morphology associated with MASLD is characterized by smaller subcortical grey matter volume and higher coherence but lower magnitudes of white matter microstructure.
Collapse
Affiliation(s)
- Wenxiao Fan
- Key Laboratory of Endocrine Glucose & Lipids Metabolism and Brain Aging, Ministry of Education, Department of Neurology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, People's Republic of China
- Shandong Institute of Brain Science and Brain-inspired Research, Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, People's Republic of China
| | - Shuping Yang
- Department of Oncology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, People's Republic of China
| | - Yiran Wei
- Key Laboratory of Endocrine Glucose & Lipids Metabolism and Brain Aging, Ministry of Education, Department of Neurology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, People's Republic of China
- Shandong Institute of Brain Science and Brain-inspired Research, Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, People's Republic of China
| | - Minle Tian
- Key Laboratory of Endocrine Glucose & Lipids Metabolism and Brain Aging, Ministry of Education, Department of Neurology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, People's Republic of China
- Shandong Institute of Brain Science and Brain-inspired Research, Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, People's Republic of China
| | - Qianying Liu
- Key Laboratory of Endocrine Glucose & Lipids Metabolism and Brain Aging, Ministry of Education, Department of Neurology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, People's Republic of China
- Shandong Institute of Brain Science and Brain-inspired Research, Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, People's Republic of China
| | - Xiaomeng Li
- Key Laboratory of Endocrine Glucose & Lipids Metabolism and Brain Aging, Ministry of Education, Department of Neurology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, People's Republic of China
- Shandong Institute of Brain Science and Brain-inspired Research, Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, People's Republic of China
| | - Jiahao Ding
- Shandong Institute of Brain Science and Brain-inspired Research, Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, People's Republic of China
| | - Xuewei Li
- Key Laboratory of Endocrine Glucose & Lipids Metabolism and Brain Aging, Ministry of Education, Department of Neurology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, People's Republic of China
- Shandong Institute of Brain Science and Brain-inspired Research, Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, People's Republic of China
| | - Ming Mao
- Key Laboratory of Endocrine Glucose & Lipids Metabolism and Brain Aging, Ministry of Education, Department of Neurology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, People's Republic of China
- Shandong Institute of Brain Science and Brain-inspired Research, Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, People's Republic of China
| | - Xiaolei Han
- Key Laboratory of Endocrine Glucose & Lipids Metabolism and Brain Aging, Ministry of Education, Department of Neurology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, People's Republic of China
- Shandong Institute of Brain Science and Brain-inspired Research, Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, People's Republic of China
| | - Yifeng Du
- Key Laboratory of Endocrine Glucose & Lipids Metabolism and Brain Aging, Ministry of Education, Department of Neurology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, People's Republic of China
- Shandong Institute of Brain Science and Brain-inspired Research, Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, People's Republic of China
| | - Chengxuan Qiu
- Key Laboratory of Endocrine Glucose & Lipids Metabolism and Brain Aging, Ministry of Education, Department of Neurology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, People's Republic of China
- Shandong Institute of Brain Science and Brain-inspired Research, Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, People's Republic of China
- Aging Research Center, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet-Stockholm University, Stockholm, Sweden
| | - Yi Dong
- Key Laboratory of Endocrine Glucose & Lipids Metabolism and Brain Aging, Ministry of Education, Department of Neurology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, People's Republic of China
- Shandong Institute of Brain Science and Brain-inspired Research, Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, People's Republic of China
| | - Yongxiang Wang
- Key Laboratory of Endocrine Glucose & Lipids Metabolism and Brain Aging, Ministry of Education, Department of Neurology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, People's Republic of China
- Shandong Institute of Brain Science and Brain-inspired Research, Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, People's Republic of China
- Aging Research Center, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet-Stockholm University, Stockholm, Sweden
| |
Collapse
|
2
|
Bao W, Bi H, Chao L, Jiang Y, Yu X, Ruan F, Wu D, Chen Z, Le K. Identifying the mediating role of brain atrophy on the relationship between DNA damage repair pathway and Alzheimer's disease: A Mendelian randomization analysis and mediation analysis. J Alzheimers Dis 2025:13872877251333811. [PMID: 40313062 DOI: 10.1177/13872877251333811] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/03/2025]
Abstract
BackgroundDNA damage and repair (DDR) and structural atrophies in different brain regions were recognized as critical factors in the onset of Alzheimer's disease (AD).ObjectiveWe utilized Mendelian randomization (MR) to examine the causal effects of the DDR-related molecular traits on AD and the potential mediating roles of different brain region volumes.MethodsIn primary analysis, we utilized public genome-wide association studies of AD and summary data from existing molecular traits datasets, including gene expression, DNA methylation, and protein levels quantitative trait loci (eQTL, mQTL, and pQTL) in both blood and brain to examine their causal associations by summary-data-based MR analysis and additional five two-sample MR methods. Subsequently, mediation analysis explored the potential mediate roles of 13 imaging-derived brain volume phenotypes in the associations between the DDR pathways and AD through a network MR design.ResultsWe found that the volumes of the right thalamus proper and global cerebral white matter mediated the causal pathways from EGFR to AD and relatively weak mediation effects of the right lateral ventricle volume in the causal pathways involving CHRNE, DNTT, and AD.ConclusionsWe identified causal relationships among DDR pathways, specific brain region volumes, and AD. Monitoring the molecular traits of these DDR-related genes and developing targeted drugs may help detect and interrupt the early progression of AD.
Collapse
Affiliation(s)
- Wei Bao
- Department of Rehabilitation Medicine, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi Province, China
- Department of Pediatrics, The First Affiliated Hospital of Nanchang University, Jiangxi Medical College, Nanchang, Jiangxi Province, China
| | - Haidi Bi
- Department of Rehabilitation Medicine, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi Province, China
| | - Lishuo Chao
- Department of Affective Disorders, The Affiliated Brain Hospital of Guangzhou Medical University (Guangzhou Huiai Hospital), Guangzhou, Guangdong Province, China
| | - Yaqing Jiang
- Department of Rehabilitation Medicine, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi Province, China
| | - Xiaoping Yu
- Department of Rehabilitation Medicine, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi Province, China
| | - Fei Ruan
- Department of Rehabilitation Medicine, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi Province, China
| | - Di Wu
- Department of Rehabilitation Medicine, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi Province, China
- Department of Pediatrics, The First Affiliated Hospital of Nanchang University, Jiangxi Medical College, Nanchang, Jiangxi Province, China
| | - Zhaoyan Chen
- Department of Rehabilitation Medicine, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi Province, China
| | - Kai Le
- Department of Rehabilitation Medicine, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi Province, China
- Department of Rehabilitation Sciences, Faculty of Health and Social Sciences, Hong Kong Polytechnic University, Hong Kong S.A.R., China
| |
Collapse
|
3
|
Vollhardt A, Frölich L, Stockbauer AC, Danek A, Schmitz C, Wahl AS. Towards a better diagnosis and treatment of dementia: Identifying common and distinct neuropathological mechanisms in Alzheimer's and vascular dementia. Neurobiol Dis 2025; 208:106845. [PMID: 39999928 DOI: 10.1016/j.nbd.2025.106845] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2024] [Revised: 02/05/2025] [Accepted: 02/15/2025] [Indexed: 02/27/2025] Open
Abstract
Alzheimer's disease (AD) and vascular dementia (VaD) together contribute to almost 90 % of all dementia cases leading to major health challenges of our time with a substantial global socioeconomic burden. While in AD, the improved understanding of Amyloid beta (Aß) mismetabolism and tau hyperphosphorylation as pathophysiological hallmarks has led to significant clinical breakthroughs, similar advances in VaD are lacking. After comparing the clinical presentation, including risk factors, disease patterns, course of diseases and further diagnostic parameters for both forms of dementia, we highlight the importance of shared pathomechanisms found in AD and VaD: Endothelial damage, blood brain barrier (BBB) breakdown and hypoperfusion inducing oxidative stress and inflammation and thus trophic uncoupling in the neurovascular unit. A dysfunctional endothelium and BBB lead to the accumulation of neurotoxic molecules and Aß through impaired clearance, which in turn leads to neurodegeneration. In this context we discuss possible neuropathological parameters, which might serve as biomarkers and thus improve diagnostic accuracy or reveal targets for novel therapeutic strategies for both forms of dementia.
Collapse
Affiliation(s)
- Alisa Vollhardt
- Department of Neuroanatomy, Institute of Anatomy, Ludwigs-Maximilians-University, Pettikoferstrasse 11, 80336 Munich, Germany
| | - Lutz Frölich
- Central Institute of Mental Health, University of Heidelberg, J5, 68159 Mannheim, Germany
| | - Anna Christina Stockbauer
- Department of Neurology, LMU University Hospital, LMU Munich, Marchioninistraße 15, 81377 Munich, Germany
| | - Adrian Danek
- Department of Neurology, LMU University Hospital, LMU Munich, Marchioninistraße 15, 81377 Munich, Germany
| | - Christoph Schmitz
- Department of Neuroanatomy, Institute of Anatomy, Ludwigs-Maximilians-University, Pettikoferstrasse 11, 80336 Munich, Germany
| | - Anna-Sophia Wahl
- Department of Neuroanatomy, Institute of Anatomy, Ludwigs-Maximilians-University, Pettikoferstrasse 11, 80336 Munich, Germany; Institute for Stroke and Dementia Research (ISD), LMU University Hospital, LMU Munich, Feodor-Lynen-Strasse 17, 81377 Munich, Germany.
| |
Collapse
|
4
|
Arndt P, Pfister M, Perosa V, Mattern H, Bernal J, John AC, Dörner M, Müller P, Braun-Dullaeus RC, Garz C, Nelke C, Kokott A, Jansen R, Gliem M, Meuth SG, Henneicke S, Vielhaber S, Neumann K, Schreiber S. Risk factors and clinical significance of neurodegenerative co-pathologies in symptomatic cerebral small vessel disease. J Neurol 2025; 272:349. [PMID: 40251424 PMCID: PMC12008077 DOI: 10.1007/s00415-025-13087-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2024] [Revised: 03/04/2025] [Accepted: 04/03/2025] [Indexed: 04/20/2025]
Abstract
BACKGROUND Cerebral small vessel disease (CSVD) often coexists with neurodegenerative pathologies, yet their role remains underexplored. This study aims to determine their prevalence, risk factors, and cognitive effects in patients with deep perforator arteriopathy (DPA) or cerebral amyloid angiopathy (CAA) using the biomarker-based ATN classification. METHODS In this cross-sectional study 186 patients (median age 75 years, 41% females, 111 with probable CAA, 75 with DPA) underwent MRI for analysis of CSVD severity and etiology, and lumbar puncture for analysis of cerebrospinal fluid amyloid-β 42/40 ratio, phosphorylated-tau, total-tau and neurofilament light. ATN profiles were related to clinical characteristics, MRI markers and cognitive performance in multivariate regression models. RESULTS Among CSVD patients, 30% had normal biomarkers (A-T-N-), 33% were within the AD pathology continuum (A + T ± N ± : 47% in CAA vs. 13% in DPA, p < .001), and 37% showed non-AD pathological changes (A-T ± N + : 53% in DPA vs. 25% in CAA, p < .001). The AD pathology continuum was associated with a severe lobar hemorrhagic phenotype and cognitive impairment, while non-AD pathological change was related to CSVD severity, history of stroke and similarly cognitive impairment. Both pathological ATN profiles were further related to lower MMSE scores (A + T ± N ± : B = - 3.3, p = .006; A-T ± N + : B = - 2.7, p = .021). CONCLUSIONS Using biomarkers, this study confirms in vivo that CSVD frequently co-occurs with neurodegenerative pathologies, exerting detrimental effects on cognitive health.
Collapse
Affiliation(s)
- Philipp Arndt
- Department of Neurology, Otto-Von-Guericke University, Leipziger Str. 44, 39120, Magdeburg, Germany
- German Center for Neurodegenerative Diseases (DZNE) Within the Helmholtz Association, Magdeburg, Germany
| | - Malte Pfister
- Department of Neurology, Otto-Von-Guericke University, Leipziger Str. 44, 39120, Magdeburg, Germany
| | - Valentina Perosa
- Department of Neurology, Otto-Von-Guericke University, Leipziger Str. 44, 39120, Magdeburg, Germany
- J. Philip Kistler Stroke Research Center, Massachusetts General Hospital/Harvard Medical School, Boston, MA, USA
| | - Hendrik Mattern
- German Center for Neurodegenerative Diseases (DZNE) Within the Helmholtz Association, Magdeburg, Germany
- Biomedical Magnetic Resonance, Faculty of Natural Sciences, Otto-Von-Guericke University, Magdeburg, Germany
| | - Jose Bernal
- German Center for Neurodegenerative Diseases (DZNE) Within the Helmholtz Association, Magdeburg, Germany
- Center for Clinical Brain Sciences, The University of Edinburgh, Edinburgh, UK
| | - Anna-Charlotte John
- Department of Neurology, Otto-Von-Guericke University, Leipziger Str. 44, 39120, Magdeburg, Germany
| | - Marc Dörner
- German Center for Neurodegenerative Diseases (DZNE) Within the Helmholtz Association, Magdeburg, Germany
- Department of Consultation-Liaison-Psychiatry and Psychosomatic Medicine, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Patrick Müller
- Department of Cardiology, Otto-Von-Guericke University, Magdeburg, Germany
- German Center for Mental Health (DZPG), Magdeburg, Germany
| | | | - Cornelia Garz
- Department of Neurology, Otto-Von-Guericke University, Leipziger Str. 44, 39120, Magdeburg, Germany
- German Center for Neurodegenerative Diseases (DZNE) Within the Helmholtz Association, Magdeburg, Germany
| | - Christopher Nelke
- Department of Neurology, Heinrich-Heine-University, Düsseldorf, Germany
| | - Alma Kokott
- Department of Neurology, Heinrich-Heine-University, Düsseldorf, Germany
| | - Robin Jansen
- Department of Neurology, Heinrich-Heine-University, Düsseldorf, Germany
| | - Michael Gliem
- Department of Neurology, Heinrich-Heine-University, Düsseldorf, Germany
| | - Sven G Meuth
- Department of Neurology, Heinrich-Heine-University, Düsseldorf, Germany
| | - Solveig Henneicke
- Department of Neurology, Otto-Von-Guericke University, Leipziger Str. 44, 39120, Magdeburg, Germany
- German Center for Neurodegenerative Diseases (DZNE) Within the Helmholtz Association, Magdeburg, Germany
| | - Stefan Vielhaber
- Department of Neurology, Otto-Von-Guericke University, Leipziger Str. 44, 39120, Magdeburg, Germany
| | - Katja Neumann
- Department of Neurology, Otto-Von-Guericke University, Leipziger Str. 44, 39120, Magdeburg, Germany
| | - Stefanie Schreiber
- Department of Neurology, Otto-Von-Guericke University, Leipziger Str. 44, 39120, Magdeburg, Germany.
- German Center for Neurodegenerative Diseases (DZNE) Within the Helmholtz Association, Magdeburg, Germany.
| |
Collapse
|
5
|
Miner AE, Groh JR, Farris C, Hattiangadi S, Cui A, Brickman AM, Alshikho M, Rabinovici GD, Rosen HJ, Cobigo Y, Asken B, Nowinski CJ, Bureau S, Shahrokhi F, Tripodis Y, Ly M, Altaras C, Lenio S, Stern RA, Rosen G, Kelley H, Huber BR, Stein TD, Mez J, McKee AC, Alosco ML. Does white matter and vascular injury from repetitive head impacts lead to a novel pattern on T2 FLAIR MRI? A hypothesis proposal and call for research. Alzheimers Dement 2025; 21:e70085. [PMID: 40145364 PMCID: PMC11947747 DOI: 10.1002/alz.70085] [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: 09/12/2024] [Revised: 02/03/2025] [Accepted: 02/03/2025] [Indexed: 03/28/2025]
Abstract
The goal of this paper is to introduce the hypothesis that white matter (WM) and vascular injury are long-term consequences of repetitive head impacts (RHI) that result in a novel T2 fluid attenuated inversion recovery (FLAIR) magnetic resonance imaging pattern. A non-systematic literature review of autopsy and FLAIR studies of RHI-exposed adults was first conducted as a foundation for our hypothesis. A case series of RHI-exposed participants is presented to illustrate the unique FLAIR WM hyperintensities (WMH) pattern. Current literature shows a direct link between RHI and later-life WM/vascular neuropathologies, and that FLAIR WMH are associated with RHI, independent of modifiable vascular risk factors. Initial observations suggest a distinctive pattern of WMH in RHI-exposed participants, termed RHI-associated WMH (RHI-WMH). RHI-WMH defining features are as follows: (1) small, punctate, non-confluent, (2) spherical, and (3) proximal to the gray matter. Our hypothesis serves as a call for research to empirically validate RHI-WMH and clarify their biological and clinical correlates. HIGHLIGHTS: Repetitive head impacts (RHI) have been associated with later-life white matter (WM) and vascular neuropathologies. T2 FLAIR MRI of RHI-exposed participants reveals a potentially unique WM hyperintensity (WMH) pattern that is termed RHI-associated WMH (RHI-WMH). RHI-WMH are characterized as (1) small, punctate, and non-confluent, (2) spherical, and (3) proximal to the gray matter at an area anatomically susceptible to impact injury, such as the depths of the cortical sulci.
Collapse
|
6
|
Tao R, Wei Z, Chen X, Wang Q, Liu X, Lu Q, Zhao J, Zhou H. Retinal vascular alterations are associated with cognitive function and neuroimaging in white matter hyperintensities. Microvasc Res 2025; 158:104763. [PMID: 39566656 DOI: 10.1016/j.mvr.2024.104763] [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: 08/20/2024] [Revised: 11/10/2024] [Accepted: 11/11/2024] [Indexed: 11/22/2024]
Abstract
AIM To reveal alterations in retinal structure, vessels, and function, and their association with cognitive function and neuroimaging in white matter hyperintensities (WMH). METHODS This study enlisted WMH and age-matched healthy controls (HC). All participants underwent six different tests: magnetic resonance imaging (MRI) of the brain, the Mini-Mental State Examination (MMSE), the Montreal Cognitive Assessment (MoCA), fundus photography, optical coherence tomography (OCT), and visual field testing. Visual field can reflect the function of optic nerve and retina. The peripapillary retinal nerve fiber layer (p-RNFL) was analyzed using OCT. Image J software was employed to measure retinal vascular caliber in fundus photographs and to compute the central retinal artery equivalent (CRAE), central retinal venous equivalent (CRVE) and arteriole-to-venule ratio (AVR). RESULTS A total of 90 WMH patients and 93 HC participants. In comparison with the HC, the WMH group exhibited reduced cognitive function scores (MoCA: P < 0.001; MMSE: P < 0.001), narrower retinal arteries (P < 0.001), smaller AVR (P < 0.001) and thinner p-RNFL thickness (total: P = 0.026; temporal: P = 0.006). About visual field, both univariate and multivariate analysis showed that mean sensitivity decreased, and mean defect increased in WMH group (P < 0.05). Additionally, correlation analysis indicated a positive correlation between CRAE and AVR with MMSE and MoCA score (r = 0.424-0.57, P < 0.001) and a negative correlation with Fazekas score (CRAE: r = -0.515, P < 0.001; AVR: r = -0.554, P < 0.001), and p-RNFL was negatively correlated with Fazekas score (total p-RNFL: r = -0.192, P = 0.009; temporal p-RNFL: r = -0.217, P = 0.003). Notably, no significant correlation was found between cognitive function and p-RNFL. CONCLUSION WMH group exhibit narrower retinal arteries, smaller arteriole-to-venule ratio, damaged p-RNFL and visual function. These alterations in retinal vessels are associate with both neuroimaging and cognitive function. Our results suggest that retinal imaging could serve as a valuable instrument for evaluating WMH and provides some new approaches to study the characteristic markers of WMH.
Collapse
Affiliation(s)
- Rui Tao
- Department of Ophthalmology, Shidong Hospital Affiliated to University of Shanghai for Science and Technology, Shanghai, China
| | - Zhenyu Wei
- Department of Neurology, Shidong Hospital Affiliated to University of Shanghai for Science and Technology, Shanghai, China
| | - Xiaoxia Chen
- Department of Ophthalmology, Shidong Hospital Affiliated to University of Shanghai for Science and Technology, Shanghai, China
| | - Qian Wang
- Department of Ophthalmology, Shidong Hospital Affiliated to University of Shanghai for Science and Technology, Shanghai, China
| | - Xiuduo Liu
- Department of Ophthalmology, Shidong Hospital Affiliated to University of Shanghai for Science and Technology, Shanghai, China
| | - Qing Lu
- Department of Ophthalmology, Shidong Hospital Affiliated to University of Shanghai for Science and Technology, Shanghai, China
| | - Jie Zhao
- Department of Ophthalmology, Shidong Hospital Affiliated to University of Shanghai for Science and Technology, Shanghai, China.
| | - Hui Zhou
- Department of Neurology, Shidong Hospital Affiliated to University of Shanghai for Science and Technology, Shanghai, China.
| |
Collapse
|
7
|
Hu HY, Li HQ, Gong WK, Huang SY, Fu Y, Hu H, Dong Q, Cheng W, Tan L, Cui M, Yu JT. Microstructural white matter injury contributes to cognitive decline: Besides amyloid and tau. J Prev Alzheimers Dis 2025; 12:100037. [PMID: 39863331 DOI: 10.1016/j.tjpad.2024.100037] [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: 10/28/2024] [Revised: 12/09/2024] [Accepted: 12/10/2024] [Indexed: 01/27/2025]
Abstract
BACKGROUND Cognitive decline and the progression to Alzheimer's disease (AD) are traditionally associated with amyloid-beta (Aβ) and tau pathologies. This study aims to evaluate the relationships between microstructural white matter injury, cognitive decline and AD core biomarkers. METHODS We conducted a longitudinal study of 566 participants using peak width of skeletonized mean diffusivity (PSMD) to quantify microstructural white matter injury. The associations of PSMD with changes in cognitive functions, AD pathologies (Aβ, tau, and neurodegeneration), and volumes of AD-signature regions of interest (ROI) or hippocampus were estimated. The associations between PSMD and the incidences of clinical progression were also tested. Covariates included age, sex, education, apolipoprotein E4 status, smoking, and hypertension. RESULTS Higher PSMD was associated with greater cognitive decline (β=-0.012, P < 0.001 for Mini-Mental State Examination score; β<0, P < 0.05 for four cognitive domains) and a higher risk of clinical progression from normal cognition to mild cognitive impairment (MCI) or AD (Hazard ratio=2.11 [1.38-3.23], P < 0.001). These associations persisted independently of amyloid status. PSMD did not predict changes in Aβ or tau levels, but predicted changes in volumes of AD-signature ROI (β=-0.003, P < 0.001) or hippocampus (β=-0.002, P = 0.010). Besides, the whole-brain PSMD could predict cognitive decline better than regional PSMDs. CONCLUSIONS PSMD may be a valuable biomarker for predicting cognitive decline and clinical progression to MCI and AD, providing insights besides traditional Aβ and tau pathways. Further research could elucidate its role in clinical assessments and therapeutic strategies.
Collapse
Affiliation(s)
- He-Ying Hu
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, Qingdao, PR China.
| | - Hong-Qi Li
- Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, PR China.
| | - Wei-Kang Gong
- Institute of Science and Technology for Brain-inspired Intelligence, Fudan University, Shanghai, PR China.
| | - Shu-Yi Huang
- Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, PR China.
| | - Yan Fu
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, Qingdao, PR China.
| | - Hao Hu
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, Qingdao, PR China.
| | - Qiang Dong
- Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, PR China.
| | - Wei Cheng
- Institute of Science and Technology for Brain-inspired Intelligence, Fudan University, Shanghai, PR China.
| | - Lan Tan
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, Qingdao, PR China.
| | - Mei Cui
- Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, PR China.
| | - Jin-Tai Yu
- Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, PR China.
| |
Collapse
|
8
|
Wang Y, Zhu L, He K, Cui L, Pan F, Guan Y, He R, Xie F, Guo Q. Urinary formic acid is associated with cerebral amyloid deposition and glucose metabolism in memory clinic patients. J Alzheimers Dis 2025; 103:1102-1111. [PMID: 39791247 DOI: 10.1177/13872877241309117] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2025]
Abstract
BACKGROUND Urinary formic acid (FA) has been reported to be a biomarker for Alzheimer's disease (AD). However, the association between FA and pathological changes in memory clinic patients is currently unclear. OBJECTIVE This study aims to investigate associations between FA and pathological changes across different cognitive statuses in memory clinic patients. METHODS A cohort of patients with mild cognitive impairment (MCI-Aβ- n = 37, MCI-Aβ+ n = 33), AD dementia (n = 39), and cognitively normal subjects (CN-Aβ- n = 98, CN-Aβ+ n = 50) were included. Comprehensive neuropsychological assessment, urinary FA, AD-related plasma biomarkers, MRI scans, [18F]-flurbetapir and [18F]-FDG PET scan data were collected from all participants. RESULTS Urinary FA levels were higher in patients with MCI and AD than in CN subjects and higher in Aβ+ (CN- Aβ+, MCI-Aβ+, AD dementia) subjects than in Aβ-subjects (CN- Aβ-, MCI-Aβ-). Urinary FA was positively associated with cerebral Aβ deposition and negatively associated with glucose metabolism, both at the global level and in multiple regions of interest cortical regions in participants with different cognitive statuses. Additionally, urinary FA levels were positively correlated with the severity of white matter hyperintensities and hippocampal atrophy. Urinary FA combined with age, Mini-Mental State Examination, plasma p-tau181, and neurofilament light chain could be used to predict Aβ deposition in the brain. CONCLUSIONS Urinary FA is associated with brain pathological changes in memory clinic patients, including cerebral Aβ deposition, glucose metabolism, white matter hyperintensities, and hippocampal atrophy. It could be used as a biomarker for the early diagnosis of AD and predicting Aβ deposition.
Collapse
Affiliation(s)
- Ying Wang
- Department of Gerontology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Liangying Zhu
- Department of Gerontology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Kun He
- Department of Nuclear Medicine & PET Center, Huashan Hospital, Fudan University, Shanghai, China
| | - Liang Cui
- Department of Gerontology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Fengfeng Pan
- Department of Gerontology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yihui Guan
- Department of Nuclear Medicine & PET Center, Huashan Hospital, Fudan University, Shanghai, China
| | - Rongqiao He
- State Key Laboratory of Brain and Cognitive Sciences, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
| | - Fang Xie
- Department of Nuclear Medicine & PET Center, Huashan Hospital, Fudan University, Shanghai, China
| | - Qihao Guo
- Department of Gerontology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
9
|
Fang Z, Chen X, Zhao Y, Zhou X, Cai X, Deng J, Cheng W, Sun W, Zhuang J, Yin Y. Quantitative assessments of white matter hyperintensities and plasma biomarkers can predict cognitive impairment and cerebral microbleeds in cerebral small vessel disease patients. Neuroscience 2025; 564:41-51. [PMID: 39522933 DOI: 10.1016/j.neuroscience.2024.11.014] [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: 04/09/2024] [Revised: 10/08/2024] [Accepted: 11/07/2024] [Indexed: 11/16/2024]
Abstract
The objective of this study is to examine the efficacy of magnetic resonance imaging (MRI) features and peripheral blood biomarkers in assessing cognitive function in patients with cerebral small vessel disease (CSVD). A total of 58 CSVD patients were recruited. Six features of white matter hyperintensities (WMHs) were derived from MRI scans. Additionally, five neurodegenerative biomarkers (Aβ40, Aβ42, t-tau, p-tau181, NfL) and 13 serum inflammatory cytokines (VILIP-1, CCL2, IL-6, IL-18, TNF-α, CX3CL, sTREM-1/2, VEGF, s-RAGE, BNDF, TGF-β1, β-NGF) were quantified. Cognitive assessments were conducted using standardized neuropsychological scales. Spearman analysis revealed that the volumetric characteristics (absolute area, upper area, bottom area, absolute area percentage, upper percentage, and bottom percentage) of WMHs were negatively correlated with performance on all cognitive scale measures except the verbal fluency test (VFT) (r < -0.3, p > 0.05), while they were positively correlated with plasma neurofilament light (NFL) levels (r > 0.4, p < 0.05). Additionally, serum tumor necrosis factor-α (TNF-α) and soluble receptor for advanced glycation end-products (s-RAGE) showed significant correlations with scales of speech function. An integrated model incorporating WMHs features, neurodegenerative biomarkers, and neuroinflammatory markers was developed, demonstrating high predictive accuracy for cognitive impairment with an area under the curve (AUC) of 0.95 (accuracy 0.88, sensitivity 0.87, specificity 0.89). Another integrated model that includes features of WMHs and inflammatory cytokines for predicting cerebral microbleeds (CMBs) achieved an AUC of 0.95 (accuracy 0.88, sensitivity 0.82, specificity 0.92). Our findings suggest that these markers have the potential to be used for the early detection of cognitive decline and CMBs in patients with CSVD.
Collapse
Affiliation(s)
- Zhuo Fang
- Department of Data and Analytics, WuXi Diagnostics Innovation Research Institute, 200131, Shanghai, China; Danaher China Corporation, 200335, Shanghai, China
| | - Xiaohan Chen
- Department of Neurology, Second Affiliated Hospital of Naval Medical University, 200003, Shanghai, China
| | - Yike Zhao
- Department of Data and Analytics, WuXi Diagnostics Innovation Research Institute, 200131, Shanghai, China
| | - Xinrui Zhou
- Department of Data and Analytics, WuXi Diagnostics Innovation Research Institute, 200131, Shanghai, China
| | - Xiao Cai
- Department of Data and Analytics, WuXi Diagnostics Innovation Research Institute, 200131, Shanghai, China
| | - Jiale Deng
- Department of Data and Analytics, WuXi Diagnostics Innovation Research Institute, 200131, Shanghai, China
| | - Wenbin Cheng
- Department of Neurology, Second Affiliated Hospital of Naval Medical University, 200003, Shanghai, China
| | - Wenjing Sun
- Department of Neurology, Second Affiliated Hospital of Naval Medical University, 200003, Shanghai, China
| | - Jianhua Zhuang
- Department of Neurology, Second Affiliated Hospital of Naval Medical University, 200003, Shanghai, China
| | - You Yin
- Department of Neurology, Second Affiliated Hospital of Naval Medical University, 200003, Shanghai, China; Department of Neurology, Shanghai East Hospital, School of Medicine, Tongji University, 200120, Shanghai, China.
| |
Collapse
|
10
|
Ruiz-Uribe NE, Manser P, Butcher B, Li Y, Blendstrup M, Baker S, Sanabria Bohorquez S, Teng E. Cross-sectional and prognostic associations of baseline [ 18F]GTP1 tau PET signal and white matter lesion volumes for cognitive and functional decline in prodromal-to-mild Alzheimer's disease. J Alzheimers Dis 2025; 103:465-475. [PMID: 39801050 DOI: 10.1177/13872877241302497] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2025]
Abstract
BACKGROUND In Alzheimer's disease (AD), tau and white matter lesion pathology are associated with clinical severity and subsequent decline, but their relative relationships with clinical assessments remain uncertain. OBJECTIVE To examine cross-sectional and prognostic associations between baseline [18F]GTP1 tau positron emission tomography (PET) standardized uptake value ratio (SUVRs) and T1 white matter hypointensity (WMHypo) volumes with clinical indices. METHODS We analyzed participants with biomarker-confirmed prodromal (n = 127) or mild (n = 233) AD with baseline [18F]GTP1 tau PET and MRI and longitudinal Clinical Dementia Rating-Sum of Boxes (CDR-SB), 13-item version of the Alzheimer's Disease Assessment Scale-Cognitive Subscale (ADAS-Cog13), Repeatable Battery for the Assessment of Neuropsychological Status (RBANS), Mini-Mental Status Examination (MMSE), and Alzheimer's Disease Cooperative Study-Activities of Daily Living (ADCS-ADL) data. RESULTS Higher baseline [18F]GTP1 SUVRs were independently associated with poorer baseline performance and faster rates of subsequent decline on all five clinical outcome measures. Higher baseline WMHypo volumes were independently associated with poorer baseline performance on the CDR-SB, ADAS-Cog13, RBANS, and MMSE and faster rates of subsequent decline on the CDR-SB and ADCS-ADL. CONCLUSIONS The independent associations of tau and white matter lesion pathology with clinical decline in AD suggest future prognostic models should include both imaging modalities.
Collapse
Affiliation(s)
- Nancy E Ruiz-Uribe
- Genentech, Inc., South San Francisco, CA, USA
- Department of Biomedical Engineering, Cornell University, Ithaca, NY, USA
| | - Paul Manser
- Genentech, Inc., South San Francisco, CA, USA
| | | | - Yihao Li
- Genentech, Inc., South San Francisco, CA, USA
| | | | - Suzanne Baker
- Genentech, Inc., South San Francisco, CA, USA
- Department of Molecular Biophysics and Integrated Bioimaging, Lawrence Berkeley National Laboratory, Berkeley, CA, USA
| | | | - Edmond Teng
- Genentech, Inc., South San Francisco, CA, USA
| |
Collapse
|
11
|
Li X, Wang Z, Zhang H, Zhao W, Ji Q, Zhang X, Jia X, Bai G, Pan Y, Wu T, Yin B, Shi L, Li Z, Ding J, Zhang J, Salat DH, Bai L. Tract-Specific White Matter Hyperintensities Disrupt Brain Networks and Associated With Cognitive Impairment in Mild Traumatic Brain Injury. Hum Brain Mapp 2024; 45:e70050. [PMID: 39611374 PMCID: PMC11605479 DOI: 10.1002/hbm.70050] [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: 07/04/2024] [Revised: 09/16/2024] [Accepted: 09/30/2024] [Indexed: 11/30/2024] Open
Abstract
Traumatic brain injury (TBI) is considered to initiate cerebrovascular pathology, involving in the development of multiple forms of neurodegeneration. However, it is unknown the relationships between imaging marker of cerebrovascular injury (white matter hyperintensity, WMH), its load on white matter tract and disrupted brain dynamics with cognitive function in mild TBI (mTBI). MRI data and neuropsychological assessments were collected from 85 mTBI patients and 52 healthy controls. Between-group difference was conducted for the tract-specific WMH volumes, white matter integrity, and dynamic brain connectivity (i.e., fractional occupancies [%], dwell times [seconds], and state transitions). Regression analysis was used to examine associations between white matter damage, brain dynamics, and cognitive function. Increased WMH volumes induced by mTBI within the thalamic radiation and corpus callosum were highest among all tract fibers, and related with altered fractional anisotropy (FA) within the same tracts. Clustering identified two brain states, segregated state characterized by the sparse inter-independent component connections, and default mode network (DMN)-centered integrated state with strongly internetwork connections between DMN and other networks. In mTBI, higher WMH loads contributed to the longer dwell time and larger fractional occupancies in DMN-centered integrated state. Every 1 mL increase in WMH volume within the left thalamic radiation was associated with a 47% increase fractional occupancies, and contributed to 65.6 s delay in completion of cognitive processing speed test. Our study provided the first evidence for the structural determinants (i.e., small vessel lesions) that mediate the spatiotemporal brain dynamics to cognitive impairments in mTBI.
Collapse
Affiliation(s)
- Xuan Li
- The Key Laboratory of Biomedical Information Engineering, Ministry of Education, Department of Biomedical Engineering, School of Life Science and TechnologyXi'an Jiaotong UniversityXi'anChina
| | - Zhuonan Wang
- PET‐CT Center, The First Affiliated Hospital of Xi'an Jiaotong UniversityXi'anChina
| | - Haonan Zhang
- The Key Laboratory of Biomedical Information Engineering, Ministry of Education, Department of Biomedical Engineering, School of Life Science and TechnologyXi'an Jiaotong UniversityXi'anChina
| | - Wenpu Zhao
- The Key Laboratory of Biomedical Information Engineering, Ministry of Education, Department of Biomedical Engineering, School of Life Science and TechnologyXi'an Jiaotong UniversityXi'anChina
| | - Qiuyu Ji
- The Key Laboratory of Biomedical Information Engineering, Ministry of Education, Department of Biomedical Engineering, School of Life Science and TechnologyXi'an Jiaotong UniversityXi'anChina
| | - Xiang Zhang
- The Key Laboratory of Biomedical Information Engineering, Ministry of Education, Department of Biomedical Engineering, School of Life Science and TechnologyXi'an Jiaotong UniversityXi'anChina
| | - Xiaoyan Jia
- The Key Laboratory of Biomedical Information Engineering, Ministry of Education, Department of Biomedical Engineering, School of Life Science and TechnologyXi'an Jiaotong UniversityXi'anChina
| | - Guanghui Bai
- Department of RadiologyThe Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical UniversityWenzhouChina
| | - Yizhen Pan
- The Key Laboratory of Biomedical Information Engineering, Ministry of Education, Department of Biomedical Engineering, School of Life Science and TechnologyXi'an Jiaotong UniversityXi'anChina
| | - Tingting Wu
- The Key Laboratory of Biomedical Information Engineering, Ministry of Education, Department of Biomedical Engineering, School of Life Science and TechnologyXi'an Jiaotong UniversityXi'anChina
| | - Bo Yin
- Department of NeurosurgeryThe Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical UniversityWenzhouChina
| | - Lei Shi
- The Key Laboratory of Biomedical Information Engineering, Ministry of Education, Department of Biomedical Engineering, School of Life Science and TechnologyXi'an Jiaotong UniversityXi'anChina
- Department of Clinical LaboratoryShuguang Hospital Affiliated to Shanghai University of Chinese Traditional MedicineShanghaiChina
| | - Zhiqi Li
- The Key Laboratory of Biomedical Information Engineering, Ministry of Education, Department of Biomedical Engineering, School of Life Science and TechnologyXi'an Jiaotong UniversityXi'anChina
| | - Jierui Ding
- The Key Laboratory of Biomedical Information Engineering, Ministry of Education, Department of Biomedical Engineering, School of Life Science and TechnologyXi'an Jiaotong UniversityXi'anChina
| | - Jie Zhang
- Department of Radiation Medicine, School of Preventive MedicineAir Force Medical UniversityXi'anChina
| | - David H. Salat
- Athinoula A. Martinos Center for Biomedical Imaging, Department of RadiologyMassachusetts General HospitalCharlestownMassachusettsUSA
| | - Lijun Bai
- The Key Laboratory of Biomedical Information Engineering, Ministry of Education, Department of Biomedical Engineering, School of Life Science and TechnologyXi'an Jiaotong UniversityXi'anChina
| |
Collapse
|
12
|
Xu Z, He S, Begum MM, Han X. Myelin Lipid Alterations in Neurodegenerative Diseases: Landscape and Pathogenic Implications. Antioxid Redox Signal 2024; 41:1073-1099. [PMID: 39575748 PMCID: PMC11971557 DOI: 10.1089/ars.2024.0676] [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: 05/14/2024] [Revised: 10/21/2024] [Accepted: 10/22/2024] [Indexed: 12/14/2024]
Abstract
Significance: Lipids, which constitute the highest portion (over 50%) of brain dry mass, are crucial for brain integrity, energy homeostasis, and signaling regulation. Emerging evidence revealed that lipid profile alterations and abnormal lipid metabolism occur during normal aging and in different forms of neurodegenerative diseases. Moreover, increasing genome-wide association studies have validated new targets on lipid-associated pathways involved in disease development. Myelin, the protective sheath surrounding axons, is crucial for efficient neural signaling transduction. As the primary site enriched with lipids, impairments of myelin are increasingly recognized as playing significant and complex roles in various neurodegenerative diseases, beyond simply being secondary effects of neuronal loss. Recent Advances: With advances in the lipidomics field, myelin lipid alterations and their roles in contributing to or reflecting the progression of diseases, including Alzheimer's disease, Parkinson's disease, Huntington's disease, amyotrophic lateral sclerosis, multiple sclerosis, and others, have recently caught great attention. Critical Issues: This review summarizes recent findings of myelin lipid alterations in the five most common neurodegenerative diseases and discusses their implications in disease pathogenesis. Future Directions: By highlighting myelin lipid abnormalities in neurodegenerative diseases, this review aims to encourage further research focused on lipids and the development of new lipid-oriented therapeutic approaches in this area. Antioxid. Redox Signal. 00, 000-000.
Collapse
Affiliation(s)
- Ziying Xu
- Sam and Ann Barshop Institute for Longevity and Aging Studies, UT Health San Antonio, San Antonio, Texas, USA
| | - Sijia He
- Sam and Ann Barshop Institute for Longevity and Aging Studies, UT Health San Antonio, San Antonio, Texas, USA
| | - Mst Marium Begum
- Sam and Ann Barshop Institute for Longevity and Aging Studies, UT Health San Antonio, San Antonio, Texas, USA
| | - Xianlin Han
- Sam and Ann Barshop Institute for Longevity and Aging Studies, UT Health San Antonio, San Antonio, Texas, USA
- Department of Medicine, UT Health San Antonio, San Antonio, Texas, USA
| |
Collapse
|
13
|
Huan S, Zhang S, Wang L, Liu C, Xu J, Ge Y, Huang Z, Cai W, Cao Y, You S. Association between fibrinogen and white matter lesions and cerebral atrophy in patients with acute ischemic stroke. J Stroke Cerebrovasc Dis 2024; 33:108008. [PMID: 39265859 DOI: 10.1016/j.jstrokecerebrovasdis.2024.108008] [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: 04/17/2024] [Revised: 08/20/2024] [Accepted: 09/09/2024] [Indexed: 09/14/2024] Open
Abstract
BACKGROUND Inflammation is a potential mechanism underlying the development of white matter lesions (WMLs) and cerebral atrophy. We aimed to investigate the relationship of fibrinogen levels with WMLs and cerebral atrophy in patients with acute ischemic stroke (AIS). METHODS A total of 701 AIS patients were enrolled. Participants were divided into four groups according to the quartiles of fibrinogen levels: Q1 < 2.58 g/L, Q2: 2.58-3.12 g/L, Q3: 3.12-3.67 g/L, Q4: ≥ 3.67 g/L. White matter hyperintensity (WMH), periventricular hyperintensity (PVH) and deep white matter hyperintensity (DWMH) were defined according to the Fazekas scale. Cerebral atrophy was defined according to global cortical atrophy scores. Univariate and multivariate logistic regression were used to explore the relationship of fibrinogen levels and WMHs, PVH, DWMH and cerebral atrophy. RESULTS Among 701 AIS patients, 498 (71.0 %), 425 (60.6 %), 442 (63.1 %), and 560 (79.9 %) had WMHs, PVH, DWMH and cerebral atrophy, respectively. After adjustment for potential covariates, the highest fibrinogen quartiles were significantly associated with increased risk of WMHs (odds ratio [OR] 1.97, 95 % confidence intervals [CI] 1.10-3.50), PVH (OR 1.85, 95 % CI 1.08-3.16) and cerebral atrophy (OR 2.53, 95 % CI 1.19-5.40) but not DWMH (OR 1.37 95 % CI 0.81-2.31) compared with the lowest fibrinogen quartile. Moreover, the association between elevated fibrinogen levels and the risk of WMLs and cerebral atrophy remained significant as continuous variables. CONCLUSIONS Increased baseline fibrinogen levels were independently associated with WMHs, PVH and cerebral atrophy in patients with ischemic stroke. Fibrinogen could be the potential blood biomarker of WMLs and cerebral atrophy.
Collapse
Affiliation(s)
- Suqiu Huan
- Department of Neurology and Suzhou Clinical Research Center of Neurological Disease, The Second Affiliated Hospital of Soochow University, Suzhou, China.
| | - Shiya Zhang
- Department of Neurology and Suzhou Clinical Research Center of Neurological Disease, The Second Affiliated Hospital of Soochow University, Suzhou, China.
| | - Lixuan Wang
- Department of Neurology and Suzhou Clinical Research Center of Neurological Disease, The Second Affiliated Hospital of Soochow University, Suzhou, China; Department of Neurology, Honghui Hospital, Xi'an Jiaotong University, Xi'an, China.
| | - Chenchen Liu
- Department of Medical Iconography, The Second Affiliated Hospital of Soochow University, Suzhou, China.
| | - Jiaping Xu
- Department of Neurology and Suzhou Clinical Research Center of Neurological Disease, The Second Affiliated Hospital of Soochow University, Suzhou, China.
| | - Yilun Ge
- Department of Neurology and Suzhou Clinical Research Center of Neurological Disease, The Second Affiliated Hospital of Soochow University, Suzhou, China.
| | - Zhichao Huang
- Department of Neurology and Suzhou Clinical Research Center of Neurological Disease, The Second Affiliated Hospital of Soochow University, Suzhou, China.
| | - Wu Cai
- Department of Medical Iconography, The Second Affiliated Hospital of Soochow University, Suzhou, China.
| | - Yongjun Cao
- Department of Neurology and Suzhou Clinical Research Center of Neurological Disease, The Second Affiliated Hospital of Soochow University, Suzhou, China.
| | - Shoujiang You
- Department of Neurology and Suzhou Clinical Research Center of Neurological Disease, The Second Affiliated Hospital of Soochow University, Suzhou, China.
| |
Collapse
|
14
|
Bernal J, Menze I, Yakupov R, Peters O, Hellmann-Regen J, Freiesleben SD, Priller J, Spruth EJ, Altenstein S, Schneider A, Fliessbach K, Wiltfang J, Schott BH, Jessen F, Rostamzadeh A, Glanz W, Incesoy EI, Buerger K, Janowitz D, Ewers M, Perneczky R, Rauchmann BS, Teipel S, Kilimann I, Laske C, Sodenkamp S, Spottke A, Esser A, Lüsebrink F, Dechent P, Hetzer S, Scheffler K, Schreiber S, Düzel E, Ziegler G. Longitudinal evidence for a mutually reinforcing relationship between white matter hyperintensities and cortical thickness in cognitively unimpaired older adults. Alzheimers Res Ther 2024; 16:240. [PMID: 39465440 PMCID: PMC11520063 DOI: 10.1186/s13195-024-01606-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2024] [Accepted: 10/18/2024] [Indexed: 10/29/2024]
Abstract
BACKGROUND For over three decades, the concomitance of cortical neurodegeneration and white matter hyperintensities (WMH) has sparked discussions about their coupled temporal dynamics. Longitudinal studies supporting this hypothesis nonetheless remain scarce. METHODS We applied global and regional bivariate latent growth curve modelling to determine the extent to which WMH and cortical thickness were interrelated over a four-year period. For this purpose, we leveraged longitudinal MRI data from 451 cognitively unimpaired participants (DELCODE; median age 69.71 [IQR 65.51, 75.50] years; 52.32% female). Participants underwent MRI sessions annually over a four-year period (1815 sessions in total, with roughly four MRI sessions per participant). We adjusted all models for demographics and cardiovascular risk. RESULTS Our findings were three-fold. First, larger WMH volumes were linked to lower cortical thickness (σ = -0.165, SE = 0.047, Z = -3.515, P < 0.001). Second, individuals with higher WMH volumes experienced more rapid cortical thinning (σ = -0.226, SE = 0.093, Z = -2.443, P = 0.007), particularly in temporal, cingulate, and insular regions. Similarly, those with lower initial cortical thickness had faster WMH progression (σ = -0.141, SE = 0.060, Z = -2.336, P = 0.009), with this effect being most pronounced in temporal, cingulate, and insular cortices. Third, faster WMH progression was associated with accelerated cortical thinning (σ = -0.239, SE = 0.139, Z = -1.710, P = 0.044), particularly in frontal, occipital, and insular cortical regions. CONCLUSIONS Our study suggests that cortical thinning and WMH progression could be mutually reinforcing rather than parallel, unrelated processes, which become entangled before cognitive deficits are detectable. TRIAL REGISTRATION German Clinical Trials Register (DRKS00007966, 04/05/2015).
Collapse
Affiliation(s)
- Jose Bernal
- Institute of Cognitive Neurology and Dementia Research, Otto-Von-Guericke University Magdeburg, Magdeburg, Germany.
- German Centre for Neurodegenerative Diseases (DZNE), Magdeburg, Germany.
- Centre for Clinical Brain Sciences, the University of Edinburgh, Edinburgh, UK.
- UK Dementia Research Institute Centre at the University of Edinburgh, Edinburgh, UK.
| | - Inga Menze
- Institute of Cognitive Neurology and Dementia Research, Otto-Von-Guericke University Magdeburg, Magdeburg, Germany
- German Centre for Neurodegenerative Diseases (DZNE), Magdeburg, Germany
| | - Renat Yakupov
- Institute of Cognitive Neurology and Dementia Research, Otto-Von-Guericke University Magdeburg, Magdeburg, Germany
- German Centre for Neurodegenerative Diseases (DZNE), Magdeburg, Germany
| | - Oliver Peters
- German Centre for Neurodegenerative Diseases (DZNE), Berlin, Germany
- Charité - Universitätsmedizin Berlin, Institute of Psychiatry and Psychotherapy, Berlin, Germany
| | - Julian Hellmann-Regen
- German Centre for Neurodegenerative Diseases (DZNE), Berlin, Germany
- Charité - Universitätsmedizin Berlin, Department of Psychiatry and Neurosciences, Campus Benjamin Franklin, Berlin, Germany
- German Centre for Mental Health (DZPG), Berlin, Germany
| | - Silka Dawn Freiesleben
- German Centre for Neurodegenerative Diseases (DZNE), Berlin, Germany
- Charité - Universitätsmedizin Berlin, Institute of Psychiatry and Psychotherapy, Berlin, Germany
| | - Josef Priller
- UK Dementia Research Institute Centre at the University of Edinburgh, Edinburgh, UK
- German Centre for Neurodegenerative Diseases (DZNE), Berlin, Germany
- Department of Psychiatry and Psychotherapy, Charité, Berlin, Germany
- School of Medicine, Department of Psychiatry and Psychotherapy, Technical University of Munich, Munich, Germany
| | - Eike Jakob Spruth
- German Centre for Neurodegenerative Diseases (DZNE), Berlin, Germany
- Department of Psychiatry and Psychotherapy, Charité, Berlin, Germany
| | - Slawek Altenstein
- German Centre for Neurodegenerative Diseases (DZNE), Berlin, Germany
- Department of Psychiatry and Psychotherapy, Charité, Berlin, Germany
| | - Anja Schneider
- German Centre for Neurodegenerative Diseases (DZNE), Bonn, Germany
- Department of Old Age Psychiatry and Cognitive Disorders, University Hospital Bonn and University of Bonn, Bonn, Germany
| | - Klaus Fliessbach
- German Centre for Neurodegenerative Diseases (DZNE), Bonn, Germany
- Department of Old Age Psychiatry and Cognitive Disorders, University Hospital Bonn and University of Bonn, Bonn, Germany
| | - Jens Wiltfang
- German Centre for Neurodegenerative Diseases (DZNE), Göttingen, Germany
- Department of Psychiatry and Psychotherapy, University Medical Centre Göttingen, University of Göttingen, Göttingen, Germany
- Neurosciences and Signalling Group, Institute of Biomedicine (iBiMED), Department of Medical Sciences, University of Aveiro, Aveiro, Portugal
| | - Björn H Schott
- German Centre for Neurodegenerative Diseases (DZNE), Göttingen, Germany
- Department of Psychiatry and Psychotherapy, University Medical Centre Göttingen, University of Göttingen, Göttingen, Germany
- Leibniz Institute for Neurobiology, Brenneckestr. 6, 39118, Magdeburg, Germany
| | - Frank Jessen
- German Centre for Neurodegenerative Diseases (DZNE), Bonn, Germany
- Department of Psychiatry, Medical Faculty, University of Cologne, Cologne, Germany
- Excellence Cluster On Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Cologne, Germany
| | - Ayda Rostamzadeh
- Department of Psychiatry, Medical Faculty, University of Cologne, Cologne, Germany
| | - Wenzel Glanz
- German Centre for Neurodegenerative Diseases (DZNE), Magdeburg, Germany
| | - Enise I Incesoy
- Institute of Cognitive Neurology and Dementia Research, Otto-Von-Guericke University Magdeburg, Magdeburg, Germany
- German Centre for Neurodegenerative Diseases (DZNE), Magdeburg, Germany
- Department for Psychiatry and Psychotherapy, University Clinic Magdeburg, Magdeburg, Germany
| | - Katharina Buerger
- German Centre for Neurodegenerative Diseases (DZNE), Munich, Germany
- Institute for Stroke and Dementia Research (ISD), University Hospital, LMU Munich, Munich, Germany
| | - Daniel Janowitz
- Institute for Stroke and Dementia Research (ISD), University Hospital, LMU Munich, Munich, Germany
| | - Michael Ewers
- German Centre for Neurodegenerative Diseases (DZNE), Munich, Germany
- Institute for Stroke and Dementia Research (ISD), University Hospital, LMU Munich, Munich, Germany
| | - Robert Perneczky
- German Centre for Neurodegenerative Diseases (DZNE), Munich, Germany
- Department of Psychiatry and Psychotherapy, University Hospital, LMU Munich, Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy) Munich, Munich, Germany
- Ageing Epidemiology Research Unit (AGE), School of Public Health, Imperial College London, London, UK
| | - Boris-Stephan Rauchmann
- Department of Psychiatry and Psychotherapy, University Hospital, LMU Munich, Munich, Germany
- Sheffield Institute for Translational Neuroscience (SITraN), University of Sheffield, Sheffield, UK
- Department of Neuroradiology, University Hospital LMU, Munich, Germany
| | - Stefan Teipel
- German Centre for Neurodegenerative Diseases (DZNE), Rostock, Germany
- Department of Psychosomatic Medicine, Rostock University Medical Centre, Rostock, Germany
| | - Ingo Kilimann
- German Centre for Neurodegenerative Diseases (DZNE), Rostock, Germany
- Department of Psychosomatic Medicine, Rostock University Medical Centre, Rostock, Germany
| | - Christoph Laske
- German Centre for Neurodegenerative Diseases (DZNE), Tübingen, Germany
- Section for Dementia Research, Hertie Institute for Clinical Brain Research and Department of Psychiatry and Psychotherapy, University of Tübingen, Tübingen, Germany
| | - Sebastian Sodenkamp
- German Centre for Neurodegenerative Diseases (DZNE), Tübingen, Germany
- Department of Psychiatry and Psychotherapy, University of Tübingen, Tübingen, Germany
| | - Annika Spottke
- German Centre for Neurodegenerative Diseases (DZNE), Bonn, Germany
- Department of Neurology, University of Bonn, Bonn, Germany
| | - Anna Esser
- German Centre for Neurodegenerative Diseases (DZNE), Bonn, Germany
| | - Falk Lüsebrink
- German Centre for Neurodegenerative Diseases (DZNE), Magdeburg, Germany
| | - Peter Dechent
- Department of Cognitive Neurology, MR-Research in Neurosciences, Georg-August-University, Göttingen, Germany
| | - Stefan Hetzer
- Berlin Centre for Advanced Neuroimaging, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Klaus Scheffler
- Department for Biomedical Magnetic Resonance, University of Tübingen, Tübingen, Germany
| | - Stefanie Schreiber
- German Centre for Neurodegenerative Diseases (DZNE), Magdeburg, Germany
- Department of Neurology, University Hospital Magdeburg, Magdeburg, Germany
| | - Emrah Düzel
- Institute of Cognitive Neurology and Dementia Research, Otto-Von-Guericke University Magdeburg, Magdeburg, Germany
- German Centre for Neurodegenerative Diseases (DZNE), Magdeburg, Germany
| | - Gabriel Ziegler
- Institute of Cognitive Neurology and Dementia Research, Otto-Von-Guericke University Magdeburg, Magdeburg, Germany
- German Centre for Neurodegenerative Diseases (DZNE), Magdeburg, Germany
| |
Collapse
|
15
|
Lan H, Qiu W, Lei X, Xu Z, Yu J, Wang H. Deep medullary vein abnormalities impact white matter hyperintensity volume through increases in interstitial free water. BMC Neurol 2024; 24:405. [PMID: 39433983 PMCID: PMC11492461 DOI: 10.1186/s12883-024-03921-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2024] [Accepted: 10/14/2024] [Indexed: 10/23/2024] Open
Abstract
BACKGROUND Our intent was to explore the mediating role of interstitial free water (FW) linking deep medullary vein (DMV) score to white matter hyperintensity (WMH) volume. METHODS Our research team conducted a forward-looking analysis of initial clinical and imaging information gathered from 125 patients with cerebral small vessel disease. We identified six anatomic DMV regions on susceptibility weighted imaging (SWI) studies. Each region earned a score of 0-3, determined by the visual conditions of vessels, summing all six to generate a DMV score. We utilized fluid-attenuated inversion recovery (FLAIR) sequences to measure the volume of WMH. Additionally, we employed diffusion tensor imaging (DTI) to assess FW value. RESULTS DMV score significantly positively correlated with FW value and with WMH volume (p < 0.05), and value of FW positively correlated with WMH volume (p < 0.05). The indirect effect of DMV score on WMH volume was mediated by FW (β = 0.281, 95% confidence interval [CI]: 0.178-0.388), whether adjusted for age and gender (β = 0.142, 95% CI: 0.058-0.240) or for age, gender and vascular risk factors (β = 0.141, 95% CI: 0.054-0.249). CONCLUSION DMV score correlate with WMH volume by virtue of FW increases in white matter.
Collapse
Affiliation(s)
- Haiyuan Lan
- Department of Radiology, Lishui Hospital of Traditional Chinese Medicine affiliated with Zhejiang Chinese Medical University, Lishui, 323000, China
| | - Weiwen Qiu
- Department of Neurology, Lishui Hospital of Traditional Chinese Medicine affiliated with Zhejiang Chinese Medical University, Lishui, 323000, China
| | - Xinjun Lei
- Department of Radiology, Lishui Hospital of Traditional Chinese Medicine affiliated with Zhejiang Chinese Medical University, Lishui, 323000, China
| | - Zhihua Xu
- Department of Radiology, Tongde Hospital of Zhejiang Province, Hangzhou, 310000, China
| | - Jie Yu
- Department of Radiology, Lishui Hospital of Traditional Chinese Medicine affiliated with Zhejiang Chinese Medical University, Lishui, 323000, China
| | - Huimei Wang
- Department of Neurology, Lishui People's Hospital, Lishui, 323000, China.
| |
Collapse
|
16
|
Chatterjee A, Lee S, Diaz V, Saloner R, Sanderson-Cimino M, deCarli C, Maillard P, Hinman J, Vossel K, Casaletto KB, Staffaroni AM, Paolillo EW, Kramer JH. Associations of cerebrovascular disease and Alzheimer's disease pathology with cognitive decline: Analysis of the National Alzheimer's Coordinating Center Uniform Data Set. Neurobiol Aging 2024; 142:1-7. [PMID: 39024720 DOI: 10.1016/j.neurobiolaging.2024.06.002] [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: 10/21/2023] [Revised: 06/10/2024] [Accepted: 06/13/2024] [Indexed: 07/20/2024]
Abstract
Cerebrovascular disease (CVD) and Alzheimer's disease (AD) often co-occur and may impact specific cognitive domains. This study's goal was to determine effects of CVD and AD burden on cross-sectional and longitudinal executive function (EF) and memory in older adults. Longitudinally followed participants from the National Alzheimer Coordinating Center database (n = 3342) were included. Cognitive outcomes were EF and memory composite scores. Baseline CVD presence was defined by moderate-to-severe white matter hyperintensities or lacunar infarct on MRI. Baseline AD pathology was defined by amyloid positivity via PET or CSF. Linear mixed models examined effects of CVD, AD, and time on cognitive outcomes, controlling for sex, education, baseline age, MoCA score, and total number of study visits. At baseline, CVD associated with lower EF (p < 0.001), while AD associated with lower EF and memory (ps < 0.001). Longitudinally only AD associated with faster declines in memory and EF (ps < 0.001). These results extend our understanding of CVD and AD pathology, highlighting that CVD does not necessarily indicate accelerated decline.
Collapse
Affiliation(s)
- Ankita Chatterjee
- Memory and Aging Center, Department of Neurology, Weill Institute for Neurosciences, University of California, San Francisco, USA.
| | - Shannon Lee
- Memory and Aging Center, Department of Neurology, Weill Institute for Neurosciences, University of California, San Francisco, USA
| | - Valentina Diaz
- Memory and Aging Center, Department of Neurology, Weill Institute for Neurosciences, University of California, San Francisco, USA
| | - Rowan Saloner
- Memory and Aging Center, Department of Neurology, Weill Institute for Neurosciences, University of California, San Francisco, USA
| | - Mark Sanderson-Cimino
- Memory and Aging Center, Department of Neurology, Weill Institute for Neurosciences, University of California, San Francisco, USA
| | - Charles deCarli
- Department of Neurology, University of California, Davis, USA
| | | | - Jason Hinman
- Mary S. Easton Center for Alzheimer's Research and Care, Department of Neurology, David Geffen School of Medicine, University of California, Los Angeles, USA
| | - Keith Vossel
- Mary S. Easton Center for Alzheimer's Research and Care, Department of Neurology, David Geffen School of Medicine, University of California, Los Angeles, USA
| | - Kaitlin B Casaletto
- Memory and Aging Center, Department of Neurology, Weill Institute for Neurosciences, University of California, San Francisco, USA
| | - Adam M Staffaroni
- Memory and Aging Center, Department of Neurology, Weill Institute for Neurosciences, University of California, San Francisco, USA
| | - Emily W Paolillo
- Memory and Aging Center, Department of Neurology, Weill Institute for Neurosciences, University of California, San Francisco, USA
| | - Joel H Kramer
- Memory and Aging Center, Department of Neurology, Weill Institute for Neurosciences, University of California, San Francisco, USA
| |
Collapse
|
17
|
Yi F, Wang J, Lin M, Li B, Han S, Wang S, Jin Y, Hu N, Chen Y, Shang X. Correspondence between white matter hyperintensities and regional grey matter volumes in Alzheimer's disease. Front Aging Neurosci 2024; 16:1429098. [PMID: 39351014 PMCID: PMC11439820 DOI: 10.3389/fnagi.2024.1429098] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Accepted: 08/30/2024] [Indexed: 10/04/2024] Open
Abstract
Objective White matter hyperintensities (WMH) are the most common neuroimaging manifestation of cerebral small vessel disease, and is frequently observed in Alzheimer's disease (AD). This study aimed to investigate the relationship between WMH and cognition and to verify the mediation of grey matter atrophy in this relationship. Methods The diffusion tensor imaging (DTI) technique analyses white matter fiber tract to assess white matter integrity. Voxel-based morphometry was applied to measure the grey matter volume (GMV). A linear regression model was applied to examine the associations between WMH and GMV, and mediation analyses was performed to determine the mediating role of regional GMV in the effect of WMH on cognitive function. Results Compared to the HC group, AD group have 8 fiber tract fractional anisotropy (FA) decreased and 16 fiber tract mean diffusivity (MD) increased. Compared to AD without WMH, AD with high WMH had 9 fiber tracts FA decreased and 13 fiber tracts MD increased. High WMH volume was negatively correlated with GMV in the frontal-parietal region. Low WMH volume was also negatively correlated with GMV except for the three regions (right angular gyrus, right superior frontal gyrus and right middle/inferior parietal gyrus), where GMV was positively correlated. Mediation analysis showed that the association between WMH and executive function or episodic memory were mediated by GMV in the frontal-parietal region. Conclusion Damage to white matter integrity was more severe in AD with WMH. Differential changes in DTI metrics may be caused by progressive myelin and axonal damage. There was a negative correlation between WMH and grey matter atrophy in frontal-parietal regions in a volume-dependent manner. This study indicates the correspondence between WMH volume and GMV in cognition, and GMV being a key modulator between WMH and cognition in AD. This result will contribute to understanding the progression of the disease process and applying targeted therapeutic intervention in the earlier stage to delay neurodegenerative changes in frontal-parietal regions to achieve better treatment outcomes and affordability.
Collapse
Affiliation(s)
- Fangyuan Yi
- Department of Neurology, The First Affiliated Hospital of China Medical University, Shenyang, China
- Department of Neurology, Jin Qiu Hospital of Liaoning Province (Geriatric Hospital of Liaoning Province), Shenyang, China
| | - Jirui Wang
- Department of Neurology, The First Affiliated Hospital of China Medical University, Shenyang, China
| | - Meiqing Lin
- Department of Neurology, The First Affiliated Hospital of China Medical University, Shenyang, China
| | - Baizhu Li
- Department of Neurology, The First Affiliated Hospital of China Medical University, Shenyang, China
| | - Shiyu Han
- Department of Neurology, The First Affiliated Hospital of China Medical University, Shenyang, China
| | - Shan Wang
- Department of Neurology, The First Affiliated Hospital of China Medical University, Shenyang, China
| | - Yingbin Jin
- Department of Neurology, The First Affiliated Hospital of China Medical University, Shenyang, China
| | - Ning Hu
- Department of Neurology, The First Affiliated Hospital of China Medical University, Shenyang, China
| | - Yutong Chen
- Department of Neurology, The First Affiliated Hospital of China Medical University, Shenyang, China
| | - Xiuli Shang
- Department of Neurology, The First Affiliated Hospital of China Medical University, Shenyang, China
| |
Collapse
|
18
|
Roh HW, Chauhan N, Seo SW, Choi SH, Kim E, Cho SH, Kim BC, Choi JW, An Y, Park B, Lee SM, Moon SY, Nam YJ, Hong S, Son SJ, Hong CH, Lee D. Assessing cognitive impairment and disability in older adults through the lens of whole brain white matter patterns. Alzheimers Dement 2024; 20:6032-6044. [PMID: 39001624 PMCID: PMC11497644 DOI: 10.1002/alz.14094] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Revised: 05/22/2024] [Accepted: 05/22/2024] [Indexed: 10/25/2024]
Abstract
INTRODUCTION This study aimed to explore the potential of whole brain white matter patterns as novel neuroimaging biomarkers for assessing cognitive impairment and disability in older adults. METHODS We conducted an in-depth analysis of magnetic resonance imaging (MRI) and amyloid positron emission tomography (PET) scans in 454 participants, focusing on white matter patterns and white matter inter-subject variability (WM-ISV). RESULTS The white matter pattern ensemble model, combining MRI and amyloid PET, demonstrated a significantly higher classification performance for cognitive impairment and disability. Participants with Alzheimer's disease (AD) exhibited higher WM-ISV than participants with subjective cognitive decline, mild cognitive impairment, and vascular dementia. Furthermore, WM-ISV correlated significantly with blood-based biomarkers (such as glial fibrillary acidic protein and phosphorylated tau-217 [p-tau217]), and cognitive function and disability scores. DISCUSSION Our results suggest that white matter pattern analysis has significant potential as an adjunct neuroimaging biomarker for clinical decision-making and determining cognitive impairment and disability. HIGHLIGHTS The ensemble model combined both magnetic resonance imaging (MRI) and amyloid positron emission tomography (PET) and demonstrated a significantly higher classification performance for cognitive impairment and disability. Alzheimer's disease (AD) revealed a notably higher heterogeneity compared to that in subjective cognitive decline, mild cognitive impairment, or vascular dementia. White matter inter-subject variability (WM-ISV) was significantly correlated with blood-based biomarkers (glial fibrillary acidic protein and phosphorylated tau-217 [p-tau217]) and with the polygenic risk score for AD. White matter pattern analysis has significant potential as an adjunct neuroimaging biomarker for clinical decision-making processes and determining cognitive impairment and disability.
Collapse
Affiliation(s)
- Hyun Woong Roh
- Department of PsychiatryAjou University School of MedicineSuwonRepublic of Korea
| | - Nishant Chauhan
- Cognitive Science Research GroupKorea Brain Research InstituteDaeguRepublic of Korea
| | - Sang Won Seo
- Department of NeurologySamsung Medical CenterSungkyunkwan University School of MedicineSeoulRepublic of Korea
| | - Seong Hye Choi
- Department of NeurologyInha University School of MedicineIncheonRepublic of Korea
| | - Eun‐Joo Kim
- Department of NeurologyPusan National University HospitalPusan National University School of Medicine and Medical Research InstituteBusanRepublic of Korea
| | - Soo Hyun Cho
- Department of NeurologyChonnam National University Medical SchoolChonnam National University HospitalGwangjuRepublic of Korea
| | - Byeong C. Kim
- Department of NeurologyChonnam National University Medical SchoolChonnam National University HospitalGwangjuRepublic of Korea
| | - Jin Wook Choi
- Department of RadiologyAjou University School of MedicineSuwonRepublic of Korea
| | - Young‐Sil An
- Department of Nuclear Medicine and Molecular ImagingAjou University School of MedicineSuwonRepublic of Korea
| | - Bumhee Park
- Department of Biomedical InformaticsAjou University School of MedicineSuwonRepublic of Korea
- Office of BiostatisticsAjou Research Institute for Innovative MedicineAjou University Medical CenterSuwonRepublic of Korea
| | - Sun Min Lee
- Department of NeurologyAjou University School of MedicineSuwonRepublic of Korea
| | - So Young Moon
- Department of NeurologyAjou University School of MedicineSuwonRepublic of Korea
| | - You Jin Nam
- Department of PsychiatryAjou University School of MedicineSuwonRepublic of Korea
| | - Sunhwa Hong
- Department of PsychiatryAjou University School of MedicineSuwonRepublic of Korea
| | - Sang Joon Son
- Department of PsychiatryAjou University School of MedicineSuwonRepublic of Korea
| | - Chang Hyung Hong
- Department of PsychiatryAjou University School of MedicineSuwonRepublic of Korea
| | - Dongha Lee
- Cognitive Science Research GroupKorea Brain Research InstituteDaeguRepublic of Korea
| |
Collapse
|
19
|
Morcillo‐Nieto AO, Zsadanyi SE, Arriola‐Infante JE, Carmona‐Iragui M, Montal V, Pegueroles J, Aranha MR, Vaqué‐Alcázar L, Padilla C, Benejam B, Videla L, Barroeta I, Fernandez S, Altuna M, Giménez S, González‐Ortiz S, Bargalló N, Ribas L, Arranz J, Torres S, Iulita MF, Belbin O, Camacho V, Alcolea D, Lleó A, Fortea J, Bejanin A. Characterization of white matter hyperintensities in Down syndrome. Alzheimers Dement 2024; 20:6527-6541. [PMID: 39087352 PMCID: PMC11497714 DOI: 10.1002/alz.14146] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Revised: 06/20/2024] [Accepted: 07/02/2024] [Indexed: 08/02/2024]
Abstract
INTRODUCTION In Down syndrome (DS), white matter hyperintensities (WMHs) are highly prevalent, yet their topography and association with sociodemographic data and Alzheimer's disease (AD) biomarkers remain largely unexplored. METHODS In 261 DS adults and 131 euploid controls, fluid-attenuated inversion recovery magnetic resonance imaging scans were segmented and WMHs were extracted in concentric white matter layers and lobar regions. We tested associations with AD clinical stages, sociodemographic data, cerebrospinal fluid (CSF) AD biomarkers, and gray matter (GM) volume. RESULTS In DS, total WMHs arose at age 43 and showed stronger associations with age than in controls. WMH volume increased along the AD continuum, particularly in periventricular regions, and frontal, parietal, and occipital lobes. Associations were found with CSF biomarkers and temporo-parietal GM volumes. DISCUSSION WMHs increase 10 years before AD symptom onset in DS and are closely linked with AD biomarkers and neurodegeneration. This suggests a direct connection to AD pathophysiology, independent of vascular risks. HIGHLIGHTS White matter hyperintensities (WMHs) increased 10 years before Alzheimer's disease symptom onset in Down syndrome (DS). WMHs were strongly associated in DS with the neurofilament light chain biomarker. WMHs were more associated in DS with gray matter volume in parieto-temporal areas.
Collapse
|
20
|
Botella Lucena P, Heneka MT. Inflammatory aspects of Alzheimer's disease. Acta Neuropathol 2024; 148:31. [PMID: 39196440 DOI: 10.1007/s00401-024-02790-2] [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: 04/26/2024] [Revised: 08/14/2024] [Accepted: 08/15/2024] [Indexed: 08/29/2024]
Abstract
Alzheimer´s disease (AD) stands out as the most common chronic neurodegenerative disorder. AD is characterized by progressive cognitive decline and memory loss, with neurodegeneration as its primary pathological feature. The role of neuroinflammation in the disease course has become a focus of intense research. While microglia, the brain's resident macrophages, have been pivotal to study central immune inflammation, recent evidence underscores the contributions of other cellular entities to the neuroinflammatory process. In this article, we review the inflammatory role of microglia and astrocytes, focusing on their interactions with AD's core pathologies, amyloid beta deposition, and tau tangle formation. Additionally, we also discuss how different modes of regulated cell death in AD may impact the chronic neuroinflammatory environment. This review aims to highlight the evolving landscape of neuroinflammatory research in AD and underscores the importance of considering multiple cellular contributors when developing new therapeutic strategies.
Collapse
Affiliation(s)
- Pablo Botella Lucena
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, 6, Avenue du Swing, Belvaux, L-4367, Esch-Belval, Luxembourg
| | - Michael T Heneka
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, 6, Avenue du Swing, Belvaux, L-4367, Esch-Belval, Luxembourg.
- Department of Infectious Diseases and Immunology, University of Massachusetts Medical School, Worcester, MA, USA.
| |
Collapse
|
21
|
Leenders F, Koole L, Slaets H, Tiane A, Hove DVD, Vanmierlo T. Navigating oligodendrocyte precursor cell aging in brain health. Mech Ageing Dev 2024; 220:111959. [PMID: 38950628 DOI: 10.1016/j.mad.2024.111959] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2024] [Revised: 06/17/2024] [Accepted: 06/24/2024] [Indexed: 07/03/2024]
Abstract
Oligodendrocyte precursor cells (OPCs) comprise 5-8 % of the adult glial cell population and stand out as the most proliferative cell type in the central nervous system (CNS). OPCs are responsible for generating oligodendrocytes (OLs), the myelinating cells of the CNS. However, OPC functions decline as we age, resulting in impaired differentiation and inadequate remyelination. This review explores the cellular and molecular changes associated with OPC aging, and their impact on OPC differentiation and functionality. Furthermore, it examines the impact of OPC aging within the context of multiple sclerosis and Alzheimer's disease, both neurodegenerative conditions wherein aged OPCs exacerbate disease progression by impeding remyelination. Moreover, various pharmacological interventions targeting pathways related to senescence and differentiation are discussed as potential strategies to rejuvenate aged OPCs. Enhancing our understanding of OPC aging mechanisms holds promise for developing new therapies to improve remyelination and repair in age-related neurodegenerative disorders.
Collapse
Affiliation(s)
- Freddy Leenders
- Department Psychiatry and Neuropsychology, Division Translational Neuroscience, Mental Health and Neuroscience Research Institute, Maastricht University, Maastricht, the Netherlands; Department of Neuroscience, Biomedical Research Institute, Faculty of Medicine and Life Sciences, Hasselt University, Diepenbeek, Belgium
| | - Lisa Koole
- Department Psychiatry and Neuropsychology, Division Translational Neuroscience, Mental Health and Neuroscience Research Institute, Maastricht University, Maastricht, the Netherlands; Department of Neuroscience, Biomedical Research Institute, Faculty of Medicine and Life Sciences, Hasselt University, Diepenbeek, Belgium
| | - Helena Slaets
- University MS Centre (UMSC) Hasselt, Pelt, Belgium; Neuro-Immune Connections and Repair Lab, Department of Immunology and Infection, Biomedical Research Institute, Hasselt University, Diepenbeek, Belgium
| | - Assia Tiane
- Department Psychiatry and Neuropsychology, Division Translational Neuroscience, Mental Health and Neuroscience Research Institute, Maastricht University, Maastricht, the Netherlands; Department of Neuroscience, Biomedical Research Institute, Faculty of Medicine and Life Sciences, Hasselt University, Diepenbeek, Belgium; University MS Centre (UMSC) Hasselt, Pelt, Belgium
| | - Daniel van den Hove
- Department Psychiatry and Neuropsychology, Division Translational Neuroscience, Mental Health and Neuroscience Research Institute, Maastricht University, Maastricht, the Netherlands
| | - Tim Vanmierlo
- Department Psychiatry and Neuropsychology, Division Translational Neuroscience, Mental Health and Neuroscience Research Institute, Maastricht University, Maastricht, the Netherlands; Department of Neuroscience, Biomedical Research Institute, Faculty of Medicine and Life Sciences, Hasselt University, Diepenbeek, Belgium; University MS Centre (UMSC) Hasselt, Pelt, Belgium.
| |
Collapse
|
22
|
Liu S, Luo X, Chong JSX, Jiaerken Y, Youn SH, Zhang M, Zhou JH. Brain structure, amyloid, and behavioral features for predicting clinical progression in subjective cognitive decline. Hum Brain Mapp 2024; 45:e26765. [PMID: 38958401 PMCID: PMC11220833 DOI: 10.1002/hbm.26765] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2023] [Revised: 05/28/2024] [Accepted: 06/10/2024] [Indexed: 07/04/2024] Open
Abstract
As a potential preclinical stage of Alzheimer's dementia, subjective cognitive decline (SCD) reveals a higher risk of future cognitive decline and conversion to dementia. However, it has not been clear whether SCD status increases the clinical progression of older adults in the context of amyloid deposition, cerebrovascular disease (CeVD), and psychiatric symptoms. We identified 99 normal controls (NC), 15 SCD individuals who developed mild cognitive impairment in the next 2 years (P-SCD), and 54 SCD individuals who did not (S-SCD) from ADNI database with both baseline and 2-year follow-up data. Total white matter hyperintensity (WMH), WMH in deep (DWMH) and periventricular (PWMH) regions, and voxel-wise grey matter volumes were compared among groups. Furthermore, using structural equation modelling method, we constructed path models to explore SCD-related brain changes longitudinally and to determine whether baseline SCD status, age, and depressive symptoms affect participants' clinical outcomes. Both SCD groups showed higher baseline amyloid PET SUVR, baseline PWMH volumes, and larger increase of PWMH volumes over time than NC. In contrast, only P-SCD had higher baseline DWMH volumes and larger increase of DWMH volumes over time than NC. No longitudinal differences in grey matter volume and amyloid was observed among NC, S-SCD, and P-SCD. Our path models demonstrated that SCD status contributed to future WMH progression. Further, baseline SCD status increases the risk of future cognitive decline, mediated by PWMH; baseline depressive symptoms directly contribute to clinical outcomes. In conclusion, both S-SCD and P-SCD exhibited more severe CeVD than NC. The CeVD burden increase was more pronounced in P-SCD. In contrast with the direct association of depressive symptoms with dementia severity progression, the effects of SCD status on future cognitive decline may manifest via CeVD pathologies. Our work highlights the importance of multi-modal longitudinal designs in understanding the SCD trajectory heterogeneity, paving the way for stratification and early intervention in the preclinical stage. PRACTITIONER POINTS: Both S-SCD and P-SCD exhibited more severe CeVD at baseline and a larger increase of CeVD burden compared to NC, while the burden was more pronounced in P-SCD. Baseline SCD status increases the risk of future PWMH and DWMH volume accumulation, mediated by baseline PWMH and DWMH volumes, respectively. Baseline SCD status increases the risk of future cognitive decline, mediated by baseline PWMH, while baseline depression status directly contributes to clinical outcome.
Collapse
Grants
- U01 AG024904 NIA NIH HHS
- W81XWH-12-2-0012 DoD Alzheimer's Disease Neuroimaging Initiative (Department of Defense)
- A20G8b0102 Research, Innovation and Enterprise (RIE) 2020 Advanced Manufacturing and Engineering (AME) Programmatic Fund (Agency for Science, Technology and Research (A*STAR), Singapore)
- NMRC/OFLCG19May-0035 National Medical Research Council, Singapore
- NMRC/CIRG/1485/2018 National Medical Research Council, Singapore
- NMRC/CSA-SI/0007/2016 National Medical Research Council, Singapore
- NMRC/MOH-00707-01 National Medical Research Council, Singapore
- NMRC/CG/435M009/2017-NUH/NUHS National Medical Research Council, Singapore
- CIRG21nov-0007 National Medical Research Council, Singapore
- HLCA23Feb-0004 National Medical Research Council, Singapore
- Yong Loo Lin School of Medicine Research Core Funding (National University of Singapore, Singapore)
- 82271936 National Natural Science Foundation of China
- 2022ZQ057 Zhejiang Provincial Administration of Traditional Chinese Medicine - Youth Talent Fund Project
- MOE-T2EP40120-0007 Ministry of Education, Singapore
- T2EP2-0223-0025 Ministry of Education, Singapore
- MOE-T2EP20220-0001 Ministry of Education, Singapore
- Alzheimer's Disease Neuroimaging Initiative (National Institutes of Health)
- DoD Alzheimer's Disease Neuroimaging Initiative (Department of Defense)
- National Medical Research Council, Singapore
- National Natural Science Foundation of China
- Ministry of Education, Singapore
Collapse
Affiliation(s)
- Siwei Liu
- Centre for Sleep and CognitionCentre for Translational Magnetic Resonance Research, Yong Loo Lin School of MedicineNational University of SingaporeSingaporeSingapore
- Human Potential Translational Research ProgramDepartment of MedicineNational University of SingaporeSingaporeSingapore
| | - Xiao Luo
- Department of Radiology, Second Affiliated HospitalZhejiang University School of MedicineHangzhouChina
| | - Joanna Su Xian Chong
- Centre for Sleep and CognitionCentre for Translational Magnetic Resonance Research, Yong Loo Lin School of MedicineNational University of SingaporeSingaporeSingapore
- Human Potential Translational Research ProgramDepartment of MedicineNational University of SingaporeSingaporeSingapore
| | - Yeerfan Jiaerken
- Department of Radiology, Second Affiliated HospitalZhejiang University School of MedicineHangzhouChina
| | - Shim Hee Youn
- Centre for Sleep and CognitionCentre for Translational Magnetic Resonance Research, Yong Loo Lin School of MedicineNational University of SingaporeSingaporeSingapore
- Human Potential Translational Research ProgramDepartment of MedicineNational University of SingaporeSingaporeSingapore
| | - Minming Zhang
- Department of Radiology, Second Affiliated HospitalZhejiang University School of MedicineHangzhouChina
| | - Juan Helen Zhou
- Centre for Sleep and CognitionCentre for Translational Magnetic Resonance Research, Yong Loo Lin School of MedicineNational University of SingaporeSingaporeSingapore
- Human Potential Translational Research ProgramDepartment of MedicineNational University of SingaporeSingaporeSingapore
- Department of Electrical and Computer EngineeringIntegrative Sciences and Engineering Programme (ISEP), NUS Graduate SchoolNational University of SingaporeSingaporeSingapore
| | | |
Collapse
|
23
|
Pradeep A, Raghavan S, Przybelski SA, Preboske GM, Schwarz CG, Lowe VJ, Knopman DS, Petersen RC, Jack CR, Graff-Radford J, Cogswell PM, Vemuri P. Can white matter hyperintensities based Fazekas visual assessment scales inform about Alzheimer's disease pathology in the population? Alzheimers Res Ther 2024; 16:157. [PMID: 38987827 PMCID: PMC11234605 DOI: 10.1186/s13195-024-01525-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2024] [Accepted: 07/02/2024] [Indexed: 07/12/2024]
Abstract
BACKGROUND White matter hyperintensities (WMH) are considered hallmark features of cerebral small vessel disease and have recently been linked to Alzheimer's disease (AD) pathology. Their distinct spatial distributions, namely periventricular versus deep WMH, may differ by underlying age-related and pathobiological processes contributing to cognitive decline. We aimed to identify the spatial patterns of WMH using the 4-scale Fazekas visual assessment and explore their differential association with age, vascular health, AD imaging markers, namely amyloid and tau burden, and cognition. Because our study consisted of scans from GE and Siemens scanners with different resolutions, we also investigated inter-scanner reproducibility and combinability of WMH measurements on imaging. METHODS We identified 1144 participants from the Mayo Clinic Study of Aging consisting of a population-based sample from Olmsted County, Minnesota with available structural magnetic resonance imaging (MRI), amyloid, and tau positron emission tomography (PET). WMH distribution patterns were assessed on FLAIR-MRI, both 2D axial and 3D, using Fazekas ratings of periventricular and deep WMH severity. We compared the association of periventricular and deep WMH scales with vascular risk factors, amyloid-PET, and tau-PET standardized uptake value ratio, automated WMH volume, and cognition using Pearson partial correlation after adjusting for age. We also evaluated vendor compatibility and reproducibility of the Fazekas scales using intraclass correlations (ICC). RESULTS Periventricular and deep WMH measurements showed similar correlations with age, cardiometabolic conditions score (vascular risk), and cognition, (p < 0.001). Both periventricular WMH and deep WMH showed weak associations with amyloidosis (R = 0.07, p = < 0.001), and none with tau burden. We found substantial agreement between data from the two scanners for Fazekas measurements (ICC = 0.82 and 0.74). The automated WMH volume had high discriminating power for identifying participants with Fazekas ≥ 2 (area under curve = 0.97) and showed poor correlation with amyloid and tau PET markers similar to the visual grading. CONCLUSION Our study investigated risk factors underlying WMH spatial patterns and their impact on global cognition, with no discernible differences between periventricular and deep WMH. We observed minimal impact of amyloidosis on WMH severity. These findings, coupled with enhanced inter-scanner reproducibility of WMH data, suggest the combinability of inter-scanner data assessed by harmonized protocols in the context of vascular contributions to cognitive impairment and dementia biomarker research.
Collapse
Affiliation(s)
| | - Sheelakumari Raghavan
- Department of Radiology, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA
| | - Scott A Przybelski
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN, 55905, USA
| | - Gregory M Preboske
- Department of Radiology, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA
| | - Christopher G Schwarz
- Department of Radiology, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA
| | - Val J Lowe
- Department of Radiology, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA
| | - David S Knopman
- Department of Neurology, Mayo Clinic, Rochester, MN, 55905, USA
| | | | - Clifford R Jack
- Department of Radiology, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA
| | | | - Petrice M Cogswell
- Department of Radiology, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA
| | - Prashanthi Vemuri
- Department of Radiology, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA.
| |
Collapse
|
24
|
Mao M, Wei Y, Wang C, Han X, Liu R, Dong Y, Song L, Cong L, Wang Y, Du Y, Qiu C. Prolonged ventricular repolarization associated with mild cognitive impairment and white matter hyperintensities: a cross-sectional study. Sci Rep 2024; 14:15162. [PMID: 38956440 PMCID: PMC11219852 DOI: 10.1038/s41598-024-65364-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2024] [Accepted: 06/19/2024] [Indexed: 07/04/2024] Open
Abstract
Prolonged ventricular repolarization has been associated with cardiovascular disease. We sought to investigate the association of prolonged ventricular repolarization with mild cognitive impairment (MCI) and the potential underlying neuropathological mechanisms in older adults. This cross-sectional study included 4328 dementia-free participants (age ≥ 65 years; 56.8% female) in the baseline examination of the Multidomain INterventions to delay dementia and Disability in rural China; of these, 989 undertook structural brain magnetic resonance imaging (MRI) scans. QT, QTc, JT, JTc, and QRS intervals were derived from 12-lead electrocardiograph. MCI, amnestic MCI (aMCI), and non-amnestic MCI (naMCI) were defined following the Petersen's criteria. Volumes of gray matter (GM), white matter, cerebrospinal fluid, total white matter hyperintensities (WMH), periventricular WMH (PWMH), and deep WMH (DWMH) were automatically estimated. Data were analyzed using logistic and general linear regression models. Prolonged QT, QTc, JT, and JTc intervals were significantly associated with an increased likelihood of MCI and aMCI, but not naMCI (p < 0.05). In the MRI subsample, QT, QTc, JT, and JTc intervals were significantly associated with larger total WMH and PWMH volumes (p < 0.05), but not with DWMH volume. Statistical interactions were detected, such that prolonged QT and JT intervals were significantly associated with reduced GM volume only among participants with coronary heart disease or without APOE ε4 allele (p < 0.05). Prolonged ventricular repolarization is associated with MCI and cerebral microvascular lesions in a general population of older adults. This underlies the importance of cognitive assessments and brain MRI examination among older adults with prolonged QT interval.
Collapse
Affiliation(s)
- Ming Mao
- Department of Neurology, Shandong Provincial Hospital affiliated to Shandong First Medical University, No. 324 Jingwuweiqi Road, Jinan, 250021, Shandong, People's Republic of China
- Key Laboratory of Endocrine Glucose and Lipids Metabolism and Brain Aging in Shandong First Medical University, Ministry of Education of the People's Republic of China, Jinan, 250021, Shandong, People's Republic of China
| | - Yiran Wei
- Department of Neurology, Shandong Provincial Hospital affiliated to Shandong First Medical University, No. 324 Jingwuweiqi Road, Jinan, 250021, Shandong, People's Republic of China
| | - Chaoqun Wang
- Department of Neurology, Shandong Provincial Hospital, Shandong University, Jinan, 250021, Shandong, People's Republic of China
| | - Xiaolei Han
- Department of Neurology, Shandong Provincial Hospital affiliated to Shandong First Medical University, No. 324 Jingwuweiqi Road, Jinan, 250021, Shandong, People's Republic of China
- Key Laboratory of Endocrine Glucose and Lipids Metabolism and Brain Aging in Shandong First Medical University, Ministry of Education of the People's Republic of China, Jinan, 250021, Shandong, People's Republic of China
| | - Rui Liu
- Department of Neurology, Shandong Provincial Hospital affiliated to Shandong First Medical University, No. 324 Jingwuweiqi Road, Jinan, 250021, Shandong, People's Republic of China
- Key Laboratory of Endocrine Glucose and Lipids Metabolism and Brain Aging in Shandong First Medical University, Ministry of Education of the People's Republic of China, Jinan, 250021, Shandong, People's Republic of China
| | - Yi Dong
- Department of Neurology, Shandong Provincial Hospital affiliated to Shandong First Medical University, No. 324 Jingwuweiqi Road, Jinan, 250021, Shandong, People's Republic of China
- Key Laboratory of Endocrine Glucose and Lipids Metabolism and Brain Aging in Shandong First Medical University, Ministry of Education of the People's Republic of China, Jinan, 250021, Shandong, People's Republic of China
| | - Lin Song
- Department of Neurology, Shandong Provincial Hospital affiliated to Shandong First Medical University, No. 324 Jingwuweiqi Road, Jinan, 250021, Shandong, People's Republic of China
- Key Laboratory of Endocrine Glucose and Lipids Metabolism and Brain Aging in Shandong First Medical University, Ministry of Education of the People's Republic of China, Jinan, 250021, Shandong, People's Republic of China
| | - Lin Cong
- Department of Neurology, Shandong Provincial Hospital affiliated to Shandong First Medical University, No. 324 Jingwuweiqi Road, Jinan, 250021, Shandong, People's Republic of China
- Key Laboratory of Endocrine Glucose and Lipids Metabolism and Brain Aging in Shandong First Medical University, Ministry of Education of the People's Republic of China, Jinan, 250021, Shandong, People's Republic of China
| | - Yongxiang Wang
- Department of Neurology, Shandong Provincial Hospital affiliated to Shandong First Medical University, No. 324 Jingwuweiqi Road, Jinan, 250021, Shandong, People's Republic of China.
- Key Laboratory of Endocrine Glucose and Lipids Metabolism and Brain Aging in Shandong First Medical University, Ministry of Education of the People's Republic of China, Jinan, 250021, Shandong, People's Republic of China.
- Institute of Brain Science and Brain-Inspired Research, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, 250021, Shandong, People's Republic of China.
- Department of Neurobiology, Care Sciences and Society, Aging Research Center and Center for Alzheimer Research, Karolinska Institutet-Stockholm University, 17177, Stockholm, Sweden.
| | - Yifeng Du
- Department of Neurology, Shandong Provincial Hospital affiliated to Shandong First Medical University, No. 324 Jingwuweiqi Road, Jinan, 250021, Shandong, People's Republic of China.
- Key Laboratory of Endocrine Glucose and Lipids Metabolism and Brain Aging in Shandong First Medical University, Ministry of Education of the People's Republic of China, Jinan, 250021, Shandong, People's Republic of China.
- Department of Neurology, Shandong Provincial Hospital, Shandong University, Jinan, 250021, Shandong, People's Republic of China.
| | - Chengxuan Qiu
- Department of Neurology, Shandong Provincial Hospital, Shandong University, Jinan, 250021, Shandong, People's Republic of China
- Department of Neurobiology, Care Sciences and Society, Aging Research Center and Center for Alzheimer Research, Karolinska Institutet-Stockholm University, 17177, Stockholm, Sweden
| |
Collapse
|
25
|
Kapoor A, Dutt S, Alitin JPM, Sible IJ, Marshall A, Shenasa F, Engstrom AC, Gaubert A, Shao X, Bradford DR, Rodgers K, Mather M, Wang DJJ, Nation DA. Older adults with reduced cerebrovascular reactivity exhibit high white matter hyperintensity burden. Neurobiol Aging 2024; 139:5-10. [PMID: 38579393 PMCID: PMC11896757 DOI: 10.1016/j.neurobiolaging.2024.03.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Revised: 03/17/2024] [Accepted: 03/22/2024] [Indexed: 04/07/2024]
Abstract
Cerebrovascular reactivity (CVR) deficits may contribute to small vessel disease, such as white matter hyperintensities (WMH). Moreover, apolipoprotein-e4 (APOE4) carriers at genetic risk for Alzheimer's disease exhibit cerebrovascular dysfunction relative to non-carriers. We examined whether older adults, and APOE4 carriers specifically, with diminished CVR would exhibit higher WMH burden. Independently living older adults (N = 125, mean age = 69.2 years; SD = 7.6; 31.2% male) free of dementia or clinical stroke underwent brain MRI to quantify cerebral perfusion during CVR to hypercapnia and hypocapnia and determine WMH volume. Adjusting for age, sex and intracranial volume, hierarchical regression analysis revealed a significant association between whole brain CVR to hypercapnia and WMH overall [B = -.02, 95% CI (-.04, -.008), p =.003] and in APOE4 carriers [B = -.03, 95% CI (-.06, -.009), p =.009]. Findings suggest deficits in cerebral vasodilatory capacity are associated with WMH burden in older adults and future studies are warranted to further delineate the effect of APOE4 on precipitating WMH.
Collapse
Affiliation(s)
- Arunima Kapoor
- Department of Psychological Science, University of California, Irvine, Irvine, CA, USA
| | - Shubir Dutt
- Department of Psychology, University of Southern California, Los Angeles, CA, USA
| | - John Paul M Alitin
- Institute for Memory Impairments and Neurological Disorders, University of California, Irvine, Irvine, CA, USA
| | - Isabel J Sible
- Department of Psychology, University of Southern California, Los Angeles, CA, USA
| | - Anisa Marshall
- Department of Psychology, University of Southern California, Los Angeles, CA, USA
| | - Fatemah Shenasa
- Department of Psychological Science, University of California, Irvine, Irvine, CA, USA
| | - Allison C Engstrom
- Department of Psychological Science, University of California, Irvine, Irvine, CA, USA
| | - Aimée Gaubert
- Institute for Memory Impairments and Neurological Disorders, University of California, Irvine, Irvine, CA, USA
| | - Xingfeng Shao
- Stevens Neuroimaging and Informatics Institute, University of Southern California, Los Angeles, CA, USA
| | - David Robert Bradford
- Center for Innovations in Brain Science, Department of Pharmacology, University of Arizona, Tucson, AZ, USA
| | - Kathleen Rodgers
- Center for Innovations in Brain Science, Department of Pharmacology, University of Arizona, Tucson, AZ, USA
| | - Mara Mather
- University of Southern California Leonard Davis School of Gerontology, USA
| | - Danny J J Wang
- Stevens Neuroimaging and Informatics Institute, University of Southern California, Los Angeles, CA, USA
| | - Daniel A Nation
- University of Southern California Leonard Davis School of Gerontology, USA; Department of Physiology and Neuroscience, Zilkha Neurogenetic Institute, University of Southern California Keck School of Medicine, USA.
| |
Collapse
|
26
|
Raichlen DA, Ally M, Aslan DH, Sayre MK, Bharadwaj PK, Maltagliati S, Lai MHC, Wilcox RR, Habeck CG, Klimentidis YC, Alexander GE. Associations between accelerometer-derived sedentary behavior and physical activity with white matter hyperintensities in middle-aged to older adults. ALZHEIMER'S & DEMENTIA (AMSTERDAM, NETHERLANDS) 2024; 16:e70001. [PMID: 39183745 PMCID: PMC11342350 DOI: 10.1002/dad2.70001] [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: 04/14/2024] [Revised: 07/30/2024] [Accepted: 08/02/2024] [Indexed: 08/27/2024]
Abstract
INTRODUCTION We examined the relationship between sedentary behavior (SB), moderate-to-vigorous physical activity (MVPA), and white matter hyperintensity (WMH) volumes, a common magnetic resonance imaging (MRI) marker associated with risk of neurodegenerative disease in middle-aged to older adults. METHODS We used data from the UK Biobank (n = 14,415; 45 to 81 years) that included accelerometer-derived measures of SB and MVPA, and WMH volumes from MRI. RESULTS Both MVPA and SB were associated with WMH volumes (βMVPA = -0.03 [-0.04, -0.01], p < 0.001; βSB = 0.02 [0.01, 0.03], p = 0.007). There was a significant interaction between SB and MVPA on WMH volumes (βSB×MVPA = -0.015 [-0.028, -0.001], p SB×MVPA = 0.03) where SB was positively associated with WMHs at low MVPA, and MVPA was negatively associated with WMHs at high SB. DISCUSSION While this study cannot establish causality, the results highlight the potential importance of considering both MVPA and SB in strategies aimed at reducing the accumulation of WMH volumes in middle-aged to older adults. Highlights SB is associated with greater WMH volumes and MVPA is associated with lower WMH volumes.Relationships between SB and WMH are strongest at low levels of MVPA.Associations between MVPA and WMH are strongest at high levels of SB.Considering both SB and MVPA may be effective strategies for reducing WMHs.
Collapse
Affiliation(s)
- David A. Raichlen
- Human and Evolutionary Biology SectionDepartment of Biological SciencesUniversity of Southern CaliforniaLos AngelesCaliforniaUSA
- Department of AnthropologyUniversity of Southern CaliforniaLos AngelesCaliforniaUSA
| | - Madeline Ally
- Department of PsychologyUniversity of ArizonaTucsonArizonaUSA
| | - Daniel H. Aslan
- Human and Evolutionary Biology SectionDepartment of Biological SciencesUniversity of Southern CaliforniaLos AngelesCaliforniaUSA
| | | | | | - Silvio Maltagliati
- Human and Evolutionary Biology SectionDepartment of Biological SciencesUniversity of Southern CaliforniaLos AngelesCaliforniaUSA
| | - Mark H. C. Lai
- Department of PsychologyUniversity of Southern CaliforniaLos AngelesCaliforniaUSA
| | - Rand R. Wilcox
- Department of PsychologyUniversity of Southern CaliforniaLos AngelesCaliforniaUSA
| | - Christian G. Habeck
- Cognitive Neuroscience DivisionDepartment of Neurology and Taub InstituteColumbia UniversityNew YorkNew YorkUSA
| | - Yann C. Klimentidis
- Department of Epidemiology and BiostatisticsMel and Enid Zuckerman College of Public HealthUniversity of ArizonaTucsonArizonaUSA
- BIO5 InstituteUniversity of ArizonaTucsonArizonaUSA
| | - Gene E. Alexander
- Department of PsychologyUniversity of ArizonaTucsonArizonaUSA
- BIO5 InstituteUniversity of ArizonaTucsonArizonaUSA
- Evelyn F. McKnight Brain InstituteUniversity of ArizonaTucsonArizonaUSA
- Department of PsychiatryUniversity of ArizonaTucsonArizonaUSA
- Neuroscience Graduate Interdisciplinary ProgramUniversity of ArizonaTucsonArizonaUSA
- Physiological Sciences Graduate Interdisciplinary ProgramUniversity of ArizonaTucsonArizonaUSA
- Arizona Alzheimer's ConsortiumPhoenixArizonaUSA
| |
Collapse
|
27
|
Zhu SG, Chen ZL, Xiao K, Wang ZW, Lu WB, Liu RP, Huang SS, Zhu JH, Zhang X, Wang JY. Association analyses of apolipoprotein E genotypes and cognitive performance in patients with Parkinson's disease. Eur J Med Res 2024; 29:334. [PMID: 38880878 PMCID: PMC11181540 DOI: 10.1186/s40001-024-01924-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2024] [Accepted: 06/06/2024] [Indexed: 06/18/2024] Open
Abstract
BACKGROUND Cognitive impairment is a common non-motor symptom of Parkinson's disease (PD). The apolipoprotein E (APOE) ε4 genotype increases the risk of Alzheimer's disease (AD). However, the effect of APOEε4 on cognitive function of PD patients remains unclear. In this study, we aimed to understand whether and how carrying APOEε4 affects cognitive performance in patients with early-stage and advanced PD. METHODS A total of 119 Chinese early-stage PD patients were recruited. Movement Disorder Society Unified Parkinson's Disease Rating Scale, Hamilton anxiety scale, Hamilton depression scale, non-motor symptoms scale, Mini-mental State Examination, Montreal Cognitive Assessment, and Fazekas scale were evaluated. APOE genotypes were determined by polymerase chain reactions and direct sequencing. Demographic and clinical information of 521 early-stage and 262 advanced PD patients were obtained from Parkinson's Progression Marker Initiative (PPMI). RESULTS No significant difference in cognitive performance was found between ApoEε4 carriers and non-carriers in early-stage PD patients from our cohort and PPMI. The cerebrospinal fluid (CSF) Amyloid Beta 42 (Aβ42) level was significantly lower in ApoEε4 carrier than non-carriers in early-stage PD patients from PPMI. In advanced PD patients from PPMI, the BJLOT, HVLT retention and SDMT scores seem to be lower in ApoEε4 carriers without reach the statistical significance. CONCLUSIONS APOEε4 carriage does not affect the cognitive performance of early-stage PD patients. However, it may promote the decline of CSF Aβ42 level and the associated amyloidopathy, which is likely to further contribute to the cognitive dysfunction of PD patients in the advanced stage.
Collapse
Affiliation(s)
- Shi-Guo Zhu
- Department of Neurology, Institute of Geriatric Neurology, the Second Affiliated Hospital and Yuying Children's Hospital, Wenzhou Medical University, Wenzhou, 325027, Zhejiang, China
| | - Zhu-Ling Chen
- Department of Neurology, Institute of Geriatric Neurology, the Second Affiliated Hospital and Yuying Children's Hospital, Wenzhou Medical University, Wenzhou, 325027, Zhejiang, China
| | - Ke Xiao
- Department of Neurology, Institute of Geriatric Neurology, the Second Affiliated Hospital and Yuying Children's Hospital, Wenzhou Medical University, Wenzhou, 325027, Zhejiang, China
| | - Zi-Wei Wang
- Department of Neurology, Institute of Geriatric Neurology, the Second Affiliated Hospital and Yuying Children's Hospital, Wenzhou Medical University, Wenzhou, 325027, Zhejiang, China
| | - Wen-Bin Lu
- Department of Neurology, Institute of Geriatric Neurology, the Second Affiliated Hospital and Yuying Children's Hospital, Wenzhou Medical University, Wenzhou, 325027, Zhejiang, China
| | - Rong-Pei Liu
- Department of Neurology, Institute of Geriatric Neurology, the Second Affiliated Hospital and Yuying Children's Hospital, Wenzhou Medical University, Wenzhou, 325027, Zhejiang, China
| | - Shi-Shi Huang
- Department of Neurology, Institute of Geriatric Neurology, the Second Affiliated Hospital and Yuying Children's Hospital, Wenzhou Medical University, Wenzhou, 325027, Zhejiang, China
| | - Jian-Hong Zhu
- Department of Preventive Medicine, Institute of Nutrition and Diseases, Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China.
| | - Xiong Zhang
- Department of Neurology, Institute of Geriatric Neurology, the Second Affiliated Hospital and Yuying Children's Hospital, Wenzhou Medical University, Wenzhou, 325027, Zhejiang, China.
| | - Jian-Yong Wang
- Department of Neurology, Institute of Geriatric Neurology, the Second Affiliated Hospital and Yuying Children's Hospital, Wenzhou Medical University, Wenzhou, 325027, Zhejiang, China.
| |
Collapse
|
28
|
Zhang J, Chen H, Wang J, Huang Q, Xu X, Wang W, Xu W, Guan Y, Liu J, Wardlaw JM, Deng Y, Xie F, Li B. Linking white matter hyperintensities to regional cortical thinning, amyloid deposition, and synaptic density loss in Alzheimer's disease. Alzheimers Dement 2024; 20:3931-3942. [PMID: 38648354 PMCID: PMC11180938 DOI: 10.1002/alz.13845] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2023] [Revised: 02/16/2024] [Accepted: 03/21/2024] [Indexed: 04/25/2024]
Abstract
INTRODUCTION We investigated the association between white matter hyperintensities (WMH) and regional cortical thickness, amyloid and tau deposition, and synaptic density in the WMH-connected cortex using multimodal images. METHODS We included 107 participants (59 with Alzheimer's disease [AD]; 27 with mild cognitive impairment; 21 cognitively normal controls) with amyloid beta (Aβ) positivity on amyloid positron emission tomography (PET). The cortex connected to WMH was identified using probabilistic tractography. RESULTS We found that WMH connected to the cortex with more severe regional degeneration as measured by cortical thickness, Aβ and tau deposition, and synaptic vesicle glycoprotein 2 A (SV2A) density using 18F-SynVesT-1 PET. In addition, higher ratios of Aβ in the deep WMH-connected versus WMH-unconnected cortex were significantly related to lower cognitive scores. Last, the cortical thickness of WMH-connected cortex reduced more than WMH-unconnected cortex over 12 months. DISCUSSION Our results suggest that WMH may be associated with AD-intrinsic processes of degeneration, in addition to vascular mechanisms. HIGHLIGHTS We studied white matter hyperintensities (WMHs) and WMH-connected cortical changes. WMHs are associated with more severe regional cortical degeneration. Findings suggest WMHs may be associated with Alzheimer's disease-intrinsic processes of degeneration.
Collapse
Affiliation(s)
- Junfang Zhang
- Department of Neurology & Institute of NeurologyRuijin Hospital affiliated with Shanghai Jiao Tong University School of MedicineShanghaiChina
- Clinical Neuroscience CenterRuijin Hospital LuWan BranchShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Haijuan Chen
- Department of Neurology & Institute of NeurologyRuijin Hospital affiliated with Shanghai Jiao Tong University School of MedicineShanghaiChina
- Clinical Neuroscience CenterRuijin Hospital LuWan BranchShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Jie Wang
- PET CenterHuashan HospitalFudan UniversityShanghaiChina
| | - Qi Huang
- PET CenterHuashan HospitalFudan UniversityShanghaiChina
| | - Xiaomeng Xu
- Department of Neurology & Institute of NeurologyRuijin Hospital affiliated with Shanghai Jiao Tong University School of MedicineShanghaiChina
| | - Wenjing Wang
- Department of Neurology & Institute of NeurologyRuijin Hospital affiliated with Shanghai Jiao Tong University School of MedicineShanghaiChina
| | - Wei Xu
- Department of Neurology & Institute of NeurologyRuijin Hospital affiliated with Shanghai Jiao Tong University School of MedicineShanghaiChina
| | - Yihui Guan
- PET CenterHuashan HospitalFudan UniversityShanghaiChina
| | - Jun Liu
- Department of Neurology & Institute of NeurologyRuijin Hospital affiliated with Shanghai Jiao Tong University School of MedicineShanghaiChina
| | - Joanna M Wardlaw
- Centre for Clinical Brain SciencesUniversity of EdinburghEdinburghUK
- UK Dementia Research InstituteUniversity of EdinburghEdinburghUK
| | - Yulei Deng
- Department of Neurology & Institute of NeurologyRuijin Hospital affiliated with Shanghai Jiao Tong University School of MedicineShanghaiChina
- Clinical Neuroscience CenterRuijin Hospital LuWan BranchShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Fang Xie
- PET CenterHuashan HospitalFudan UniversityShanghaiChina
| | - Binyin Li
- Department of Neurology & Institute of NeurologyRuijin Hospital affiliated with Shanghai Jiao Tong University School of MedicineShanghaiChina
- Clinical Neuroscience CenterRuijin Hospital LuWan BranchShanghai Jiao Tong University School of MedicineShanghaiChina
| |
Collapse
|
29
|
Silbert LC, Roese NE, Krajbich V, Hurworth J, Lahna D, Schwartz DL, Dodge HH, Woltjer RL. White matter hyperintensities and the surrounding normal appearing white matter are associated with water channel disruption in the oldest old. Alzheimers Dement 2024; 20:3839-3851. [PMID: 38629888 PMCID: PMC11180942 DOI: 10.1002/alz.13816] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2023] [Revised: 02/08/2024] [Accepted: 03/08/2024] [Indexed: 06/18/2024]
Abstract
INTRODUCTION Age-related magnetic resonance imaging (MRI) T2 white matter hyperintensities (WMHs) are common and associated with neurological decline. We investigated the histopathological underpinnings of MRI WMH and surrounding normal appearing white matter (NAWM), with a focus on astroglial phenotypes. METHODS Brain samples from 51 oldest old Oregon Alzheimer's Disease Research Center participants who came to autopsy underwent post mortem (PM) 7 tesla MRI with targeted histopathological sampling of WMHs and NAWM. Stained slides were digitized and quantified. Mixed-effects models determined differences in molecular characteristics between WMHs and the NAWM and across NAWM. RESULTS PM MRI-targeted WMHs are characterized by demyelination, microglial activation, and prominent astrocytic alterations, including disrupted aquaporin (AQP) expression. Similar changes occur within the surrounding NAWM in a pattern of decreasing severity with increased distance from WMHs. DISCUSSION Decreased AQP expression within WMH and proximal NAWM suggest an overwhelmed system wherein water homeostasis is no longer maintained, contributing to WM damage in older individuals. HIGHLIGHTS Post mortem magnetic resonance imaging (MRI) was used to characterize the pathology of white matter hyperintensities (WMHs) and surrounding normal appearing white matter (NAWM). Stained immunohistochemical (IHC) slides from targeted WMH and NAWM samples were digitized and quantified. WMHs and NAWM were associated with inflammation, demyelination, and gliosis. WMHs and NAWM astrocytic changes included decreased AQP1 and AQP4 expression. Abnormal NAWM pathology diminished in severity with increasing distance from WMH.
Collapse
Affiliation(s)
- Lisa C. Silbert
- National Institute on Aging, Layton Aging & Alzheimer's Disease Research CenterDepartment of NeurologyOregon Health & Science UniversityPortlandOregonUSA
- Department of NeurologyPortland Veterans Affairs Health Care SystemPortlandOregonUSA
| | - Natalie E. Roese
- National Institute on Aging, Layton Aging & Alzheimer's Disease Research CenterDepartment of NeurologyOregon Health & Science UniversityPortlandOregonUSA
| | - Victoria Krajbich
- Department of PathologyOregon Health & Science UniversityPortlandOregonUSA
| | - Justin Hurworth
- National Institute on Aging, Layton Aging & Alzheimer's Disease Research CenterDepartment of NeurologyOregon Health & Science UniversityPortlandOregonUSA
| | - David Lahna
- National Institute on Aging, Layton Aging & Alzheimer's Disease Research CenterDepartment of NeurologyOregon Health & Science UniversityPortlandOregonUSA
| | - Daniel L. Schwartz
- National Institute on Aging, Layton Aging & Alzheimer's Disease Research CenterDepartment of NeurologyOregon Health & Science UniversityPortlandOregonUSA
- Advanced Imaging Research CenterOregon Health & Science UniversityPortlandOregonUSA
| | - Hiroko H. Dodge
- Department of NeurologyMassachusetts General Hospital, Harvard Medical SchoolBostonMassachusettsUSA
| | - Randall L. Woltjer
- National Institute on Aging, Layton Aging & Alzheimer's Disease Research CenterDepartment of NeurologyOregon Health & Science UniversityPortlandOregonUSA
- Department of PathologyOregon Health & Science UniversityPortlandOregonUSA
| |
Collapse
|
30
|
Patel V, Edison P. Cardiometabolic risk factors and neurodegeneration: a review of the mechanisms underlying diabetes, obesity and hypertension in Alzheimer's disease. J Neurol Neurosurg Psychiatry 2024; 95:581-589. [PMID: 38290839 PMCID: PMC11103343 DOI: 10.1136/jnnp-2023-332661] [Citation(s) in RCA: 22] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Accepted: 01/09/2024] [Indexed: 02/01/2024]
Abstract
A growing body of evidence suggests that cardiometabolic risk factors play a significant role in Alzheimer's disease (AD). Diabetes, obesity and hypertension are highly prevalent and can accelerate neurodegeneration and perpetuate the burden of AD. Insulin resistance and enzymes including insulin degrading enzymes are implicated in AD where breakdown of insulin is prioritised over amyloid-β. Leptin resistance and inflammation demonstrated by higher plasma and central nervous system levels of interleukin-6 (IL-6), IL-1β and tumour necrosis factor-α, are mechanisms connecting obesity and diabetes with AD. Leptin has been shown to ameliorate AD pathology and enhance long-term potentiation and hippocampal-dependent cognitive function. The renin-aldosterone angiotensin system, involved in hypertension, has been associated with AD pathology and neurotoxic reactive oxygen species, where angiotensin binds to specific angiotensin-1 receptors in the hippocampus and cerebral cortex. This review aims to consolidate the evidence behind putative processes stimulated by obesity, diabetes and hypertension, which leads to increased AD risk. We focus on how novel knowledge can be applied clinically to facilitate recognition of efficacious treatment strategies for AD.
Collapse
Affiliation(s)
- Vijay Patel
- Department of Brain Sciences, Imperial College London, London, UK
| | - Paul Edison
- Department of Brain Sciences, Imperial College London, London, UK
- Cardiff University, Cardiff, UK
| |
Collapse
|
31
|
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
|
32
|
Qiu T, Hong H, Zeng Q, Xu X, Wang Y, Zhu L, Zhang L, Li K, Dai S, Li X, Xie F, Zhang Y, Luo X. Effect of cerebral small vessel disease on the integrity of cholinergic system in mild cognitive impairment patients: a longitudinal study. J Neurol 2024; 271:2704-2715. [PMID: 38381177 PMCID: PMC11055699 DOI: 10.1007/s00415-024-12218-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2023] [Revised: 01/22/2024] [Accepted: 01/22/2024] [Indexed: 02/22/2024]
Abstract
We aimed to investigate the effect of cerebral small vessel disease (SVD) on cholinergic system integrity in mild cognitive impairment (MCI) patients. Nucleus basalis of Meynert (NBM) volume and cholinergic pathways integrity was evaluated at baseline, 1-, 2-, and 4-year follow-ups in 40 cognitively unimpaired (CU) participants, 29 MCI patients without SVD, and 23 MCI patients with SVD. We compared cholinergic markers among three groups and examined their associations with SVD burden in MCI patients. We used linear mixed models to assess longitudinal changes in cholinergic markers over time among groups. Mediation analysis was employed to investigate the mediating role of cholinergic system degeneration between SVD and cognitive impairment. Increased mean diffusivity (MD) in medial and lateral pathways was observed in MCI patients with SVD compared to those without SVD and CU participants. Both MCI groups showed decreased NBM volume compared to CU participants, while there was no significant difference between the two MCI groups. Longitudinally, compared to CU participants, MCI patients with SVD displayed a more rapid change in MD in both pathways, but not in NBM volume. Furthermore, SVD burden was associated with cholinergic pathway disruption and its faster rate of change in MCI patients. However, mediation analyses showed that cholinergic pathways did not mediate significant indirect effects of SVD burden on cognitive impairment. Our findings suggest that SVD could accelerate the degeneration of cholinergic pathways in MCI patients. However, they do not provide evidence to support that SVD could contribute to cognitive impairment through cholinergic system injury.
Collapse
Affiliation(s)
- Tiantian Qiu
- Department of Radiology, Linyi People's Hospital, Linyi, China
| | - Hui Hong
- Department of Radiology, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Qingze Zeng
- Department of Radiology, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Xiaopei Xu
- Department of Radiology, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Yanyan Wang
- Laboratory Medicine Center, Linyi People's Hospital, Linyi, China
| | - Lixin Zhu
- Department of Radiology, Linyi People's Hospital, Linyi, China
| | - Lige Zhang
- Department of Radiology, Linyi People's Hospital, Linyi, China
| | - Kaicheng Li
- Department of Radiology, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Shouping Dai
- Department of Radiology, Linyi People's Hospital, Linyi, China
| | - Xiaodong Li
- Department of Radiology, Linyi People's Hospital, Linyi, China
| | - Fei Xie
- Department of Equipment and Medical Engineering, Linyi People's Hospital, Linyi, China
| | - Yusong Zhang
- Department of Radiology, Linyi People's Hospital, Linyi, China
| | - Xiao Luo
- Department of Radiology, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China.
| |
Collapse
|
33
|
Xhima K, Ottoy J, Gibson E, Zukotynski K, Scott C, Feliciano GJ, Adamo S, Kuo PH, Borrie MJ, Chertkow H, Frayne R, Laforce R, Noseworthy MD, Prato FS, Sahlas DJ, Smith EE, Sossi V, Thiel A, Soucy J, Tardif J, Goubran M, Black SE, Ramirez J. Distinct spatial contributions of amyloid pathology and cerebral small vessel disease to hippocampal morphology. Alzheimers Dement 2024; 20:3687-3695. [PMID: 38574400 PMCID: PMC11095424 DOI: 10.1002/alz.13791] [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: 09/30/2023] [Revised: 01/22/2024] [Accepted: 02/09/2024] [Indexed: 04/06/2024]
Abstract
INTRODUCTION Cerebral small vessel disease (SVD) and amyloid beta (Aβ) pathology frequently co-exist. The impact of concurrent pathology on the pattern of hippocampal atrophy, a key substrate of memory impacted early and extensively in dementia, remains poorly understood. METHODS In a unique cohort of mixed Alzheimer's disease and moderate-severe SVD, we examined whether total and regional neuroimaging measures of SVD, white matter hyperintensities (WMH), and Aβ, as assessed by 18F-AV45 positron emission tomography, exert additive or synergistic effects on hippocampal volume and shape. RESULTS Frontal WMH, occipital WMH, and Aβ were independently associated with smaller hippocampal volume. Frontal WMH had a spatially distinct impact on hippocampal shape relative to Aβ. In contrast, hippocampal shape alterations associated with occipital WMH spatially overlapped with Aβ-vulnerable subregions. DISCUSSION Hippocampal degeneration is differentially sensitive to SVD and Aβ pathology. The pattern of hippocampal atrophy could serve as a disease-specific biomarker, and thus guide clinical diagnosis and individualized treatment strategies for mixed dementia.
Collapse
Affiliation(s)
- Kristiana Xhima
- Dr. Sandra E. Black Centre for Brain Resilience and RecoveryLC Campbell Cognitive Neurology, Hurvitz Brain Sciences Program, Sunnybrook Research Institute, University of TorontoTorontoOntarioCanada
| | - Julie Ottoy
- Dr. Sandra E. Black Centre for Brain Resilience and RecoveryLC Campbell Cognitive Neurology, Hurvitz Brain Sciences Program, Sunnybrook Research Institute, University of TorontoTorontoOntarioCanada
| | - Erin Gibson
- Dr. Sandra E. Black Centre for Brain Resilience and RecoveryLC Campbell Cognitive Neurology, Hurvitz Brain Sciences Program, Sunnybrook Research Institute, University of TorontoTorontoOntarioCanada
| | - Katherine Zukotynski
- Dr. Sandra E. Black Centre for Brain Resilience and RecoveryLC Campbell Cognitive Neurology, Hurvitz Brain Sciences Program, Sunnybrook Research Institute, University of TorontoTorontoOntarioCanada
- Departments of Medicine and RadiologyMcMaster UniversityHamiltonOntarioCanada
- Department of Medical ImagingSchulich School of Medicine and Dentistry, Western UniversityLondonOntarioCanada
| | - Christopher Scott
- Dr. Sandra E. Black Centre for Brain Resilience and RecoveryLC Campbell Cognitive Neurology, Hurvitz Brain Sciences Program, Sunnybrook Research Institute, University of TorontoTorontoOntarioCanada
| | - Ginelle J. Feliciano
- Dr. Sandra E. Black Centre for Brain Resilience and RecoveryLC Campbell Cognitive Neurology, Hurvitz Brain Sciences Program, Sunnybrook Research Institute, University of TorontoTorontoOntarioCanada
| | - Sabrina Adamo
- Dr. Sandra E. Black Centre for Brain Resilience and RecoveryLC Campbell Cognitive Neurology, Hurvitz Brain Sciences Program, Sunnybrook Research Institute, University of TorontoTorontoOntarioCanada
| | - Phillip H. Kuo
- Departments of Medical Imaging, Medicine, Biomedical EngineeringUniversity of ArizonaTucsonArizonaUSA
| | - Michael J. Borrie
- Schulich School of Medicine and DentistryWestern UniversityLondonOntarioCanada
| | - Howard Chertkow
- Rotman Research InstituteBaycrest Health SciencesTorontoOntarioCanada
| | - Richard Frayne
- Departments of Radiology and Clinical NeuroscienceHotchkiss Brain Institute, University of CalgaryCalgaryAlbertaCanada
| | - Robert Laforce
- Clinique Interdisciplinaire de Mémoire, Département des Sciences NeurologiquesUniversité Laval, Quebec CityQuebecCanada
| | - Michael D. Noseworthy
- Departments of Medicine and RadiologyMcMaster UniversityHamiltonOntarioCanada
- Department of Electrical and Computer EngineeringMcMaster UniversityHamiltonOntarioCanada
| | - Frank S. Prato
- Schulich School of Medicine and DentistryWestern UniversityLondonOntarioCanada
| | | | - Eric E. Smith
- Department of Clinical Neurosciences and Hotchkiss Brain InstituteUniversity of CalgaryCalgaryAlbertaCanada
| | - Vesna Sossi
- Physics and Astronomy Department and DM Center for Brain HealthUniversity of British ColumbiaVancouverBritish ColumbiaCanada
| | - Alexander Thiel
- Department of Neurology and NeurosurgeryMcGill UniversityMontrealQuebecCanada
| | - Jean‐Paul Soucy
- Montreal Neurological InstituteMcGill UniversityMontrealQuebecCanada
| | | | - Maged Goubran
- Dr. Sandra E. Black Centre for Brain Resilience and RecoveryLC Campbell Cognitive Neurology, Hurvitz Brain Sciences Program, Sunnybrook Research Institute, University of TorontoTorontoOntarioCanada
- Department of Medical BiophysicsUniversity of TorontoTorontoOntarioCanada
- Physical Sciences Platform, Sunnybrook Research InstituteUniversity of TorontoTorontoOntarioCanada
| | - Sandra E. Black
- Dr. Sandra E. Black Centre for Brain Resilience and RecoveryLC Campbell Cognitive Neurology, Hurvitz Brain Sciences Program, Sunnybrook Research Institute, University of TorontoTorontoOntarioCanada
- Division of NeurologyDepartment of MedicineUniversity of TorontoTorontoOntarioCanada
| | - Joel Ramirez
- Dr. Sandra E. Black Centre for Brain Resilience and RecoveryLC Campbell Cognitive Neurology, Hurvitz Brain Sciences Program, Sunnybrook Research Institute, University of TorontoTorontoOntarioCanada
| | | |
Collapse
|
34
|
Albar NY, Hassaballa H, Shikh H, Albar Y, Ibrahim AS, Mousa AH, Alshanberi AM, Elgebaly A, Bahbah EI. The interaction between insulin resistance and Alzheimer's disease: a review article. Postgrad Med 2024; 136:377-395. [PMID: 38804907 DOI: 10.1080/00325481.2024.2360887] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2024] [Accepted: 05/23/2024] [Indexed: 05/29/2024]
Abstract
Insulin serves multiple functions as a growth-promoting hormone in peripheral tissues. It manages glucose metabolism by promoting glucose uptake into cells and curbing the production of glucose in the liver. Beyond this, insulin fosters cell growth, drives differentiation, aids protein synthesis, and deters degradative processes like glycolysis, lipolysis, and proteolysis. Receptors for insulin and insulin-like growth factor-1 are widely expressed in the central nervous system. Their widespread presence in the brain underscores the varied and critical functions of insulin signaling there. Insulin aids in bolstering cognition, promoting neuron extension, adjusting the release and absorption of catecholamines, and controlling the expression and positioning of gamma-aminobutyric acid (GABA). Importantly, insulin can effortlessly traverse the blood-brain barrier. Furthermore, insulin resistance (IR)-induced alterations in insulin signaling might hasten brain aging, impacting its plasticity and potentially leading to neurodegeneration. Two primary pathways are responsible for insulin signal transmission: the phosphoinositide 3-kinase (PI3K)/protein kinase B (AKT) pathway, which oversees metabolic responses, and the mitogen-activated protein kinase (MAPK) pathway, which guides cell growth, survival, and gene transcription. This review aimed to explore the potential shared metabolic traits between Alzheimer's disease (AD) and IR disorders. It delves into the relationship between AD and IR disorders, their overlapping genetic markers, and shared metabolic indicators. Additionally, it addresses existing therapeutic interventions targeting these intersecting pathways.
Collapse
Affiliation(s)
- Nezar Y Albar
- Internal Medicine Department, Dr. Samir Abbas Hospital, Jeddah, Saudi Arabia
| | | | - Hamza Shikh
- Ibn Sina National College for Medical Studies, Jeddah, Saudi Arabia
| | - Yassin Albar
- Fakeeh College of Medical Sciences, Jeddah, Saudi Arabia
| | | | - Ahmed Hafez Mousa
- Department of Neurosurgery, Postgraduate Medical Education, Mohammed Bin Rashid University of Medicine and Health Sciences, Dubai, United Arab Emirates
- Department of Neurosurgery, Rashid Hospital, Dubai Academic Health Cooperation, Dubai, United Arab Emirates
| | - Asim Muhammed Alshanberi
- Department of Community Medicine and Pilgrims Health Care, Umm Alqura University, Makkah, Saudi Arabia
- Medicine Program, Batterjee Medical College, Jeddah, Saudi Arabia
| | - Ahmed Elgebaly
- Smart Health Academic Unit, University of East London, London, UK
| | - Eshak I Bahbah
- Faculty of Medicine, Al-Azhar University, Damietta, Egypt
| |
Collapse
|
35
|
Biesbroek JM, Coenen M, DeCarli C, Fletcher EM, Maillard PM, Barkhof F, Barnes J, Benke T, Chen CPLH, Dal‐Bianco P, Dewenter A, Duering M, Enzinger C, Ewers M, Exalto LG, Franzmeier N, Hilal S, Hofer E, Koek HL, Maier AB, McCreary CR, Papma JM, Paterson RW, Pijnenburg YAL, Rubinski A, Schmidt R, Schott JM, Slattery CF, Smith EE, Sudre CH, Steketee RME, Teunissen CE, van den Berg E, van der Flier WM, Venketasubramanian N, Venkatraghavan V, Vernooij MW, Wolters FJ, Xin X, Kuijf HJ, Biessels GJ. Amyloid pathology and vascular risk are associated with distinct patterns of cerebral white matter hyperintensities: A multicenter study in 3132 memory clinic patients. Alzheimers Dement 2024; 20:2980-2989. [PMID: 38477469 PMCID: PMC11032573 DOI: 10.1002/alz.13765] [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: 10/30/2023] [Revised: 01/30/2024] [Accepted: 01/31/2024] [Indexed: 03/14/2024]
Abstract
INTRODUCTION White matter hyperintensities (WMH) are associated with key dementia etiologies, in particular arteriolosclerosis and amyloid pathology. We aimed to identify WMH locations associated with vascular risk or cerebral amyloid-β1-42 (Aβ42)-positive status. METHODS Individual patient data (n = 3,132; mean age 71.5 ± 9 years; 49.3% female) from 11 memory clinic cohorts were harmonized. WMH volumes in 28 regions were related to a vascular risk compound score (VRCS) and Aß42 status (based on cerebrospinal fluid or amyloid positron emission tomography), correcting for age, sex, study site, and total WMH volume. RESULTS VRCS was associated with WMH in anterior/superior corona radiata (B = 0.034/0.038, p < 0.001), external capsule (B = 0.052, p < 0.001), and middle cerebellar peduncle (B = 0.067, p < 0.001), and Aß42-positive status with WMH in posterior thalamic radiation (B = 0.097, p < 0.001) and splenium (B = 0.103, p < 0.001). DISCUSSION Vascular risk factors and Aß42 pathology have distinct signature WMH patterns. This regional vulnerability may incite future studies into how arteriolosclerosis and Aß42 pathology affect the brain's white matter. HIGHLIGHTS Key dementia etiologies may be associated with specific patterns of white matter hyperintensities (WMH). We related WMH locations to vascular risk and cerebral Aβ42 status in 11 memory clinic cohorts. Aβ42 positive status was associated with posterior WMH in splenium and posterior thalamic radiation. Vascular risk was associated with anterior and infratentorial WMH. Amyloid pathology and vascular risk have distinct signature WMH patterns.
Collapse
|
36
|
Bachmann D, von Rickenbach B, Buchmann A, Hüllner M, Zuber I, Studer S, Saake A, Rauen K, Gruber E, Nitsch RM, Hock C, Treyer V, Gietl A. White matter hyperintensity patterns: associations with comorbidities, amyloid, and cognition. Alzheimers Res Ther 2024; 16:67. [PMID: 38561806 PMCID: PMC10983708 DOI: 10.1186/s13195-024-01435-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2023] [Accepted: 03/23/2024] [Indexed: 04/04/2024]
Abstract
BACKGROUND White matter hyperintensities (WMHs) are often measured globally, but spatial patterns of WMHs could underlie different risk factors and neuropathological and clinical correlates. We investigated the spatial heterogeneity of WMHs and their association with comorbidities, Alzheimer's disease (AD) risk factors, and cognition. METHODS In this cross-sectional study, we studied 171 cognitively unimpaired (CU; median age: 65 years, range: 50 to 89) and 51 mildly cognitively impaired (MCI; median age: 72, range: 53 to 89) individuals with available amyloid (18F-flutementamol) PET and FLAIR-weighted images. Comorbidities were assessed using the Cumulative Illness Rating Scale (CIRS). Each participant's white matter was segmented into 38 parcels, and WMH volume was calculated in each parcel. Correlated principal component analysis was applied to the parceled WMH data to determine patterns of WMH covariation. Adjusted and unadjusted linear regression models were used to investigate associations of component scores with comorbidities and AD-related factors. Using multiple linear regression, we tested whether WMH component scores predicted cognitive performance. RESULTS Principal component analysis identified four WMH components that broadly describe FLAIR signal hyperintensities in posterior, periventricular, and deep white matter regions, as well as basal ganglia and thalamic structures. In CU individuals, hypertension was associated with all patterns except the periventricular component. MCI individuals showed more diverse associations. The posterior and deep components were associated with renal disorders, the periventricular component was associated with increased amyloid, and the subcortical gray matter structures was associated with sleep disorders, endocrine/metabolic disorders, and increased amyloid. In the combined sample (CU + MCI), the main effects of WMH components were not associated with cognition but predicted poorer episodic memory performance in the presence of increased amyloid. No interaction between hypertension and the number of comorbidities on component scores was observed. CONCLUSION Our study underscores the significance of understanding the regional distribution patterns of WMHs and the valuable insights that risk factors can offer regarding their underlying causes. Moreover, patterns of hyperintensities in periventricular regions and deep gray matter structures may have more pronounced cognitive implications, especially when amyloid pathology is also present.
Collapse
Affiliation(s)
- Dario Bachmann
- Institute for Regenerative Medicine, University of Zurich, Campus Schlieren, Wagistrasse 12, 8952, Zurich, Schlieren, Switzerland.
- Department of Health Sciences and Technology, ETH Zürich, 8093, Zurich, Switzerland.
| | | | - Andreas Buchmann
- Institute for Regenerative Medicine, University of Zurich, Campus Schlieren, Wagistrasse 12, 8952, Zurich, Schlieren, Switzerland
| | - Martin Hüllner
- Department of Nuclear Medicine, University Hospital of Zurich, University of Zurich, 8091, Zurich, Switzerland
| | - Isabelle Zuber
- Institute for Regenerative Medicine, University of Zurich, Campus Schlieren, Wagistrasse 12, 8952, Zurich, Schlieren, Switzerland
| | - Sandro Studer
- Institute for Regenerative Medicine, University of Zurich, Campus Schlieren, Wagistrasse 12, 8952, Zurich, Schlieren, Switzerland
| | - Antje Saake
- Institute for Regenerative Medicine, University of Zurich, Campus Schlieren, Wagistrasse 12, 8952, Zurich, Schlieren, Switzerland
| | - Katrin Rauen
- Institute for Regenerative Medicine, University of Zurich, Campus Schlieren, Wagistrasse 12, 8952, Zurich, Schlieren, Switzerland
- Department of Geriatric Psychiatry, Psychiatric Hospital Zurich, 8032, Zurich, Switzerland
- Neuroscience Center Zurich, University of Zurich, 8057, Zurich, Switzerland
| | - Esmeralda Gruber
- Institute for Regenerative Medicine, University of Zurich, Campus Schlieren, Wagistrasse 12, 8952, Zurich, Schlieren, Switzerland
| | - Roger M Nitsch
- Institute for Regenerative Medicine, University of Zurich, Campus Schlieren, Wagistrasse 12, 8952, Zurich, Schlieren, Switzerland
- Neurimmune AG, 8952, Zurich, Schlieren, Switzerland
| | - Christoph Hock
- Institute for Regenerative Medicine, University of Zurich, Campus Schlieren, Wagistrasse 12, 8952, Zurich, Schlieren, Switzerland
- Neurimmune AG, 8952, Zurich, Schlieren, Switzerland
| | - Valerie Treyer
- Institute for Regenerative Medicine, University of Zurich, Campus Schlieren, Wagistrasse 12, 8952, Zurich, Schlieren, Switzerland
- Department of Nuclear Medicine, University Hospital of Zurich, University of Zurich, 8091, Zurich, Switzerland
| | - Anton Gietl
- Institute for Regenerative Medicine, University of Zurich, Campus Schlieren, Wagistrasse 12, 8952, Zurich, Schlieren, Switzerland
- Department of Geriatric Psychiatry, Psychiatric Hospital Zurich, 8032, Zurich, Switzerland
| |
Collapse
|
37
|
Zhou R, Cai Q, Liu C, Hui J, Kang M, Gou Y, Liu Y, Shi P, Wang B, Zhang F. Association between white matter hyperintensity and anxiety/depression. Cereb Cortex 2024; 34:bhae149. [PMID: 38602744 DOI: 10.1093/cercor/bhae149] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2023] [Revised: 03/12/2024] [Accepted: 03/13/2024] [Indexed: 04/12/2024] Open
Abstract
Although previous studies have explored the associations of white matter hyperintensity with psychiatric disorders, the sample size is small and the conclusions are inconsistent. The present study aimed to further systematically explore the association in a larger sample. All data were extracted from the UK Biobank. First, general linear regression models and logistic regression models were used to assess the association between white matter hyperintensity volume and anxiety/depression. White matter hyperintensity has been classified into periventricular white matter hyperintensity and deep white matter hyperintensity. Anxiety was determined by General Anxiety Disorder-7 score (n = 17,221) and self-reported anxiety (n = 15,333), depression was determined by Patient Health Questionnaire-9 score (n = 17,175), and self-reported depression (n = 14,519). Moreover, we employed Cox proportional hazard models to explore the association between white matter hyperintensity volume and anxiety/depression. The covariates included in fully adjusted model are age, gender, body mass index, Townsend deprivation index, healthy physical activity, cigarette consumption, alcohol consumption, educational attainment, diabetes, hypertension, and coronary heart disease. The results of the fully adjusted model showed that white matter hyperintensity volume was significantly associated with General Anxiety Disorder-7 score (periventricular white matter hyperintensity: β = 0.152, deep white matter hyperintensity: β = 0.094) and Patient Health Questionnaire-9 score (periventricular white matter hyperintensity: β = 0.168). Logistic regression analysis results indicated that periventricular white matter hyperintensity volume (odds ratio = 1.153) was significantly associated with self-reported anxiety. After applying the Cox proportional hazard models, we found that larger white matter hyperintensity volume was associated with increased risk of depression (periventricular white matter hyperintensity: hazard ratio = 1.589, deep white matter hyperintensity: hazard ratio = 1.200), but not anxiety. In summary, our findings support a positive association between white matter hyperintensity volume and depression.
Collapse
Affiliation(s)
- Ruixue Zhou
- Key Laboratory of Trace Elements and Endemic Diseases, Collaborative Innovation Center of Endemic Disease and Health Promotion for Silk Road Region, School of Public Health, Health Science Center, Xi'an Jiaotong University, Xi'an, 710061, P. R. China
| | - Qingqing Cai
- Key Laboratory of Trace Elements and Endemic Diseases, Collaborative Innovation Center of Endemic Disease and Health Promotion for Silk Road Region, School of Public Health, Health Science Center, Xi'an Jiaotong University, Xi'an, 710061, P. R. China
| | - Chen Liu
- Key Laboratory of Trace Elements and Endemic Diseases, Collaborative Innovation Center of Endemic Disease and Health Promotion for Silk Road Region, School of Public Health, Health Science Center, Xi'an Jiaotong University, Xi'an, 710061, P. R. China
| | - Jingni Hui
- Key Laboratory of Trace Elements and Endemic Diseases, Collaborative Innovation Center of Endemic Disease and Health Promotion for Silk Road Region, School of Public Health, Health Science Center, Xi'an Jiaotong University, Xi'an, 710061, P. R. China
| | - Meijuan Kang
- Key Laboratory of Trace Elements and Endemic Diseases, Collaborative Innovation Center of Endemic Disease and Health Promotion for Silk Road Region, School of Public Health, Health Science Center, Xi'an Jiaotong University, Xi'an, 710061, P. R. China
| | - Yifan Gou
- Key Laboratory of Trace Elements and Endemic Diseases, Collaborative Innovation Center of Endemic Disease and Health Promotion for Silk Road Region, School of Public Health, Health Science Center, Xi'an Jiaotong University, Xi'an, 710061, P. R. China
| | - Ye Liu
- Key Laboratory of Trace Elements and Endemic Diseases, Collaborative Innovation Center of Endemic Disease and Health Promotion for Silk Road Region, School of Public Health, Health Science Center, Xi'an Jiaotong University, Xi'an, 710061, P. R. China
| | - Panxing Shi
- Key Laboratory of Trace Elements and Endemic Diseases, Collaborative Innovation Center of Endemic Disease and Health Promotion for Silk Road Region, School of Public Health, Health Science Center, Xi'an Jiaotong University, Xi'an, 710061, P. R. China
| | - Bingyi Wang
- Key Laboratory of Trace Elements and Endemic Diseases, Collaborative Innovation Center of Endemic Disease and Health Promotion for Silk Road Region, School of Public Health, Health Science Center, Xi'an Jiaotong University, Xi'an, 710061, P. R. China
| | - Feng Zhang
- Key Laboratory of Trace Elements and Endemic Diseases, Collaborative Innovation Center of Endemic Disease and Health Promotion for Silk Road Region, School of Public Health, Health Science Center, Xi'an Jiaotong University, Xi'an, 710061, P. R. China
- Department of Psychiatry, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| |
Collapse
|
38
|
Pradeep A, Raghavan S, Przybelski SA, Preboske G, Schwarz CG, Lowe VJ, Knopman DS, Petersen RC, Jack CR, Graff-Radford J, Cogswell PM, Vemuri P. Can white matter hyperintensities based Fazekas visual assessment scales inform about Alzheimer's disease pathology in the population? RESEARCH SQUARE 2024:rs.3.rs-4017874. [PMID: 38558965 PMCID: PMC10980106 DOI: 10.21203/rs.3.rs-4017874/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/04/2024]
Abstract
Background White matter hyperintensities (WMH) are considered hallmark features of cerebral small vessel disease and have recently been linked to Alzheimer's disease pathology. Their distinct spatial distributions, namely periventricular versus deep WMH, may differ by underlying age-related and pathobiological processes contributing to cognitive decline. We aimed to identify the spatial patterns of WMH using the 4-scale Fazekas visual assessment and explore their differential association with age, vascular health, Alzheimer's imaging markers, namely amyloid and tau burden, and cognition. Because our study consisted of scans from GE and Siemens scanners with different resolutions, we also investigated inter-scanner reproducibility and combinability of WMH measurements on imaging. Methods We identified 1144 participants from the Mayo Clinic Study of Aging consisting of older adults from Olmsted County, Minnesota with available structural magnetic resonance imaging (MRI), amyloid, and tau positron emission tomography (PET). WMH distribution patterns were assessed on FLAIR-MRI, both 2D axial and 3D, using Fazekas ratings of periventricular and deep WMH severity. We compared the association of periventricular and deep WMH scales with vascular risk factors, amyloid-PET and tau-PET standardized uptake value ratio, WMH volume, and cognition using Pearson partial correlation after adjusting for age. We also evaluated vendor compatibility and reproducibility of the Fazekas scales using intraclass correlations (ICC). Results Periventricular and deep WMH measurements showed similar correlations with age, cardiometabolic conditions score (vascular risk), and cognition, (p < 0.001). Both periventricular WMH and deep WMH showed weak associations with amyloidosis (R = 0.07, p = < 0.001), and none with tau burden. We found substantial agreement between data from the two scanners for Fazekas measurements (ICC = 0.78). The automated WMH volume had high discriminating power for identifying participants with Fazekas ≥ 2 (area under curve = 0.97). Conclusion Our study investigates risk factors underlying WMH spatial patterns and their impact on global cognition, with no discernible differences between periventricular and deep WMH. We observed minimal impact of amyloidosis on WMH severity. These findings, coupled with enhanced inter-scanner reproducibility of WMH data, suggest the combinability of inter-scanner data assessed by harmonized protocols in the context of vascular contributions to cognitive impairment and dementia biomarker research.
Collapse
|
39
|
Cha WJ, Yi D, Ahn H, Byun MS, Chang YY, Choi JM, Kim K, Choi H, Jung G, Kang KM, Sohn CH, Lee YS, Kim YK, Lee DY. Association between brain amyloid deposition and longitudinal changes of white matter hyperintensities. Alzheimers Res Ther 2024; 16:50. [PMID: 38454444 PMCID: PMC10918927 DOI: 10.1186/s13195-024-01417-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2023] [Accepted: 02/20/2024] [Indexed: 03/09/2024]
Abstract
BACKGROUND Growing evidence suggests that not only cerebrovascular disease but also Alzheimer's disease (AD) pathological process itself cause cerebral white matter degeneration, resulting in white matter hyperintensities (WMHs). Some preclinical evidence also indicates that white matter degeneration may precede or affect the development of AD pathology. This study aimed to clarify the direction of influence between in vivo AD pathologies, particularly beta-amyloid (Aβ) and tau deposition, and WMHs through longitudinal approach. METHODS Total 282 older adults including cognitively normal and cognitively impaired individuals were recruited from the Korean Brain Aging Study for the Early Diagnosis and Prediction of Alzheimer's Disease (KBASE) cohort. The participants underwent comprehensive clinical and neuropsychological assessment, [11C] Pittsburgh Compound B PET for measuring Aβ deposition, [18F] AV-1451 PET for measuring tau deposition, and MRI scans with fluid-attenuated inversion recovery image for measuring WMH volume. The relationships between Aβ or tau deposition and WMH volume were examined using multiple linear regression analysis. In this analysis, baseline Aβ or tau were used as independent variables, and change of WMH volume over 2 years was used as dependent variable to examine the effect of AD pathology on increase of WMH volume. Additionally, we set baseline WMH volume as independent variable and longitudinal change of Aβ or tau deposition for 2 years as dependent variables to investigate whether WMH volume could precede AD pathologies. RESULTS Baseline Aβ deposition, but not tau deposition, had significant positive association with longitudinal change of WMH volume over 2 years. Baseline WMH volume was not related with any of longitudinal change of Aβ or tau deposition for 2 years. We also found a significant interaction effect between baseline Aβ deposition and sex on longitudinal change of WMH volume. Subsequent subgroup analyses showed that high baseline Aβ deposition was associated with increase of WMH volume over 2 years in female, but not in male. CONCLUSIONS Our findings suggest that Aβ deposition accelerates cerebral WMHs, particularly in female, whereas white matter degeneration appears not influence on longitudinal Aβ increase. The results also did not support any direction of influence between tau deposition and WMHs.
Collapse
Affiliation(s)
- Woo-Jin Cha
- Department of Neuropsychiatry, Seoul National University Hospital, Seoul, Republic of Korea
| | - Dahyun Yi
- Institute of Human Behavioral Medicine, Medical Research Center, Seoul National University, Seoul, Republic of Korea
| | - Hyejin Ahn
- Interdisciplinary program of cognitive science, Seoul National University College of Humanities, Seoul, Republic of Korea
| | - Min Soo Byun
- Department of Neuropsychiatry, Seoul National University Hospital, Seoul, Republic of Korea
- Department of Psychiatry, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Yoon Young Chang
- Department of Psychiatry, Inje University Sanggye Paik Hospital, Seoul, Republic of Korea
| | - Jung-Min Choi
- Department of Neuropsychiatry, Seoul National University Hospital, Seoul, Republic of Korea
| | - Kyungtae Kim
- Department of Neuropsychiatry, Seoul National University Hospital, Seoul, Republic of Korea
| | - Hyeji Choi
- Department of Neuropsychiatry, Seoul National University Hospital, Seoul, Republic of Korea
| | - Gijung Jung
- Institute of Human Behavioral Medicine, Medical Research Center, Seoul National University, Seoul, Republic of Korea
| | - Koung Mi Kang
- Department of Radiology, Seoul National University Hospital, Seoul, Republic of Korea
| | - Chul-Ho Sohn
- Department of Radiology, Seoul National University Hospital, Seoul, Republic of Korea
| | - Yun-Sang Lee
- Department of Nuclear Medicine, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Yu Kyeong Kim
- Department of Nuclear Medicine, Seoul Metropolitan Government-Seoul National University Boramae Medical Center, Seoul, Republic of Korea
| | - Dong Young Lee
- Department of Neuropsychiatry, Seoul National University Hospital, Seoul, Republic of Korea.
- Institute of Human Behavioral Medicine, Medical Research Center, Seoul National University, Seoul, Republic of Korea.
- Interdisciplinary program of cognitive science, Seoul National University College of Humanities, Seoul, Republic of Korea.
- Department of Psychiatry, Seoul National University College of Medicine, Seoul, Republic of Korea.
| |
Collapse
|
40
|
Royse SK, Snitz BE, Hengenius JB, Huppert TJ, Roush RE, Ehrenkranz RE, Wilson JD, Bertolet M, Reese AC, Cisneros G, Potopenko K, Becker JT, Cohen AD, Shaaban CE. Unhealthy white matter connectivity, cognition, and racialization in older adults. Alzheimers Dement 2024; 20:1483-1496. [PMID: 37828730 PMCID: PMC10947965 DOI: 10.1002/alz.13494] [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: 04/16/2023] [Revised: 09/06/2023] [Accepted: 09/10/2023] [Indexed: 10/14/2023]
Abstract
INTRODUCTION White matter hyperintensities (WMH) may promote clinical Alzheimer's disease (AD) disparities between Black American (BA) and non-Hispanic White (nHW) populations. Using a novel measurement, unhealthy white matter connectivity (UWMC), we interrogated racialized group differences in associations between WMH in AD pathology-affected regions and cognition. METHODS UWMC is the proportion of white matter fibers that pass through WMH for every pair of brain regions. Individual regression models tested associations of UWMC in beta-amyloid (Aβ) or tau pathology-affected regions with cognition overall, stratified by racialized group, and with a racialized group interaction. RESULTS In 201 older adults ranging from cognitively unimpaired to AD, BA participants exhibited greater UWMC and worse cognition than nHW participants. UWMC was negatively associated with cognition in 17 and 5 Aβ- and tau-affected regions, respectively. Racialization did not modify these relationships. DISCUSSION Differential UWMC burden, not differential UWMC-and-cognition associations, may drive clinical AD disparities between racialized groups. HIGHLIGHTS Unhealthy white matter connectivity (UWMC) in Alzheimer's disease (AD) pathology-affected brain regions is associated with cognition. Relationships between UWMC and cognition are similar between Black American (BA) and non-Hispanic White (nHW) individuals. More UWMC may partially drive higher clinical AD burden in BA versus nHW populations. UWMC risk factors, particularly social and environmental, should be identified.
Collapse
Affiliation(s)
- Sarah K. Royse
- Department of EpidemiologyUniversity of PittsburghPittsburghPennsylvaniaUSA
- Department of RadiologyUniversity of PittsburghPittsburghPennsylvaniaUSA
| | - Beth E. Snitz
- Department of NeurologyUniversity of PittsburghPittsburghPennsylvaniaUSA
| | - James B. Hengenius
- Department of EpidemiologyUniversity of PittsburghPittsburghPennsylvaniaUSA
| | - Theodore J. Huppert
- Department of Electrical EngineeringUniversity of PittsburghPittsburghPennsylvaniaUSA
| | - Rebecca E. Roush
- Department of NeurologyUniversity of PittsburghPittsburghPennsylvaniaUSA
| | | | - James D. Wilson
- Department of Mathematics and StatisticsUniversity of San FranciscoSan FranciscoCaliforniaUSA
| | - Marnie Bertolet
- Department of EpidemiologyUniversity of PittsburghPittsburghPennsylvaniaUSA
- Department of BiostatisticsUniversity of PittsburghPittsburghPennsylvaniaUSA
| | | | - Geraldine Cisneros
- Department of PsychologyUniversity of PittsburghPittsburghPennsylvaniaUSA
| | - Katey Potopenko
- Department of PsychologyUniversity of PittsburghPittsburghPennsylvaniaUSA
| | - James T. Becker
- Department of NeurologyUniversity of PittsburghPittsburghPennsylvaniaUSA
- Department of BiostatisticsUniversity of PittsburghPittsburghPennsylvaniaUSA
- Department of PsychiatryUniversity of PittsburghPittsburghPennsylvaniaUSA
| | - Ann D. Cohen
- Department of PsychiatryUniversity of PittsburghPittsburghPennsylvaniaUSA
| | | |
Collapse
|
41
|
Rubinski A, Dewenter A, Zheng L, Franzmeier N, Stephenson H, Deming Y, Duering M, Gesierich B, Denecke J, Pham AV, Bendlin B, Ewers M. Florbetapir PET-assessed demyelination is associated with faster tau accumulation in an APOE ε4-dependent manner. Eur J Nucl Med Mol Imaging 2024; 51:1035-1049. [PMID: 38049659 PMCID: PMC10881623 DOI: 10.1007/s00259-023-06530-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2023] [Accepted: 11/14/2023] [Indexed: 12/06/2023]
Abstract
PURPOSE The main objectives were to test whether (1) a decrease in myelin is associated with enhanced rate of fibrillar tau accumulation and cognitive decline in Alzheimer's disease, and (2) whether apolipoprotein E (APOE) ε4 genotype is associated with worse myelin decrease and thus tau accumulation. METHODS To address our objectives, we repurposed florbetapir-PET as a marker of myelin in the white matter (WM) based on previous validation studies showing that beta-amyloid (Aβ) PET tracers bind to WM myelin. We assessed 43 Aβ-biomarker negative (Aβ-) cognitively normal participants and 108 Aβ+ participants within the AD spectrum with florbetapir-PET at baseline and longitudinal flortaucipir-PET as a measure of fibrillar tau (tau-PET) over ~ 2 years. In linear regression analyses, we tested florbetapir-PET in the whole WM and major fiber tracts as predictors of tau-PET accumulation in a priori defined regions of interest (ROIs) and fiber-tract projection areas. In mediation analyses we tested whether tau-PET accumulation mediates the effect of florbetapir-PET in the whole WM on cognition. Finally, we assessed the role of myelin alteration on the association between APOE and tau-PET accumulation. RESULTS Lower florbetapir-PET in the whole WM or at a given fiber tract was predictive of faster tau-PET accumulation in Braak stages or the connected grey matter areas in Aβ+ participants. Faster tau-PET accumulation in higher cortical brain areas mediated the association between a decrease in florbetapir-PET in the WM and a faster rate of decline in global cognition and episodic memory. APOE ε4 genotype was associated with a worse decrease in the whole WM florbetapir-PET and thus enhanced tau-PET accumulation. CONCLUSION Myelin alterations are associated in an APOE ε4 dependent manner with faster tau progression and cognitive decline, and may therefore play a role in the etiology of AD.
Collapse
Affiliation(s)
- Anna Rubinski
- Institute for Stroke and Dementia Research, University Hospital, Ludwig-Maximilian-University Munich, Munich, Germany
| | - Anna Dewenter
- Institute for Stroke and Dementia Research, University Hospital, Ludwig-Maximilian-University Munich, Munich, Germany
| | - Lukai Zheng
- Institute for Stroke and Dementia Research, University Hospital, Ludwig-Maximilian-University Munich, Munich, Germany
| | - Nicolai Franzmeier
- Institute for Stroke and Dementia Research, University Hospital, Ludwig-Maximilian-University Munich, Munich, Germany
| | - Henry Stephenson
- Department of Medicine, School of Medicine and Public Health, University of Wisconsin- Madison, Madison, WI, USA
| | - Yuetiva Deming
- Department of Medicine, School of Medicine and Public Health, University of Wisconsin- Madison, Madison, WI, USA
| | - Marco Duering
- Institute for Stroke and Dementia Research, University Hospital, Ludwig-Maximilian-University Munich, Munich, Germany
- Medical Image Analysis Center (MIAC) and Department of Biomedical Engineering, University of Basel, Basel, Switzerland
| | - Benno Gesierich
- Medical Image Analysis Center (MIAC) and Department of Biomedical Engineering, University of Basel, Basel, Switzerland
| | - Jannis Denecke
- Institute for Stroke and Dementia Research, University Hospital, Ludwig-Maximilian-University Munich, Munich, Germany
| | - An-Vi Pham
- Institute for Stroke and Dementia Research, University Hospital, Ludwig-Maximilian-University Munich, Munich, Germany
- Department of Neuroradiology, School of Medicine, Technical University of Munich, Munich, Germany
| | - Barbara Bendlin
- Department of Medicine, School of Medicine and Public Health, University of Wisconsin- Madison, Madison, WI, USA
| | - Michael Ewers
- Institute for Stroke and Dementia Research, University Hospital, Ludwig-Maximilian-University Munich, Munich, Germany.
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany.
| |
Collapse
|
42
|
Morales CD, Cotton-Samuel D, Lao PJ, Chang JF, Pyne JD, Alshikho MJ, Lippert RV, Bista K, Hale C, Edwards NC, Igwe KC, Deters K, Zimmerman ME, Brickman AM. Small vessel cerebrovascular disease is associated with cognition in prospective Alzheimer's clinical trial participants. Alzheimers Res Ther 2024; 16:25. [PMID: 38308344 PMCID: PMC10836014 DOI: 10.1186/s13195-024-01395-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Accepted: 01/21/2024] [Indexed: 02/04/2024]
Abstract
BACKGROUND Secondary prevention clinical trials for Alzheimer's disease (AD) target amyloid accumulation in asymptomatic, amyloid-positive individuals, but it is unclear to what extent other pathophysiological processes, such as small vessel cerebrovascular disease, account for participant performance on the primary cognitive outcomes in those trials. White matter hyperintensities are areas of increased signal on T2-weighted magnetic resonance imaging (MRI) that reflect small vessel cerebrovascular disease. They are associated with cognitive functioning in older adults and with clinical presentation and course of AD, particularly when distributed in posterior brain regions. The purpose of this study was to examine to what degree regional WMH volume is associated with performance on the primary cognitive outcome measure in the Anti-Amyloid Treatment in Asymptomatic Alzheimer's Disease (A4) study, a secondary prevention trial. METHODS Data from 1791 participants (59.5% women, mean age (SD) 71.6 (4.74)) in the A4 study and the Longitudinal Evaluation of Amyloid Risk and Neurodegeneration (LEARN) companion study at the screening visit were used to quantify WMH volumes on T2-weighted fluid-attenuated inversion recovery (FLAIR) MR images. Cognition was assessed with the preclinical Alzheimer cognitive composite (PACC). We tested the association of total and regional WMH volumes with PACC performance, adjusting for age, education, and amyloid positivity status, with general linear models. We also considered interactions between WMH and amyloid positivity status. RESULTS Increased frontal and parietal lobe WMH volume was associated with poorer performance on the PACC. While amyloid positivity was also associated with lower cognitive test scores, WMH volumes did not interact with amyloid positivity status. CONCLUSION These results highlight the potential of small vessel cerebrovascular disease to drive AD-related cognitive profiles. Measures of small vessel cerebrovascular disease should be considered when evaluating outcome in trials, both as potential effect modifiers and as a possible target for intervention or prevention.
Collapse
Affiliation(s)
- Clarissa D Morales
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Gertrude H. Sergievsky Center, Department of Neurology, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY, 10032, USA
| | - Dejania Cotton-Samuel
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Gertrude H. Sergievsky Center, Department of Neurology, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY, 10032, USA
| | - Patrick J Lao
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Gertrude H. Sergievsky Center, Department of Neurology, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY, 10032, USA
| | - Julia F Chang
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Gertrude H. Sergievsky Center, Department of Neurology, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY, 10032, USA
| | - Jeffrey D Pyne
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Gertrude H. Sergievsky Center, Department of Neurology, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY, 10032, USA
| | - Mohamad J Alshikho
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Gertrude H. Sergievsky Center, Department of Neurology, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY, 10032, USA
| | - Rafael V Lippert
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Gertrude H. Sergievsky Center, Department of Neurology, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY, 10032, USA
| | - Kelsang Bista
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Gertrude H. Sergievsky Center, Department of Neurology, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY, 10032, USA
| | - Christiane Hale
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Gertrude H. Sergievsky Center, Department of Neurology, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY, 10032, USA
| | - Natalie C Edwards
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Gertrude H. Sergievsky Center, Department of Neurology, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY, 10032, USA
| | - Kay C Igwe
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Gertrude H. Sergievsky Center, Department of Neurology, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY, 10032, USA
| | - Kacie Deters
- Department of Integrative Biology & Physiology, University of California, Los Angeles, CA, USA
| | | | - Adam M Brickman
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Gertrude H. Sergievsky Center, Department of Neurology, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY, 10032, USA.
| |
Collapse
|
43
|
Sampatakakis SN, Mourtzi N, Charisis S, Mamalaki E, Ntanasi E, Hatzimanolis A, Ramirez A, Lambert JC, Yannakoulia M, Kosmidis MH, Dardiotis E, Hadjigeorgiou G, Sakka P, Scarmeas N. Genetic Predisposition for White Matter Hyperintensities and Risk of Mild Cognitive Impairment and Alzheimer's Disease: Results from the HELIAD Study. Curr Issues Mol Biol 2024; 46:934-947. [PMID: 38275674 PMCID: PMC10814944 DOI: 10.3390/cimb46010060] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2023] [Revised: 01/13/2024] [Accepted: 01/19/2024] [Indexed: 01/27/2024] Open
Abstract
The present study investigated the association of genetic predisposition for white matter hyperintensities (WMHs) with incident amnestic mild cognitive impairment (aMCI) or Alzheimer's disease (AD), as well as whether such an association was influenced by age, sex, and cognitive reserve. Overall, 537 individuals without aMCI or dementia at baseline were included. Among them, 62 individuals developed aMCI/AD at follow up. Genetic propensity to WMH was estimated using a polygenic risk score for WMHs (PRS WMH). The association of PRS WMH with aMCI/AD incidence was examined using COX models. A higher PRS WMH was associated with a 47.2% higher aMCI/AD incidence (p = 0.015) in the fully adjusted model. Subgroup analyses showed significant results in the older age group, in which individuals with a higher genetic predisposition for WMHs had a 3.4-fold higher risk for developing aMCI/AD at follow up (p < 0.001), as well as in the lower cognitive reserve (CR, proxied by education years) group, in which individuals with a higher genetic predisposition for WMHs had an over 2-fold higher risk (p = 0.013). Genetic predisposition for WMHs was associated with aMCI/AD incidence, particularly in the group of participants with a low CR. Thus, CR might be a modifier in the relationship between genetic predisposition for WMHs and incident aMCI/AD.
Collapse
Affiliation(s)
- Stefanos N. Sampatakakis
- 1st Department of Neurology, Aiginition Hospital, Athens Medical School, National and Kapodistrian University, 11528 Athens, Greece; (S.N.S.); (N.M.); (E.M.); (E.N.)
| | - Niki Mourtzi
- 1st Department of Neurology, Aiginition Hospital, Athens Medical School, National and Kapodistrian University, 11528 Athens, Greece; (S.N.S.); (N.M.); (E.M.); (E.N.)
| | - Sokratis Charisis
- Department of Neurology, UT Health San Antonio, San Antonio, TX 78229, USA;
| | - Eirini Mamalaki
- 1st Department of Neurology, Aiginition Hospital, Athens Medical School, National and Kapodistrian University, 11528 Athens, Greece; (S.N.S.); (N.M.); (E.M.); (E.N.)
| | - Eva Ntanasi
- 1st Department of Neurology, Aiginition Hospital, Athens Medical School, National and Kapodistrian University, 11528 Athens, Greece; (S.N.S.); (N.M.); (E.M.); (E.N.)
| | - Alexandros Hatzimanolis
- Department of Psychiatry, Aiginition Hospital, Athens Medical School, National and Kapodistrian University, 11528 Athens, Greece;
| | - Alfredo Ramirez
- Division of Neurogenetics and Molecular Psychiatry, Department of Psychiatry and Psychotherapy, Medical Faculty, University of Cologne, 50923 Cologne, Germany
- Department of Neurodegenerative Diseases and Geriatric Psychiatry, University Hospital Bonn, 53127 Bonn, Germany
- German Center for Neurodegenerative Diseases (DZNE Bonn), 53127 Bonn, Germany
- Department of Psychiatry, Glenn Biggs Institute for Alzheimer’s and Neurodegenerative Diseases, San Antonio, TX 78229, USA
- Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, 50923 Cologne, Germany
| | - Jean-Charles Lambert
- Inserm, CHU Lille, Institut Pasteur de Lille, U1167-RID-AGE Facteurs de Risque et Déterminants Moléculaires des Maladies Liés au Vieillissement, University of Lille, 59000 Lille, France;
| | - Mary Yannakoulia
- Department of Nutrition and Dietetics, Harokopio University, 17676 Athens, Greece;
| | - Mary H. Kosmidis
- Lab of Neuropsychology and Behavioral Neuroscience, School of Psychology, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece;
| | - Efthimios Dardiotis
- Department of Neurology, University Hospital of Larissa, Faculty of Medicine, School of Health Sciences, University of Thessaly, 41334 Larissa, Greece;
| | | | - Paraskevi Sakka
- Athens Association of Alzheimer’s Disease and Related Disorders, 11636 Marousi, Greece;
| | - Nikolaos Scarmeas
- 1st Department of Neurology, Aiginition Hospital, Athens Medical School, National and Kapodistrian University, 11528 Athens, Greece; (S.N.S.); (N.M.); (E.M.); (E.N.)
- Department of Neurology, The Gertrude H. Sergievsky Center, Taub Institute for Research in Alzheimer’s Disease and the Aging Brain, Columbia University, New York, NY 10027, USA
| |
Collapse
|
44
|
Liu SW, Ma XT, Yu S, Weng XF, Li M, Zhu J, Liu CF, Hu H. Bridging Reduced Grip Strength and Altered Executive Function: Specific Brain White Matter Structural Changes in Patients with Alzheimer's Disease. Clin Interv Aging 2024; 19:93-107. [PMID: 38250174 PMCID: PMC10799618 DOI: 10.2147/cia.s438782] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2023] [Accepted: 01/09/2024] [Indexed: 01/23/2024] Open
Abstract
Objective To investigate the correlation between specific fiber tracts and grip strength and cognitive function in patients with Alzheimer's disease (AD) by fixel-based analysis (FBA). Methods AD patients were divided into AD with low grip strength (AD-LGS, n=29) and AD without low grip strength (AD-nLGS, n=25), along with 31 normal controls (NC). General data, neuropsychological tests, grip strength and cranial magnetic resonance imaging (MRI) scans were collected. FBA evaluated white matter (WM) fiber metrics, including fiber density (FD), fiber cross-sectional (FC), and fiber density and cross-sectional area (FDC). The mean fiber indicators of the fiber tracts of interest (TOI) were extracted in cerebral region of significant statistical differences in FBA to further compare the differences between groups and analyze the correlation between fiber properties and neuropsychological test scores. Results Compared to AD-nLGS group, AD-LGS group showed significant reductions in FDC in several cerebral regions. In AD patients, FDC values of bilateral uncinate fasciculus and left superior longitudinal fasciculus were positively correlated with Clock Drawing Test scores, while FDC of splenium of corpus callosum, bilateral anterior cingulate tracts, forceps major, and bilateral inferior longitudinal fasciculus were positively correlated with the Executive Factor Score of Memory and Executive Screening scale scores. Conclusion Reduced grip strength in AD patients is associated with extensive impairment of WM structural integrity. Changes in FDC of specific WM fiber tracts related to executive function play a significant mediating role in the reduction of grip strength in AD patients.
Collapse
Affiliation(s)
- Shan-Wen Liu
- Department of Neurology, the Second Affiliated Hospital of Soochow University, Suzhou, 215004, People’s Republic of China
| | - Xiao-Ting Ma
- Department of Neurology, the Second Affiliated Hospital of Soochow University, Suzhou, 215004, People’s Republic of China
| | - Shuai Yu
- Department of Neurology, the Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Suzhou, 215000, People’s Republic of China
| | - Xiao-Fen Weng
- Department of Geriatric Medicine, the Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Suzhou, 215000, People’s Republic of China
| | - Meng Li
- Department of Imaging, the Second Affiliated Hospital of Soochow University, Suzhou, 215004, People’s Republic of China
| | - Jiangtao Zhu
- Department of Imaging, the Second Affiliated Hospital of Soochow University, Suzhou, 215004, People’s Republic of China
| | - Chun-Feng Liu
- Department of Neurology, the Second Affiliated Hospital of Soochow University, Suzhou, 215004, People’s Republic of China
| | - Hua Hu
- Department of Neurology, the Second Affiliated Hospital of Soochow University, Suzhou, 215004, People’s Republic of China
| |
Collapse
|
45
|
Holtman IR, Glass CK, Nott A. Interpretation of Neurodegenerative GWAS Risk Alleles in Microglia and their Interplay with Other Cell Types. ADVANCES IN NEUROBIOLOGY 2024; 37:531-544. [PMID: 39207711 DOI: 10.1007/978-3-031-55529-9_29] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/04/2024]
Abstract
Microglia have been implicated in numerous neurodegenerative and neuroinflammatory disorders; however, the causal contribution of this immune cell type is frequently debated. Genetic studies offer a unique vantage point in that they infer causality over a secondary consequence. Genome-wide association studies (GWASs) have identified hundreds of loci in the genome that are associated with susceptibility to neurodegenerative disorders. GWAS studies implicate microglia in the pathogenesis of Alzheimer's disease (AD), Parkinson's disease (PD), multiple sclerosis (MS), and to a lesser degree suggest a role for microglia in vascular dementia (VaD), frontotemporal dementia (FTD), and amyotrophic lateral sclerosis (ALS), and other neurodegenerative and neuropsychiatric disorders. The contribution and function of GWAS risk loci on disease progression is an ongoing field of study, in which large genomic datasets, and an extensive framework of computational tools, have proven to be crucial. Several GWAS risk loci are shared between disorders, pointing towards common pleiotropic mechanisms. In this chapter, we introduce key concepts in GWAS and post-GWAS interpretation of neurodegenerative disorders, with a focus on GWAS risk genes implicated in microglia, their interplay with other cell types and shared convergence of GWAS risk loci on microglia.
Collapse
Affiliation(s)
- Inge R Holtman
- Department of Biomedical Sciences, Section Molecular Neurobiology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Christopher K Glass
- Department of Cellular and Molecular Medicine, School of Medicine, UC San Diego, La Jolla, CA, USA.
- Department of Medicine, School of Medicine, UC San Diego, La Jolla, CA, USA.
| | - Alexi Nott
- Department of Brain Sciences, Imperial College London, London, UK
- UK Dementia Research Institute, Imperial College London, London, UK
| |
Collapse
|
46
|
Ye YC, Chai SF, Li XR, Wu MN, Cai HY, Wang ZJ. Intermittent fasting and Alzheimer's disease-Targeting ketone bodies as a potential strategy for brain energy rescue. Metab Brain Dis 2024; 39:129-146. [PMID: 37823968 DOI: 10.1007/s11011-023-01288-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/15/2023] [Accepted: 09/01/2023] [Indexed: 10/13/2023]
Abstract
Alzheimer's disease (AD) lacks effective clinical treatments. As the disease progresses, the cerebral glucose hypometabolism that appears in the preclinical phase of AD gradually worsens, leading to increasingly severe brain energy disorders. This review analyzes the brain energy deficit in AD and its etiology, brain energy rescue strategies based on ketone intervention, the effects and mechanisms of IF, the differences in efficacy between IF and ketogenic diet and the duality of IF. The evidence suggests that brain energy deficits lead to the development and progression of AD pathology. IF, which improves brain energy impairments by promoting ketone metabolism, thus has good therapeutic potential for AD.
Collapse
Affiliation(s)
- Yu- Cai Ye
- Department of Physiology, Key Laboratory of Cellular Physiology, Ministry of Education, Shanxi Medical University, Taiyuan, People's Republic of China
| | - Shi-Fan Chai
- Department of Physiology, Key Laboratory of Cellular Physiology, Ministry of Education, Shanxi Medical University, Taiyuan, People's Republic of China
| | - Xin-Ru Li
- Department of Physiology, Key Laboratory of Cellular Physiology, Ministry of Education, Shanxi Medical University, Taiyuan, People's Republic of China
| | - Mei-Na Wu
- Department of Physiology, Key Laboratory of Cellular Physiology, Ministry of Education, Shanxi Medical University, Taiyuan, People's Republic of China
| | - Hong-Yan Cai
- Department of Microbiology and Immunology, Shanxi Medical University, Taiyuan, People's Republic of China
| | - Zhao-Jun Wang
- Department of Physiology, Key Laboratory of Cellular Physiology, Ministry of Education, Shanxi Medical University, Taiyuan, People's Republic of China.
| |
Collapse
|
47
|
Zhang Z, Lim MJR. Incident Dementia After Spontaneous Intracerebral Hemorrhage. J Alzheimers Dis 2024; 99:41-51. [PMID: 38640161 DOI: 10.3233/jad-240111] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/21/2024]
Abstract
Post-stroke cognitive impairment and dementia (PSCID) is a complication that affects long-term functional outcomes after stroke. Studies on dementia after long-term follow-up in stroke have focused predominantly on ischemic stroke, which may be different from the development of dementia after spontaneous intracerebral hemorrhage (ICH). In this review, we summarize the existing data and hypotheses on the development of dementia after spontaneous ICH, review the management of post-ICH dementia, and suggest areas for future research. Dementia after spontaneous ICH has a cumulative incidence of up to 32.0-37.4% at 5 years post-ICH. Although the pathophysiology of post-ICH dementia has not been fully understood, two main theoretical frameworks can be considered: 1) the triggering role of ICH (both primary and secondary brain injury) in precipitating cognitive decline and dementia; and 2) the contributory role of pre-existing brain pathology (including small vessel disease and neurodegenerative pathology), reduced cognitive reserve, and genetic factors predisposing to cognitive dysfunction. These pathophysiological pathways may have synergistic effects that converge on dysfunction of the neurovascular unit and disruptions in functional connectivity leading to dementia post-ICH. Management of post-ICH dementia may include screening and monitoring, cognitive therapy, and pharmacotherapy. Non-invasive brain stimulation is an emerging therapeutic modality under investigation for safety and efficacy. Our review highlights that there remains a paucity of data and standardized reporting on incident dementia after spontaneous ICH. Further research is imperative for determining the incidence, risk factors, and pathophysiology of post-ICH dementia, in order to identify new therapies for the treatment of this debilitating condition.
Collapse
Affiliation(s)
- Zheting Zhang
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore
| | | |
Collapse
|
48
|
Hotz I, Deschwanden PF, Mérillat S, Jäncke L. Associations between white matter hyperintensities, lacunes, entorhinal cortex thickness, declarative memory and leisure activity in cognitively healthy older adults: A 7-year study. Neuroimage 2023; 284:120461. [PMID: 37981203 DOI: 10.1016/j.neuroimage.2023.120461] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2023] [Revised: 11/02/2023] [Accepted: 11/16/2023] [Indexed: 11/21/2023] Open
Abstract
INTRODUCTION Cerebral small vessel disease (cSVD) is a growing epidemic that affects brain health and cognition. Therefore, a more profound understanding of the interplay between cSVD, brain atrophy, and cognition in healthy aging is of great importance. In this study, we examined the association between white matter hyperintensities (WMH) volume, number of lacunes, entorhinal cortex (EC) thickness, and declarative memory in cognitively healthy older adults over a seven-year period, controlling for possible confounding factors. Because there is no cure for cSVD to date, the neuroprotective potential of an active lifestyle has been suggested. Supporting evidence, however, is scarce. Therefore, a second objective of this study is to examine the relationship between leisure activities, cSVD, EC thickness, and declarative memory. METHODS We used a longitudinal dataset, which consisted of five measurement time points of structural MRI and psychometric cognitive ability and survey data, collected from a sample of healthy older adults (baseline N = 231, age range: 64-87 years, age M = 70.8 years), to investigate associations between cSVD MRI markers, EC thickness and verbal and figural memory performance. Further, we computed physical, social, and cognitive leisure activity scores from survey-based assessments and examined their associations with brain structure and declarative memory. To provide more accurate estimates of the trajectories and cross-domain correlations, we applied latent growth curve models controlling for potential confounders. RESULTS Less age-related thinning of the right (β = 0.92, p<.05) and left EC (β = 0.82, p<.05) was related to less declarative memory decline; and a thicker EC at baseline predicted less declarative memory loss (β = 0.54, p<.05). Higher baseline levels of physical (β = 0.24, p<.05), and social leisure activity (β = 0.27, p<.01) predicted less thinning of right EC. No relation was found between WMH or lacunes and declarative memory or between leisure activity and declarative memory. Higher education was initially related to more physical activity (β = 0.16, p<.05) and better declarative memory (β = 0.23, p<.001), which, however, declined steeper in participants with higher education (β = -.35, p<.05). Obese participants were less physically (β = -.18, p<.01) and socially active (β = -.13, p<.05) and had thinner left EC (β = -.14, p<.05) at baseline. Antihypertensive medication use (β = -.26, p<.05), and light-to-moderate alcohol consumption (β = -.40, p<.001) were associated with a smaller increase in the number of lacunes whereas a larger increase in the number of lacunes was observed in current smokers (β = 0.30, p<.05). CONCLUSIONS Our results suggest complex relationships between cSVD MRI markers (total WMH, number of lacunes, right and left EC thickness), declarative memory, and confounding factors such as antihypertensive medication, obesity, and leisure activitiy. Thus, leisure activities and having good cognitive reserve counteracting this neurodegeneration. Several confounding factors seem to contribute to the extent or progression/decline of cSVD, which needs further investigation in the future. Since there is still no cure for cSVD, modifiable confounding factors should be studied more intensively in the future to maintain or promote brain health and thus cognitive abilities in older adults.
Collapse
Affiliation(s)
- Isabel Hotz
- Dynamics of Healthy Aging, University Research Priority Program (URPP), University of Zurich, Stampfenbachstrasse 73, Zurich CH-8006, Switzerland.
| | - Pascal Frédéric Deschwanden
- Dynamics of Healthy Aging, University Research Priority Program (URPP), University of Zurich, Stampfenbachstrasse 73, Zurich CH-8006, Switzerland
| | - Susan Mérillat
- Dynamics of Healthy Aging, University Research Priority Program (URPP), University of Zurich, Stampfenbachstrasse 73, Zurich CH-8006, Switzerland
| | - Lutz Jäncke
- Dynamics of Healthy Aging, University Research Priority Program (URPP), University of Zurich, Stampfenbachstrasse 73, Zurich CH-8006, Switzerland
| |
Collapse
|
49
|
Zhong S, Lou J, Ma K, Shu Z, Chen L, Li C, Ye Q, Zhou L, Shen Y, Ye X, Zhang J. Disentangling in-vivo microstructural changes of white and gray matter in mild cognitive impairment and Alzheimer's disease: a systematic review and meta-analysis. Brain Imaging Behav 2023; 17:764-777. [PMID: 37752311 DOI: 10.1007/s11682-023-00805-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/14/2023] [Indexed: 09/28/2023]
Abstract
The microstructural characteristics of white and gray matter in mild cognitive impairment (MCI) and the early-stage of Alzheimer's disease (AD) remain unclear. This study aimed to systematically identify the microstructural damages of MCI/AD in studies using neurite orientation dispersion and density imaging (NODDI), and explore their correlations with cognitive performance. Multiple databases were searched for eligible studies. The 10 eligible NODDI studies were finally included. Patients with MCI/AD showed overall significant reductions in neurite density index (NDI) of specific white matter structures in bilateral hemispheres (left hemisphere: -0.40 [-0.53, -0.27], P < 0.001; right: -0.33 [-0.47, -0.19], P < 0.001), involving the bilateral superior longitudinal fasciculus (SLF), uncinate fasciculus (UF), the left posterior thalamic radiation (PTR), and the left cingulum. White matter regions exhibited significant increased orientation dispersion index (ODI) (left: 0.25 [0.02, 0.48], P < 0.05; right: 0.27 [0.07, 0.46], P < 0.05), including the left cingulum, the right UF, and the bilateral parahippocampal cingulum (PHC), and PTR. Additionally, the ODI of gray matter showed significant reduction in bilateral hippocampi (left: -0.97 [-1.42, -0.51], P < 0.001; right: -0.90 [-1.35, -0.45], P < 0.001). The cognitive performance in MCI/AD was significantly associated with NDI (r = 0.50, P < 0.001). Our findings highlight the microstructural changes in MCI/AD were characterized by decreased fiber orientation dispersion in the hippocampus, and decreased neurite density and increased fiber orientation dispersion in specific white matter tracts, including the cingulum, UF, and PTR. Moreover, the decreased NDI may indicate the declined cognitive level of MCI/AD patients.
Collapse
Affiliation(s)
- Shuchang Zhong
- Center for Rehabilitation Medicine, Rehabilitation & Sports Medicine Research Institute of Zhejiang Province, Department of Rehabilitation Medicine, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Jingjing Lou
- Center for Rehabilitation Medicine, Rehabilitation & Sports Medicine Research Institute of Zhejiang Province, Department of Rehabilitation Medicine, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Ke Ma
- Center for Rehabilitation Medicine, Rehabilitation & Sports Medicine Research Institute of Zhejiang Province, Department of Rehabilitation Medicine, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Zhenyu Shu
- Department of Radiology, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Lin Chen
- Center for Rehabilitation Medicine, Rehabilitation & Sports Medicine Research Institute of Zhejiang Province, Department of Rehabilitation Medicine, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Chao Li
- Center for Rehabilitation Medicine, Rehabilitation & Sports Medicine Research Institute of Zhejiang Province, Department of Rehabilitation Medicine, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Qing Ye
- Center for Rehabilitation Medicine, Rehabilitation & Sports Medicine Research Institute of Zhejiang Province, Department of Rehabilitation Medicine, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Liang Zhou
- Center for Rehabilitation Medicine, Rehabilitation & Sports Medicine Research Institute of Zhejiang Province, Department of Rehabilitation Medicine, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Ye Shen
- Center for Rehabilitation Medicine, Rehabilitation & Sports Medicine Research Institute of Zhejiang Province, Department of Rehabilitation Medicine, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Xiangming Ye
- Center for Rehabilitation Medicine, Rehabilitation & Sports Medicine Research Institute of Zhejiang Province, Department of Rehabilitation Medicine, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Jie Zhang
- Center for Rehabilitation Medicine, Rehabilitation & Sports Medicine Research Institute of Zhejiang Province, Department of Rehabilitation Medicine, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, China.
| |
Collapse
|
50
|
Wang R, Wu X, Zhang Z, Cao L, Kwapong WR, Wang H, Tao W, Ye C, Liu J, Wu B. Retinal ganglion cell-inner plexiform layer, white matter hyperintensities, and their interaction with cognition in older adults. Front Aging Neurosci 2023; 15:1240815. [PMID: 38035269 PMCID: PMC10685347 DOI: 10.3389/fnagi.2023.1240815] [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: 06/15/2023] [Accepted: 10/11/2023] [Indexed: 12/02/2023] Open
Abstract
Purpose We explored the interaction of optical coherence tomography (OCT) parameters and white matter hyperintensities with cognitive measures in our older adult cohort. Methods This observational study enrolled participants who underwent a comprehensive neuropsychological battery, structural 3-T brain magnetic resonance imaging (MRI), visual acuity examination, and OCT imaging. Cerebral small vessel disease (CSVD) markers were read on MR images; lacune, cerebral microbleeds (CMB), white matter hyperintensities (WMH), and enlarged perivascular spaces (EPVS), were defined according to the STRIVE standards. Retinal nerve fiber layer (RNFL) and ganglion cell-inner plexiform layer (GCIPL) thicknesses (μm) were measured on the OCT tool. Results Older adults with cognitive impairment (CI) showed lower RNFL (p = 0.001), GCIPL (p = 0.009) thicknesses, and lower hippocampal volume (p = 0.004) when compared to non-cognitively impaired (NCI). RNFL (p = 0.006) and GCIPL thicknesses (p = 0.032) correlated with MoCA scores. GCIPL thickness (p = 0.037), total WMH (p = 0.003), PWMH (p = 0.041), and DWMH (p = 0.001) correlated with hippocampal volume in our older adults after adjusting for covariates. With hippocampal volume as the outcome, a significant interaction (p < 0.05) between GCIPL and PWMH and total WMH was observed in our older adults. Conclusion Both GCIPL thinning and higher WMH burden (especially PWMH) are associated with hippocampal volume and older adults with both pathologies are more susceptible to subclinical cognitive decline.
Collapse
Affiliation(s)
- Ruilin Wang
- Ophthalmology Department, West China Hospital, Sichuan University, Chengdu, China
| | - Xinmao Wu
- Neurology Department, West China Hospital, Sichuan University, Chengdu, China
| | - Zengyi Zhang
- Neurology Department, West China Hospital, Sichuan University, Chengdu, China
| | - Le Cao
- Ophthalmology Department, West China Hospital, Sichuan University, Chengdu, China
| | | | - Hang Wang
- Neurology Department, West China Hospital, Sichuan University, Chengdu, China
| | - Wendan Tao
- Neurology Department, West China Hospital, Sichuan University, Chengdu, China
| | - Chen Ye
- Neurology Department, West China Hospital, Sichuan University, Chengdu, China
| | - Junfeng Liu
- Neurology Department, West China Hospital, Sichuan University, Chengdu, China
| | - Bo Wu
- Neurology Department, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|